Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 73
1.
Nervenarzt ; 92(5): 433-440, 2021 May.
Article De | MEDLINE | ID: mdl-33502576

Besides cognitive behavioral therapy (CBT), psychopharmacotherapy belongs to the first-line treatment approaches for anxiety disorders according to all national and international guidelines. According to studies and meta-analyses, modern antidepressants in particular have been proven to be effective. Depending on the substance, there are approvals for panic disorder, generalized anxiety disorder and social phobia. There are also approvals for other substance groups, e.g. anticonvulsants for generalized anxiety disorder. Benzodiazepines should be used with caution in view of the risk of dependency. Although effective and well-tolerated medications are available, up to 30% of patients still do not respond or do not respond adequately to treatment. Consequently, research efforts to develop new substances are important. Based on a better understanding of the complex neurobiological mechanisms underlying anxiety disorders, a large number of substances are currently undergoing clinical trials. Modulators of current and new transmitter systems, in particular the glutamatergic and the endocannabinoid systems as well as neuropeptides, are being discussed as innovative substances. Strategies are also being investigated which, in combination with psychotherapy, aim at optimizing fear extinction memory. First studies are also underway on the use of psychedelic agents in combination with psychotherapy for anxiety.


Cognitive Behavioral Therapy , Panic Disorder , Anxiety Disorders/diagnosis , Anxiety Disorders/drug therapy , Extinction, Psychological , Fear , Humans
2.
Psychoneuroendocrinology ; 110: 104430, 2019 12.
Article En | MEDLINE | ID: mdl-31542636

BACKGROUND: Obesity is a risk factor for stress-related mental disorders such as post-traumatic stress disorder. The underlying mechanism through which obesity affects mental health remains poorly understood but dysregulation of the ghrelin system may be involved. Stress increases plasma ghrelin levels, which stimulates food intake as a potential stress-coping mechanism. However, diet-induced obesity induces ghrelin resistance which in turn may have deleterious effects on stress-coping. In our study, we explored whether disruption of ghrelin receptor function though high-fat diet or genetic ablation affects fear processing, anxiety-like behavior and saccharin preference in mice. METHODS: Adult male C57BL6/J mice were placed on a standard diet or high-fat diet for a total period of 8 weeks. We first established that high-fat diet exposure for 4 weeks elicits ghrelin resistance, evidenced by a blunted hyperphagic response following administration of a ghrelin receptor agonist. We then carried out an experiment in which we subjected mice to auditory fear conditioning after 4 weeks of diet exposure and evaluated effects on fear extinction, anxiety-like behavior and saccharin preference. To explore whether fear conditioning as such may influence the effect of diet exposure, we also subjected mice to auditory fear conditioning prior to diet onset and 4 weeks later we investigated auditory fear extinction, anxiety-like behavior and saccharin preference. In a final experiment, we further assessed lack of ghrelin receptor function by investigating auditory fear processing, anxiety-like behavior and saccharin preference in ghrelin receptor knockout mice and their wild-type littermates. RESULTS: High-fat diet exposure had no significant effect on auditory fear conditioning and its subsequent extinction or on anxiety-like behavior but significantly lowered saccharin preference. Similarly, ghrelin receptor knockout mice did not differ significantly from their wild-type littermates for auditory fear processing or anxiety-like behavior but showed significantly lower saccharin preference compared to wild-type littermates. CONCLUSION: Taken together, our data suggest that disruption of ghrelin receptor function per se does not affect fear or anxiety-like behavior but may decrease saccharin preference in mice.


Anxiety/genetics , Fear , Food Preferences , Receptors, Ghrelin/genetics , Saccharin/administration & dosage , Animals , Anxiety/metabolism , Anxiety/physiopathology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Fear/psychology , Food Preferences/psychology , Gene Deletion , Ghrelin/physiology , Hyperphagia/genetics , Hyperphagia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Ghrelin/physiology
3.
Genes Brain Behav ; 17(3): e12427, 2018 03.
Article En | MEDLINE | ID: mdl-29028150

The current treatment of post-traumatic stress disorder (PTSD), phobias and other anxiety disorders, remains insufficient particularly in producing long-lasting full symptom control. Dysfunctional fear processing is common in these disorders, including a deficiency in fear-inhibitory mechanisms and impairment in the ability to discriminate between safety and danger cues. Research has aimed to elucidate brain circuitries, neurotransmitters and downstream signaling pathways important in the alleviation of aberrant fear, with a specific focus on mechanisms modulating fear memory and its behavioral expression. MicroRNAs (miRNA) as "fine tuners" of gene expression at the post-transcriptional level have emerged as critical regulators of such mechanisms important in both, the generation and the inhibition of fear memories. Along these lines, abnormal expression of miRNAs has been associated with different fear-related disorders. After providing an updated overview on the involvement of miRNAs in fear learning mechanisms, we summarize and discuss in particular those studies in which the implication of miRNAs in successful inhibition of fear has been explored. For a better overview, we dissociate the different modes of fear alleviation investigated in this regard and present studies in rodents demonstrating that specific miRNAs are involved in the destabilization of fear by interfering with consolidation/reconsolidation mechanisms or that they are associated with the generation of fear extinction or safety learning. Finally, we discuss the potential of miRNAs as biomarkers and novel therapeutic targets, as well as the challenges involved in applying the discovered mechanisms in the development of improved treatments of fear- and trauma-related disorders.


Fear/physiology , Fear/psychology , MicroRNAs/genetics , Animals , Anxiety Disorders/physiopathology , Brain/physiopathology , Conditioning, Classical/physiology , Humans , Learning/physiology , Memory/physiology , MicroRNAs/therapeutic use , Neuronal Plasticity/genetics , Signal Transduction/physiology , Stress Disorders, Post-Traumatic/physiopathology
4.
Transl Psychiatry ; 6(12): e974, 2016 12 06.
Article En | MEDLINE | ID: mdl-27922638

Extinction-based exposure therapy is used to treat anxiety- and trauma-related disorders; however, there is the need to improve its limited efficacy in individuals with impaired fear extinction learning and to promote greater protection against return-of-fear phenomena. Here, using 129S1/SvImJ mice, which display impaired fear extinction acquisition and extinction consolidation, we revealed that persistent and context-independent rescue of deficient fear extinction in these mice was associated with enhanced expression of dopamine-related genes, such as dopamine D1 (Drd1a) and -D2 (Drd2) receptor genes in the medial prefrontal cortex (mPFC) and amygdala, but not hippocampus. Moreover, enhanced histone acetylation was observed in the promoter of the extinction-regulated Drd2 gene in the mPFC, revealing a potential gene-regulatory mechanism. Although enhancing histone acetylation, via administering the histone deacetylase (HDAC) inhibitor MS-275, does not induce fear reduction during extinction training, it promoted enduring and context-independent rescue of deficient fear extinction consolidation/retrieval once extinction learning was initiated as shown following a mild conditioning protocol. This was associated with enhanced histone acetylation in neurons of the mPFC and amygdala. Finally, as a proof-of-principle, mimicking enhanced dopaminergic signaling by L-dopa treatment rescued deficient fear extinction and co-administration of MS-275 rendered this effect enduring and context-independent. In summary, current data reveal that combining dopaminergic and epigenetic mechanisms is a promising strategy to improve exposure-based behavior therapy in extinction-impaired individuals by initiating the formation of an enduring and context-independent fear-inhibitory memory.


Dopamine/physiology , Extinction, Psychological/physiology , Fear/physiology , Histone Acetyltransferases/physiology , Signal Transduction/physiology , Amygdala/physiology , Animals , Benzamides/pharmacology , Combined Modality Therapy , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Extinction, Psychological/drug effects , Fear/drug effects , Implosive Therapy , Levodopa/pharmacology , Male , Mice , Prefrontal Cortex/physiology , Pyridines/pharmacology , Signal Transduction/drug effects
5.
J Neurophysiol ; 114(4): 2500-8, 2015 Oct.
Article En | MEDLINE | ID: mdl-26334021

Substance P (SP) is implicated in stress regulation and affective and anxiety-related behavior. Particularly high expression has been found in the main output region of the amygdala complex, the central amygdala (CE). Here we investigated the cellular mechanisms of SP in CE in vitro, taking advantage of glutamic acid decarboxylase-green fluorescent protein (GAD67-GFP) knockin mice that yield a reliable labeling of GABAergic neurons, which comprise 95% of the neuronal population in the lateral section of CE (CEl). In GFP-positive neurons within CEl, SP caused a membrane depolarization and increase in input resistance, associated with an increase in action potential firing frequency. Under voltage-clamp conditions, the SP-specific membrane current reversed at -101.5 ± 2.8 mV and displayed inwardly rectifying properties indicative of a membrane K(+) conductance. Moreover, SP responses were blocked by the neurokinin type 1 receptor (NK1R) antagonist L-822429 and mimicked by the NK1R agonist [Sar(9),Met(O2)(11)]-SP. Immunofluorescence staining confirmed localization of NK1R in GFP-positive neurons in CEl, predominantly in PKCδ-negative neurons (80%) and in few PKCδ-positive neurons (17%). Differences in SP responses were not observed between the major types of CEl neurons (late firing, regular spiking, low-threshold bursting). In addition, SP increased the frequency and amplitude of GABAergic synaptic events in CEl neurons depending on upstream spike activity. These data indicate a NK1R-mediated increase in excitability and GABAergic activity in CEl neurons, which seems to mostly involve the PKCδ-negative subpopulation. This influence can be assumed to increase reciprocal interactions between CElon and CEloff pathways, thereby boosting the medial CE (CEm) output pathway and contributing to the anxiogenic-like action of SP in the amygdala.


Central Amygdaloid Nucleus/physiology , GABAergic Neurons/physiology , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Animals , Central Amygdaloid Nucleus/drug effects , Fluorescent Antibody Technique , GABAergic Neurons/drug effects , Gene Knock-In Techniques , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Mice, Transgenic , Neurokinin-1 Receptor Antagonists/pharmacology , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium/metabolism , Protein Kinase C-delta/metabolism , Tissue Culture Techniques
7.
Pharmacol Ther ; 149: 150-90, 2015 May.
Article En | MEDLINE | ID: mdl-25550231

Pathological fear and anxiety are highly debilitating and, despite considerable advances in psychotherapy and pharmacotherapy they remain insufficiently treated in many patients with PTSD, phobias, panic and other anxiety disorders. Increasing preclinical and clinical evidence indicates that pharmacological treatments including cognitive enhancers, when given as adjuncts to psychotherapeutic approaches [cognitive behavioral therapy including extinction-based exposure therapy] enhance treatment efficacy, while using anxiolytics such as benzodiazepines as adjuncts can undermine long-term treatment success. The purpose of this review is to outline the literature showing how pharmacological interventions targeting neurotransmitter systems including serotonin, dopamine, noradrenaline, histamine, glutamate, GABA, cannabinoids, neuropeptides (oxytocin, neuropeptides Y and S, opioids) and other targets (neurotrophins BDNF and FGF2, glucocorticoids, L-type-calcium channels, epigenetic modifications) as well as their downstream signaling pathways, can augment fear extinction and strengthen extinction memory persistently in preclinical models. Particularly promising approaches are discussed in regard to their effects on specific aspects of fear extinction namely, acquisition, consolidation and retrieval, including long-term protection from return of fear (relapse) phenomena like spontaneous recovery, reinstatement and renewal of fear. We also highlight the promising translational value of the preclinial research and the clinical potential of targeting certain neurochemical systems with, for example d-cycloserine, yohimbine, cortisol, and L-DOPA. The current body of research reveals important new insights into the neurobiology and neurochemistry of fear extinction and holds significant promise for pharmacologically-augmented psychotherapy as an improved approach to treat trauma and anxiety-related disorders in a more efficient and persistent way promoting enhanced symptom remission and recovery.


Anxiety/drug therapy , Extinction, Psychological/drug effects , Implosive Therapy , Nootropic Agents/pharmacology , Phobic Disorders/drug therapy , Stress Disorders, Post-Traumatic/drug therapy , Synaptic Transmission/drug effects , Animals , Anxiety/therapy , Combined Modality Therapy , Humans , Models, Neurological , Nootropic Agents/therapeutic use , Signal Transduction/drug effects
8.
Mol Psychiatry ; 20(7): 901-12, 2015 Jul.
Article En | MEDLINE | ID: mdl-25510511

Traumatic fear memories are highly durable but also dynamic, undergoing repeated reactivation and rehearsal over time. Although overly persistent fear memories underlie anxiety disorders, such as posttraumatic stress disorder, the key neural and molecular mechanisms underlying fear memory durability remain unclear. Postsynaptic density 95 (PSD-95) is a synaptic protein regulating glutamate receptor anchoring, synaptic stability and certain types of memory. Using a loss-of-function mutant mouse lacking the guanylate kinase domain of PSD-95 (PSD-95(GK)), we analyzed the contribution of PSD-95 to fear memory formation and retrieval, and sought to identify the neural basis of PSD-95-mediated memory maintenance using ex vivo immediate-early gene mapping, in vivo neuronal recordings and viral-mediated knockdown (KD) approaches. We show that PSD-95 is dispensable for the formation and expression of recent fear memories, but essential for the formation of precise and flexible fear memories and for the maintenance of memories at remote time points. The failure of PSD-95(GK) mice to retrieve remote cued fear memory was associated with hypoactivation of the infralimbic (IL) cortex (but not the anterior cingulate cortex (ACC) or prelimbic cortex), reduced IL single-unit firing and bursting, and attenuated IL gamma and theta oscillations. Adeno-associated virus-mediated PSD-95 KD in the IL, but not the ACC, was sufficient to impair recent fear extinction and remote fear memory, and remodel IL dendritic spines. Collectively, these data identify PSD-95 in the IL as a critical mechanism supporting the durability of fear memories over time. These preclinical findings have implications for developing novel approaches to treating trauma-based anxiety disorders that target the weakening of overly persistent fear memories.


Cerebral Cortex/physiology , Fear/physiology , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Memory/physiology , Action Potentials/physiology , Animals , Cerebral Cortex/cytology , Conditioning, Classical/physiology , Cues , Dendritic Spines/metabolism , Disks Large Homolog 4 Protein , Electrodes, Implanted , Electroshock , Extinction, Psychological/physiology , Female , Freezing Reaction, Cataleptic/physiology , Gamma Rhythm/physiology , Gene Knockdown Techniques , Guanylate Kinases/genetics , Male , Membrane Proteins/genetics , Mice, Mutant Strains , Olfactory Perception/physiology , Pyramidal Cells/cytology , Pyramidal Cells/physiology , Theta Rhythm/physiology
9.
Transl Psychiatry ; 4: e359, 2014 Feb 11.
Article En | MEDLINE | ID: mdl-24518397

The continuum of physiological anxiety up to psychopathology is not merely dependent on genes, but is orchestrated by the interplay of genetic predisposition, gene x environment and epigenetic interactions. Accordingly, inborn anxiety is considered a polygenic, multifactorial trait, likely to be shaped by environmentally driven plasticity at the genomic level. We here took advantage of the extreme genetic predisposition of the selectively bred high (HAB) and low anxiety (LAB) mouse model exhibiting high vs low anxiety-related behavior and tested whether and how beneficial (enriched environment) vs detrimental (chronic mild stress) environmental manipulations are capable of rescuing phenotypes from both ends of the anxiety continuum. We provide evidence that (i) even inborn and seemingly rigid behavioral and neuroendocrine phenotypes can bidirectionally be rescued by appropriate environmental stimuli, (ii) corticotropin-releasing hormone receptor 1 (Crhr1), critically involved in trait anxiety, shows bidirectional alterations in its expression in the basolateral amygdala (BLA) upon environmental stimulation, (iii) these alterations are linked to an increased methylation status of its promoter and, finally, (iv) binding of the transcription factor Yin Yang 1 (YY1) to the Crhr1 promoter contributes to its gene expression in a methylation-sensitive manner. Thus, Crhr1 in the BLA is critically involved as plasticity gene in the bidirectional epigenetic rescue of extremes in trait anxiety.


Anxiety/genetics , Basolateral Nuclear Complex/metabolism , Gene Expression/genetics , Gene-Environment Interaction , Genetic Predisposition to Disease , Receptors, Corticotropin-Releasing Hormone/genetics , Animals , Environment , Epigenesis, Genetic , Mice , Mice, Inbred Strains , Receptors, Corticotropin-Releasing Hormone/metabolism , YY1 Transcription Factor/metabolism
10.
J Neuroendocrinol ; 25(1): 48-55, 2013 Jan.
Article En | MEDLINE | ID: mdl-22861486

Accumulating evidence implicates the dorsomedial hypothalamic nucleus (DMH) in the regulation of autonomic and neuroendocrine stress responses. However, although projections from the DMH to the paraventricular hypothalamic nucleus (PVN), which is the critical site of the neuroendocrine stress axis, have been described, the impact of DMH neurones in the modulation of hypothalamic-pituitary-adrenal (HPA) axis activation during stress is not fully understood. The present study aimed to investigate the role of the DMH in HPA axis responses to different types of stimuli. Male Sprague-Dawley rats fitted with a chronic jugular venous catheter were exposed to either an emotional stressor (elevated platform-exposure) or immune challenge (systemic interleukin-1ß administration). Bilateral electrolytic lesions of the DMH disinhibited HPA axis responses to the emotional stressor, as indicated by higher plasma adrenocorticotrophic hormone levels during and after elevated platform exposure in lesioned animals compared to sham-lesioned controls. Moreover, DMH-lesioned animals showed increased neuronal activation in the PVN, as indicated by a higher c-Fos expression after elevated-platform exposure compared to controls. By contrast, DMH-lesions had no effects on HPA axis responses to immune challenge. Taken together, our data suggest an inhibitory role of DMH neurones on stress-induced HPA axis activation that is dependent upon the nature of the stimulus being important in response to an emotional stressor but not to immune challenge.


Dorsomedial Hypothalamic Nucleus/physiopathology , Emotions/physiology , Hypothalamo-Hypophyseal System/physiopathology , Neural Inhibition/physiology , Pituitary-Adrenal System/physiopathology , Stress, Psychological/physiopathology , Adrenocorticotropic Hormone/blood , Animals , Dorsomedial Hypothalamic Nucleus/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Interleukin-1beta/pharmacology , Male , Neural Inhibition/drug effects , Neural Pathways/drug effects , Neural Pathways/physiopathology , Neurons/drug effects , Neurons/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
11.
Transl Psychiatry ; 2: e171, 2012 Oct 16.
Article En | MEDLINE | ID: mdl-23047242

Adult neurogenesis has been implicated in affective disorders and the action of antidepressants (ADs) although the functional significance of this association is still unclear. The use of animal models closely mimicking human comorbid affective and anxiety disorders seen in the majority of patients should provide relevant novel information. Here, we used a unique genetic mouse model displaying higher trait anxiety (HAB) and comorbid depression-like behavior. We demonstrate that HABs have a lower rate of hippocampal neurogenesis and impaired functional integration of newly born neurons as compared with their normal anxiety/depression-like behavior (NAB) controls. In HABs, chronic treatment with the AD fluoxetine alleviated their higher depression-like behavior and protected them from relapse for 3 but not 7 weeks after discontinuation of the treatment without affecting neurogenesis. Similar to what has been observed in depressed patients, fluoxetine treatment induced anxiogenic-like effects during the early treatment phase in NABs along with a reduction in neurogenesis. On the other hand, treatment with AD drugs with a particularly strong anxiolytic component, namely the neurokinin-1-receptor-antagonist L-822 429 or tianeptine, increased the reduced rate of neurogenesis in HABs up to NAB levels. In addition, challenge-induced hypoactivation of dentate gyrus (DG) neurons in HABs was normalized by all three drugs. Overall, these data suggest that AD-like effects in a psychopathological mouse model are commonly associated with modulation of DG hypoactivity but not neurogenesis, suggesting normalization of hippocampal hypoactivity as a neurobiological marker indicating successful remission. Finally, rather than to higher depression-related behavior, neurogenesis seems to be linked to pathological anxiety.


Antidepressive Agents/pharmacology , Anxiety/physiopathology , Dentate Gyrus/drug effects , Depression/physiopathology , Fluoxetine/pharmacology , Neurogenesis/drug effects , Analysis of Variance , Animals , Antidepressive Agents/therapeutic use , Anxiety/complications , Anxiety/drug therapy , Behavior, Animal , Biomarkers , Dentate Gyrus/pathology , Depression/complications , Depression/drug therapy , Disease Models, Animal , Female , Fluoxetine/therapeutic use , Mice , Piperidines/pharmacology , Recurrence , Remission Induction , Thiazepines/pharmacology
13.
J Psychiatr Res ; 46(2): 239-47, 2012 Feb.
Article En | MEDLINE | ID: mdl-22074993

Sub-chronic tryptophan depletion (SCTD) is proposed as an animal model for depression. Aims were to test the hypothesis and optimise the time of SCTD-induced depression-related behaviour and associated biochemical changes. Sprague Dawley rats were treated with a low tryptophan (TRP) containing diet for 0, 7 or 14 days. Peripheral and central neurochemical markers were measured. SCTD-induced depression-related behaviour was assessed by the forced swim test (FST). Model sensitivity to antidepressants was tested by concomitant treatment with paroxetine. SCTD-induced significant reductions in weight gain and measures of peripheral and central TRP. Corticosterone, aldosterone and kynurenine (K), increased whilst kynurenic acid (KA), an NMDA antagonist decreased. 5-HT(2) receptor binding Bmax was enhanced but was reversed by paroxetine. Corticosterone and aldosterone were significantly negatively-correlated to weight gain. SCTD increased floating time and reduced swimming time in the FST but were reversed by paroxetine. Aldosterone was increased at 7 and 14 days, whereas other changes maximised at 14 days. Aldosterone may be an early marker or causal link for depression development. Increased corticosterone and brain tissue 5-HT-receptor density may be correlates of depressive behaviour. Consequential increases in NMDA signalling through increased K/KA ratios suggest the model may be useful for testing novel antidepressants.


Depression/etiology , Tryptophan/deficiency , Aldosterone/blood , Analysis of Variance , Animals , Body Weight/physiology , Brain/metabolism , Calcium-Binding Proteins/blood , Catecholamines/metabolism , Chromatography, High Pressure Liquid/methods , Corticosterone/blood , DNA-Binding Proteins/blood , Depression/pathology , Diet/adverse effects , Disease Models, Animal , Kynurenic Acid/metabolism , Kynurenine/metabolism , Kynurenine/pharmacokinetics , Male , Nerve Tissue Proteins/blood , Nucleobindins , Paroxetine/therapeutic use , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Selective Serotonin Reuptake Inhibitors/therapeutic use , Swimming/psychology , Time Factors , Tritium/pharmacokinetics
14.
Neuropharmacology ; 62(1): 304-12, 2012 Jan.
Article En | MEDLINE | ID: mdl-21835188

Preclinical and some clinical studies suggest a relationship between perturbation in magnesium (Mg(2+)) homeostasis and pathological anxiety, although the underlying mechanisms remain largely unknown. Since there is evidence that Mg(2+) modulates the hypothalamic-pituitary adrenal (HPA) axis, we tested whether enhanced anxiety-like behaviour can be reliably elicited by dietary Mg(2+) deficiency and whether Mg(2+) deficiency is associated with altered HPA axis function. Compared with controls, Mg(2+) deficient mice did indeed display enhanced anxiety-related behaviour in a battery of established anxiety tests. The enhanced anxiety-related behaviour of Mg(2+) deficient mice was sensitive to chronic desipramine treatment in the hyponeophagia test and to acute diazepam treatment in the open arm exposure test. Mg(2+) deficiency caused an increase in the transcription of the corticotropin releasing hormone in the paraventricular hypothalamic nucleus (PVN), and elevated ACTH plasma levels, pointing to an enhanced set-point of the HPA axis. Chronic treatment with desipramine reversed the identified abnormalities of the stress axis. Functional mapping of neuronal activity using c-Fos revealed hyper-excitability in the PVN of anxious Mg(2+) deficient mice and its normalisation through diazepam treatment. Overall, the present findings demonstrate the robustness and validity of the Mg(2+) deficiency model as a mouse model of enhanced anxiety, showing sensitivity to treatment with anxiolytics and antidepressants. It is further suggested that dysregulations in the HPA axis may contribute to the hyper-emotionality in response to dietary induced hypomagnesaemia. This article is part of a Special Issue entitled 'Anxiety and Depression'.


Anxiety/etiology , Anxiety/pathology , Hypothalamo-Hypophyseal System/physiopathology , Magnesium Deficiency/complications , Pituitary-Adrenal System/physiopathology , Adrenocorticotropic Hormone/blood , Analysis of Variance , Animals , Anxiety/blood , Anxiety/drug therapy , Corticosterone , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Dark Adaptation , Desipramine/pharmacology , Desipramine/therapeutic use , Disease Models, Animal , Exploratory Behavior , Fever , Food Deprivation , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Magnesium , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Paroxetine/pharmacology , Paroxetine/therapeutic use , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Radioimmunoassay , Reaction Time/drug effects , Reaction Time/physiology , Stress, Psychological/drug therapy
15.
Neuroscience ; 183: 71-80, 2011 Jun 02.
Article En | MEDLINE | ID: mdl-21458543

Anxiety disorders are the most prevalent central nervous system diseases imposing a high social burden to our society. Emotional processing is particularly controlled by GABA-ergic transmission in the amygdala. Using in situ hybridization and immunohistochemistry we now investigated changes in the expression of GABA synthesizing enzymes (GAD65 and GAD67), GABA(A) (α1-5, ß1-3, γ1-2) and GABA(B) receptor subunits (GBBR1, GBBR2) in amygdaloid nuclei of high anxiety-related behavior (HAB) mice in comparison to mice selected for normal anxiety-related behavior (NAB). Levels of GAD65 and GAD67 mRNAs and protein, as well as those of GABA were increased in the amygdala of HAB mice. Relative to NAB controls, mRNA expression of the GABA(A) receptor subunits ß1, ß2 and γ2 was specifically increased in the basolateral amygdala of HAB mice while transcription of α5 and γ1 subunits was reduced in the central and medial amygdala. On the protein level, increases in ß2 and γ2 subunit immunoreactivities were evident in the basolateral amygdala of HAB mice. No change in GABA(B) receptor expression was observed. These findings point towards an imbalanced GABA-ergic neurotransmission in the amygdala of HAB mice. On the other hand, FosB, a marker for neuronal activity, was increased in principal neurons of the basolateral amygdala in HAB mice, reflecting activation of excitatory neurons, possibly as a consequence of reduced GABA-ergic tonic inhibition through α5 and γ1 containing receptors. Ultimately these mechanisms may lead to the compensatory activation of GABA transmission, as indicated by the increased expression of GAD65/67 in HAB mice.


Amygdala/metabolism , Anxiety/metabolism , Gene Expression Regulation/physiology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Analysis of Variance , Animals , Anxiety/pathology , Disease Models, Animal , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Male , Maze Learning , Mice , Protein Subunits/genetics , Protein Subunits/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Statistics, Nonparametric
16.
Genes Brain Behav ; 8(8): 744-52, 2009 Nov.
Article En | MEDLINE | ID: mdl-19674120

Fear extinction is impaired in psychiatric disorders such as post-traumatic stress disorder and schizophrenia, which have a major genetic component. However, the genetic factors underlying individual variability in fear extinction remain to be determined. By comparing a panel of inbred mouse strains, we recently identified a strain, 129S1/SvImJ (129S1), that exhibits a profound and selective deficit in Pavlovian fear extinction, and associated abnormalities in functional activation of a key prefrontal-amygdala circuit, as compared with C57BL/6J. The first aim of the present study was to assess fear extinction across multiple 129 substrains representing the strain's four different genetic lineages (parental, steel, teratoma and contaminated). Results showed that 129P1/ReJ, 129P3/J, 129T2/SvEmsJ and 129X1/SvJ exhibited poor fear extinction, relative to C57BL/6J, while 129S1 showed evidence of fear incubation. On the basis of these results, the second aim was to further characterize the nature and specificity of the extinction phenotype in 129S1, as an exemplar of the 129 substrains. Results showed that the extinction deficit in 129S1 was neither the result of a failure to habituate to a sensitized fear response nor an artifact of a fear response to (unconditioned) tone per se. A stronger conditioning protocol (i.e. five x higher intensity shocks) produced an increase in fear expression in 129S1, relative to C57BL/6J, due to rapid rise in freezing during tone presentation. Taken together, these data show that impaired fear extinction is a phenotypic feature common across 129 substrains, and provide preliminary evidence that impaired fear extinction in 129S1 may reflect a pro-fear incubation-like process.


Conditioning, Psychological/physiology , Extinction, Psychological/physiology , Fear/physiology , Genetic Variation/genetics , Genome/genetics , Acoustic Stimulation , Animals , Avoidance Learning/physiology , Brain Chemistry/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Neuropsychological Tests , Phenotype , Species Specificity
17.
Stress ; 12(1): 58-69, 2009 Jan.
Article En | MEDLINE | ID: mdl-19116889

Chronic subordinate colony (CSC) housing has been recently validated as a murine model of chronic psychosocial stress which induces alterations of stress-related parameters including decreased body-weight gain and an increased level of anxiety in comparison with single housed control (SHC) mice. By using immunohistochemical immediate early gene (IEG) mapping we investigated whether CSC housing causes alterations in neuronal activation patterns in limbic areas including the amygdala, hippocampus, septum and the periaqueductal gray (PAG) and hypothalamic paraventricular nucleus (PVN). While CSC housing increased basal Zif-268 expression in the nucleus accumbens shell compared to SHC, IEG responses to subsequent open arm (OA) exposure were attenuated in the ventral and intermediate sub-regions of the lateral septum, parvocellular PVN and the dorsal CA3 region of the hippocampus of CSC compared with SHC mice. In contrast, a potentiated c-Fos response in CSC mice was observed in the dorsomedial PAG after OA exposure. Confirming previous findings obtained on the elevated plus-maze, an enhanced anxiety-related behavior in CSC compared with SHC mice was also observed during OA exposure. In order to investigate the appropriate control conditions for CSC housing, group housed control (GHC) mice were additionally included in the behavioral testing. Interestingly, GHC as well as CSC mice showed significantly less risk assessment/exploratory behavior during OA exposure compared with SHC mice indicating that group housing itself is stressful for mice and not an adequate control for the CSC paradigm. Overall, CSC housing is an ethologically relevant chronic psychosocial stressor which results in an elevated sensitivity to a subsequent novel, aversive challenge. However, the CSC-induced increase in anxiety-related behavior was accompanied by differences in neuronal activation, compared with SHC, in defined sub-regions of brain areas known to be involved in the processing of emotionality and stress responses.


Brain/physiopathology , Social Dominance , Stress, Psychological/physiopathology , Animals , Anxiety/psychology , Early Growth Response Protein 1/metabolism , Gene Expression , Housing, Animal , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/metabolism
18.
Neuroscience ; 158(4): 1717-30, 2009 Feb 18.
Article En | MEDLINE | ID: mdl-19121371

There is growing evidence that neuropeptide Y (NPY) acting through Y1 and Y2 receptors has a prominent role in modulating anxiety- and depression-like behavior in rodents. However, a role of other Y-receptors like that of Y4 receptors in this process is poorly understood. We now investigated male Y2, Y4 single and Y2/Y4 double knockout mice in behavioral paradigms for changes in motor activity, anxiety and depression-like behavior. Motor activity was increased in Y2, Y4 and Y2/Y4 knockout mice under changing and stressful conditions, but not altered in a familiar environment. Y4 and Y2 knockout mice revealed an anxiolytic phenotype in the light/dark test, marble burying test and in stress-induced hyperthermia, and reduced depression-like behavior in the forced swim and tail suspension tests. In Y2/Y4 double knockout mice, the response in the light/dark test and in the forced swim test was further enhanced compared with Y4 and Y2 knockout mice, respectively. High levels of Y4 binding sites were observed in brain stem nuclei including nucleus of solitary tract and area postrema. Lower levels were found in the medial amygdala and hypothalamus. Peripheral administration of pancreatic polypeptide (PP) induced Y4 receptor-dependent c-Fos expression in brain stem, hypothalamus and amygdala. PP released peripherally from the pancreas in response to food intake, may act not only as a satiety signal but also modulate anxiety-related locomotion.


Depression/genetics , Depression/physiopathology , Exploratory Behavior/physiology , Motor Activity/genetics , Receptors, Neuropeptide Y/deficiency , Amygdala/metabolism , Animals , Autoradiography/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hindlimb Suspension/methods , Hyperthermia, Induced , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Polypeptide/pharmacology , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Swimming/physiology
19.
Amino Acids ; 36(1): 147-58, 2009 Jan.
Article En | MEDLINE | ID: mdl-18975044

A relationship between zinc (Zn)-deficiency and mood disorders has been suspected. Here we examined for the first time whether experimentally-induced Zn-deficiency in mice would alter depression- and anxiety-related behaviour assessed in established tests and whether these alterations would be sensitive to antidepressant treatment. Mice receiving a Zn-deficient diet (40% of daily requirement) had similar homecage and open field activity compared to normally fed mice, but displayed enhanced depression-like behaviour in both the forced swim and tail suspension tests which was reversed by chronic desipramine treatment. An anxiogenic effect of Zn-deficiency prevented by chronic desipramine and Hypericum perforatum treatment was observed in the novelty suppressed feeding test, but not in other anxiety tests performed. Zn-deficient mice showed exaggerated stress-evoked immediate-early gene expression in the amygdala which was normalised following DMI treatment. Taken together these data support the link between low Zn levels and depression-like behaviour and suggest experimentally-induced Zn deficiency as a putative model of depression in mice.


Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Depression/drug therapy , Depression/metabolism , Limbic System/drug effects , Limbic System/physiopathology , Zinc/deficiency , Animals , Body Weight/drug effects , Limbic System/metabolism , Limbic System/physiology , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects
...