Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Psychoneuroendocrinology ; 48: 41-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24995583

ABSTRACT

Depression and anxiety disorders are often characterized by altered hypothalamic-pituitary-adrenal (HPA) axis re-/activity. However, the presence of a molecular link between dysbalanced neuroendocrine regulation and psychopathologies is not yet fully established. Earlier, we reported that high (HAB), normal (NAB) and low (LAB) anxiety-related behavior mice express divergent anxiety-related and passive/active coping phenotypes. Here, we studied mechanisms that might contribute to the different HPA axis reactivity observed in HAB, NAB and LAB mice and their involvement in the regulation of anxiety-related behavior and passive/active coping style. We found that HAB mice respond with significantly reduced corticosterone (CORT) secretion to an acute stressful stimulus and a blunted response in the Dex/CRH test compared to NAB and LAB mice. At the molecular level, higher expression of the glucocorticoid receptor (GR/Nr3c1) and decreased corticotropin-releasing hormone receptor 1 (CRHR1) expression were observed in the pituitary of HAB mice. We further analyzed whether these stress mediators differed between the HAB, NAB and LAB lines in limbic system-associated brain regions and whether their interplay contributes to the phenotype. Interestingly, not only in the pituitary but also in almost all brain regions investigated, GR expression was significantly higher in HAB mice. In contrast, the amount of CORT in the brain structures analyzed was significantly lower in these animals. The expression of CRHR1 varied in the prefrontal cortex only. Since glucocorticoids regulate both GR and CRHR1, we treated HAB and NAB mice chronically with CORT. After 6 weeks of administration, reduced anxiety- and depression-like behaviors were observed in HAB mice, whereas increased anxiety was found in NABs. In both groups, GR, but not CRHR1, were significantly reduced. Taken together, our study proposes HAB mice as an animal model of simultaneous features of increased anxiety-related and depression-like behaviors with blunted HPA axis reactivity suggesting a dysregulated GR/CORT system as one key mechanism behind their phenotype.


Subject(s)
Anxiety Disorders/pathology , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Behavior, Animal/physiology , Corticotropin-Releasing Hormone/metabolism , Cortisone/metabolism , Disease Models, Animal , Hypothalamo-Hypophyseal System/physiopathology , Mice , Neurosecretory Systems/physiology , Pituitary-Adrenal System/physiopathology , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Glucocorticoid/genetics , Signal Transduction , Transcriptome
2.
Front Behav Neurosci ; 7: 103, 2013.
Article in English | MEDLINE | ID: mdl-23966915

ABSTRACT

We established mouse models of extremes in trait anxiety, which are based on selective breeding for low vs. normal vs. high open-arm exploration on the elevated plus-maze. Genetically selected low anxiety-related behavior (LAB) coincided with hyperactivity in the home cage. Given the fact that several psychiatric disorders such as schizophrenia, mania, and attention deficit hyperactivity disorder (ADHD) share hyperactivity symptom, we systematically examined LAB mice with respect to unique and overlapping endophenotypes of the three diseases. To this end Venn diagrams were used as an instrument for discrimination of possible models. We arranged the endophenotypes in Venn diagrams and translated them into different behavioral tests. LAB mice showed elevated levels of locomotion in the open field (OF) test with deficits in habituation, compared to mice bred for normal (NAB) and high anxiety-related behavior (HAB). Cross-breeding of hypoactive HAB and hyperactive LAB mice resulted in offspring showing a low level of locomotion comparable to HAB mice, indicating that the HAB alleles are dominant over LAB alleles in determining the level of locomotion. In a holeboard test, LAB mice spent less time in hole exploration, as shown in patients with schizophrenia and ADHD; however, LAB mice displayed no impairments in social interaction and prepulse inhibition (PPI), implying a unlikelihood of LAB as an animal model of schizophrenia. Although LAB mice displayed hyperarousal, active coping styles, and cognitive deficits, symptoms shared by mania and ADHD, they failed to reveal the classic manic endophenotypes, such as increased hedonia and object interaction. The neuroleptic haloperidol reduced locomotor activity in all mouse lines. The mood stabilizer lithium and the psychostimulant amphetamine, in contrast, selectively reduced hyperactivity in LAB mice. Based on the behavioral and pharmacological profiles, LAB mice are suggested as a novel rodent model of ADHD-like symptoms.

3.
BMC Genomics ; 13: 579, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23114097

ABSTRACT

BACKGROUND: The hypothalamic-pituitary-adrenal (HPA) axis is essential to control physiological stress responses in mammals. Its dysfunction is related to several mental disorders, including anxiety and depression. The aim of this study was to identify genetic loci underlying the endocrine regulation of the HPA axis. METHOD: High (HAB) and low (LAB) anxiety-related behaviour mice were established by selective inbreeding of outbred CD-1 mice to model extremes in trait anxiety. Additionally, HAB vs. LAB mice exhibit comorbid characteristics including a differential corticosterone response upon stress exposure. We crossbred HAB and LAB lines to create F1 and F2 offspring. To identify the contribution of the endocrine phenotypes to the total phenotypic variance, we examined multiple behavioural paradigms together with corticosterone secretion-based phenotypes in F2 mice by principal component analysis. Further, to pinpoint the genomic loci of the quantitative trait of the HPA axis stress response, we conducted genome-wide multipoint oligogenic linkage analyses based on Bayesian Markov chain Monte Carlo approach as well as parametric linkage in three-generation pedigrees, followed by a two-dimensional scan for epistasis and association analysis in freely segregating F2 mice using 267 single-nucleotide polymorphisms (SNPs), which were identified to consistently differ between HAB and LAB mice as genetic markers. RESULTS: HPA axis reactivity measurements and behavioural phenotypes were represented by independent principal components and demonstrated no correlation. Based on this finding, we identified one single quantitative trait locus (QTL) on chromosome 3 showing a very strong evidence for linkage (2ln (L-score) > 10, LOD > 23) and significant association (lowest Bonferroni adjusted p < 10-28) to the neuroendocrine stress response. The location of the linkage peak was estimated at 42.3 cM (95% confidence interval: 41.3 - 43.3 cM) and was shown to be in epistasis (p-adjusted < 0.004) with the locus at 35.3 cM on the same chromosome. The QTL harbours genes involved in steroid synthesis and cardiovascular effects. CONCLUSION: The very prominent effect on stress-induced corticosterone secretion of the genomic locus on chromosome 3 and its involvement in epistasis highlights the critical role of this specific locus in the regulation of the HPA axis.


Subject(s)
Anxiety/genetics , Anxiety/physiopathology , Chromosomes, Mammalian/genetics , Endocrine System/physiology , Quantitative Trait Loci/genetics , Stress, Physiological/genetics , Adrenal Glands/physiopathology , Animals , Endocrine System/metabolism , Female , Genetic Markers/genetics , Hypothalamus/physiopathology , Male , Mice , Phenotype , Pituitary Gland/physiopathology
4.
Neurobiol Learn Mem ; 98(1): 56-65, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22579802

ABSTRACT

Patients diagnosed for anxiety disorders often display faster acquisition and slower extinction of learned fear. To gain further insights into the mechanisms underlying these phenomenona, we studied conditioned fear in mice originating form a bi-directional selective breeding approach, which is based on elevated plus-maze behavior and results in CD1-derived high (HAB), normal (NAB), and low (LAB) anxiety-related behavior mice. HAB mice displayed pronounced cued-conditioned fear compared to NAB/CD1 and LAB mice that coincided with increased phosphorylation of the protein kinase B (AKT) in the basolateral amygdala 45 min after conditioning. No similar changes were observed after non-associative immediate shock presentations. Fear extinction of recent but not older fear memories was preserved. However, HAB mice were more prone to relapse of conditioned fear with the passage of time. HAB mice also displayed higher levels of contextual fear compared to NAB and LAB mice and exaggerated avoidance following step-down avoidance training. Interestingly, HAB mice showed lower and LAB mice higher levels of acoustic startle responses compared to NAB controls. The increase in arousal observed in LAB mice coincided with the general absence of conditioned freezing. Taken together, our results suggest that the genetic predisposition to high anxiety-related behavior may increase the risk of forming traumatic memories, phobic-like fear and avoidance behavior following aversive encounters, with a clear bias towards passive coping styles. In contrast, genetic predisposition to low anxiety-related and high risk-taking behavior seems to be associated with an increase in active coping styles. Our data imply changes in AKT phosphorylation as a therapeutic target for the prevention of exaggerated fear memories.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Avoidance Learning/physiology , Behavior, Animal/physiology , Conditioning, Psychological/physiology , Fear/physiology , Proto-Oncogene Proteins c-akt/metabolism , Acoustic Stimulation , Animals , Anxiety/psychology , Cues , Disease Models, Animal , Electroshock , Extinction, Psychological/physiology , Male , Mice , Phosphorylation
5.
PLoS One ; 6(8): e23604, 2011.
Article in English | MEDLINE | ID: mdl-21897848

ABSTRACT

Behavioral endophenotypes are determined by a multitude of counteracting but precisely balanced molecular and physiological mechanisms. In this study, we aim to identify potential novel molecular targets that contribute to the multigenic trait "anxiety". We used microarrays to investigate the gene expression profiles of different brain regions within the limbic system of mice which were selectively bred for either high (HAB) or low (LAB) anxiety-related behavior, and also show signs of comorbid depression-like behavior. We identified and confirmed sex-independent differences in the basal expression of 13 candidate genes, using tissue from the entire brain, including coronin 7 (Coro7), cathepsin B (Ctsb), muscleblind-like 1 (Mbnl1), metallothionein 1 (Mt1), solute carrier family 25 member 17 (Slc25a17), tribbles homolog 2 (Trib2), zinc finger protein 672 (Zfp672), syntaxin 3 (Stx3), ATP-binding cassette, sub-family A member 2 (Abca2), ectonucleotide pyrophosphatase/phosphodiesterase 5 (Enpp5), high mobility group nucleosomal binding domain 3 (Hmgn3) and pyruvate dehydrogenase beta (Pdhb). Additionally, we confirmed brain region-specific differences in the expression of synaptotagmin 4 (Syt4).Our identification of about 90 polymorphisms in Ctsb suggested that this gene might play a critical role in shaping our mouse model's behavioral endophenotypes. Indeed, the assessment of anxiety-related and depression-like behaviors of Ctsb knock-out mice revealed an increase in depression-like behavior in females. Altogether, our results suggest that Ctsb has significant effects on emotionality, irrespective of the tested mouse strain, making it a promising target for future pharmacotherapy.


Subject(s)
Anxiety/enzymology , Anxiety/genetics , Cathepsin B/genetics , Gene Expression Profiling , Animals , Behavior, Animal , Brain/metabolism , Cathepsin B/deficiency , Endophenotypes , Female , Gene Knockout Techniques , In Situ Hybridization , Male , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Sequence Analysis, DNA
6.
Biol Psychiatry ; 70(11): 1074-82, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21791337

ABSTRACT

BACKGROUND: Although anxiety disorders are the most prevalent psychiatric disorders, no molecular biomarkers exist for their premorbid diagnosis, accurate patient subcategorization, or treatment efficacy prediction. To unravel the neurobiological underpinnings and identify candidate biomarkers and affected pathways for anxiety disorders, we interrogated the mouse model of high anxiety-related behavior (HAB), normal anxiety-related behavior (NAB), and low anxiety-related behavior (LAB) employing a quantitative proteomics and metabolomics discovery approach. METHODS: We compared the cingulate cortex synaptosome proteomes of HAB and LAB mice by in vivo (15)N metabolic labeling and mass spectrometry and quantified the cingulate cortex metabolomes of HAB/NAB/LAB mice. The combined data sets were used to identify divergent protein and metabolite networks by in silico pathway analysis. Selected differentially expressed proteins and affected pathways were validated with immunochemical and enzymatic assays. RESULTS: Altered levels of up to 300 proteins and metabolites were found between HAB and LAB mice. Our data reveal alterations in energy metabolism, mitochondrial import and transport, oxidative stress, and neurotransmission, implicating a previously nonhighlighted role of mitochondria in modulating anxiety-related behavior. CONCLUSIONS: Our results offer insights toward a molecular network of anxiety pathophysiology with a focus on mitochondrial contribution and provide the basis for pinpointing affected pathways in anxiety-related behavior.


Subject(s)
Anxiety/metabolism , Anxiety/physiopathology , Metabolomics , Mitochondria/metabolism , Proteomics , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Anxiety/drug therapy , Anxiety/genetics , Behavior, Animal/physiology , Citric Acid Cycle/genetics , Disease Models, Animal , Energy Metabolism/genetics , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology , Gyrus Cinguli/ultrastructure , Mass Spectrometry , Metabolic Networks and Pathways/genetics , Mice , Mitochondria/genetics , Models, Biological , Nitrogen Isotopes/administration & dosage , Nitrogen Isotopes/blood , Nitrogen Isotopes/metabolism , Oxidative Stress/genetics , Phosphorylation/genetics , Synaptic Transmission/genetics , Synaptosomes/metabolism
7.
PLoS One ; 6(2): e16849, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21386891

ABSTRACT

The propensity to develop an anxiety disorder is thought to be determined by genetic and environmental factors. Here we investigated the relationship between a genetic predisposition to trait anxiety and experience-based learned fear in a psychopathological mouse model. Male CD-1 mice selectively bred for either high (HAB), or normal (NAB) anxiety-related behaviour on the elevated plus maze were subjected to classical fear conditioning. During conditioning both mouse lines showed increased fear responses as assessed by freezing behaviour. However, 24 h later, HAB mice displayed more pronounced conditioned responses to both a contextual or cued stimulus when compared with NAB mice. Interestingly, 6 h and already 1 h after fear conditioning, freezing levels were high in HAB mice but not in NAB mice. These results suggest that trait anxiety determines stronger fear memory and/or a weaker ability to inhibit fear responses in the HAB line. The enhanced fear response of HAB mice was attenuated by treatment with either the α(2,3,5)-subunit selective benzodiazepine partial agonist L-838,417, corticosterone or the selective neurokinin-1 receptor antagonist L-822,429. Overall, the HAB mouse line may represent an interesting model (i) for identifying biological factors underlying misguided conditioned fear responses and (ii) for studying novel anxiolytic pharmacotherapies for patients with fear-associated disorders, including post-traumatic stress disorder and phobias.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety Disorders/drug therapy , Anxiety Disorders/pathology , Disease Models, Animal , Fear/physiology , Mice , Animals , Anxiety Disorders/complications , Anxiety Disorders/psychology , Behavior, Animal , Corticosterone/therapeutic use , Disease Progression , Drug Evaluation, Preclinical , Expressed Emotion/physiology , Fluorobenzenes/therapeutic use , Male , Neurokinin-1 Receptor Antagonists , Phobic Disorders/complications , Phobic Disorders/drug therapy , Phobic Disorders/pathology , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/psychology , Piperidines/therapeutic use , Psychopathology , Triazoles/therapeutic use , Up-Regulation
8.
Soc Neurosci ; 6(2): 156-68, 2011.
Article in English | MEDLINE | ID: mdl-20661836

ABSTRACT

Brain arginine vasopressin (AVP) not only regulates male social behavior and emotionality, but also promotes maternal behavior, as has been shown in rats. In our CD1 mice breed for high (HAB) or low (LAB) anxiety-related behavior, LAB mice have markedly less AVP mRNA expression in the hypothalamic paraventricular nucleus compared with HAB mice. Together these findings suggest that HAB and LAB mice represent a good model to assess the role of AVP in mouse maternal behavior. Therefore, we studied maternal care of HAB and LAB mouse dams and investigated the impact of maternal care on the offspring's anxiety in a cross-fostering paradigm. In comparison with HAB dams, LABs displayed less maternal care. Daily acute intracerebroventricular infusions of AVP in early lactation increased maternal care of LAB dams and acted anxiogenically. Cross-fostering on postnatal day 5 did not alter separation-induced high and low ultrasonic vocalization calling frequency, a measure of inborn anxiety, in HAB and LAB offspring, respectively. However, adult cross-fostered HAB mice displayed a trend towards decreased anxiety on the elevated plus-maze, which was still significantly higher compared with LAB mice. The low levels of depressive-like behavior, stress-reactivity, and hypothalamic AVP mRNA expression in adult LAB offspring were found to be independent of cross-fostering. In conclusion, the HAB/LAB differences in maternal care and anxiety are robust and strongly depend on differences in the AVP system. The seemingly rigid genetic predisposition to hyperanxiety can only be moderately attenuated by the received nurturing.


Subject(s)
Anxiety/genetics , Behavior, Animal/physiology , Brain/metabolism , Maternal Behavior/physiology , Vasopressins/metabolism , Animals , Anxiety/metabolism , Female , Male , Mice
9.
J Neurosci ; 30(27): 9103-16, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20610744

ABSTRACT

Urocortin 3 (UCN3) is strongly expressed in specific nuclei of the rodent brain, at sites distinct from those expressing urocortin 1 and urocortin 2, the other endogenous ligands of corticotropin-releasing hormone receptor type 2 (CRH-R2). To determine the physiological role of UCN3, we generated UCN3-deficient mice, in which the UCN3 open reading frame was replaced by a tau-lacZ reporter gene. By means of this reporter gene, the nucleus parabrachialis and the premammillary nucleus were identified as previously unknown sites of UCN3 expression. Additionally, the introduced reporter gene enabled the visualization of axonal projections of UCN3-expressing neurons from the superior paraolivary nucleus to the inferior colliculus and from the posterodorsal part of the medial amygdala to the principal nucleus of the bed nucleus of the stria terminalis, respectively. The examination of tau-lacZ reporter gene activity throughout the brain underscored a predominant expression of UCN3 in nuclei functionally connected to the accessory olfactory system. Male and female mice were comprehensively phenotyped but none of the applied tests provided indications for a role of UCN3 in the context of hypothalamic-pituitary-adrenocortical axis regulation, anxiety- or depression-related behavior. However, inspired by the prevalent expression throughout the accessory olfactory system, we identified alterations in social discrimination abilities of male and female UCN3 knock-out mice that were also present in male CRH-R2 knock-out mice. In conclusion, our results suggest a novel role for UCN3 and CRH-R2 related to the processing of social cues and to the establishment of social memories.


Subject(s)
Discrimination, Psychological/physiology , Interpersonal Relations , Receptors, Corticotropin-Releasing Hormone/metabolism , Recognition, Psychology/physiology , Urocortins/metabolism , Acoustic Stimulation/methods , Animals , Brain/cytology , Brain/metabolism , Circadian Rhythm/physiology , Corticosterone/blood , Fear/physiology , Female , Gene Expression Regulation/genetics , Hypothalamo-Hypophyseal System/metabolism , Inhibition, Psychological , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Odorants , Olfactory Pathways/physiology , Perception/physiology , Pituitary-Adrenal System/embryology , Radioimmunoassay/methods , Receptors, Corticotropin-Releasing Hormone/deficiency , Reflex, Startle/genetics , Statistics, Nonparametric , Swimming/physiology , Urocortins/deficiency
10.
PLoS One ; 4(11): e7821, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19915716

ABSTRACT

The identification of differentially regulated proteins in animal models of psychiatric diseases is essential for a comprehensive analysis of associated psychopathological processes. Mass spectrometry is the most relevant method for analyzing differences in protein expression of tissue and body fluid proteomes. However, standardization of sample handling and sample-to-sample variability are problematic. Stable isotope metabolic labeling of a proteome represents the gold standard for quantitative mass spectrometry analysis. The simultaneous processing of a mixture of labeled and unlabeled samples allows a sensitive and accurate comparative analysis between the respective proteomes. Here, we describe a cost-effective feeding protocol based on a newly developed (15)N bacteria diet based on Ralstonia eutropha protein, which was applied to a mouse model for trait anxiety. Tissue from (15)N-labeled vs. (14)N-unlabeled mice was examined by mass spectrometry and differences in the expression of glyoxalase-1 (GLO1) and histidine triad nucleotide binding protein 2 (Hint2) proteins were correlated with the animals' psychopathological behaviors for methodological validation and proof of concept, respectively. Additionally, phenotyping unraveled an antidepressant-like effect of the incorporation of the stable isotope (15)N into the proteome of highly anxious mice. This novel phenomenon is of considerable relevance to the metabolic labeling method and could provide an opportunity for the discovery of candidate proteins involved in depression-like behavior. The newly developed (15)N bacteria diet provides researchers a novel tool to discover disease-relevant protein expression differences in mouse models using quantitative mass spectrometry.


Subject(s)
Nitrogen Isotopes/metabolism , Proteomics/methods , Animals , Anxiety/genetics , Cupriavidus necator/metabolism , Depression/genetics , Disease Models, Animal , Histidine/chemistry , Lactoylglutathione Lyase/biosynthesis , Mass Spectrometry/methods , Maze Learning , Mice , Phenotype , Proteome
11.
PLoS One ; 4(4): e5129, 2009.
Article in English | MEDLINE | ID: mdl-19357765

ABSTRACT

BACKGROUND: To investigate neurobiological correlates of trait anxiety, CD1 mice were selectively bred for extremes in anxiety-related behavior, with high (HAB) and low (LAB) anxiety-related behavior mice additionally differing in behavioral tests reflecting depression-like behavior. METHODOLOGY/ PRINCIPAL FINDINGS: In this study, microarray analysis, in situ hybridization, quantitative real-time PCR and immunohistochemistry revealed decreased expression of the vasopressin gene (Avp) in the hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei of adult LAB mice compared to HAB, NAB (normal anxiety-related behavior) and HABxLAB F1 intercross controls, without detecting differences in receptor expression or density. By sequencing the regions 2.5 kbp up- and downstream of the Avp gene locus, we could identify several polymorphic loci, differing between the HAB and LAB lines. In the gene promoter, a deletion of twelve bp Delta(-2180-2191) is particularly likely to contribute to the reduced Avp expression detected in LAB animals under basal conditions. Indeed, allele-specific transcription analysis of F1 animals revealed a hypomorphic LAB-specific Avp allele with a reduced transcription rate by 75% compared to the HAB-specific allele, thus explaining line-specific Avp expression profiles and phenotypic features. Accordingly, intra-PVN Avp mRNA levels were found to correlate with anxiety-related and depression-like behaviors. In addition to this correlative evidence, a significant, though moderate, genotype/phenotype association was demonstrated in 258 male mice of a freely-segregating F2 panel, suggesting a causal contribution of the Avp promoter deletion to anxiety-related behavior. DISCUSSION: Thus, the identification of polymorphisms in the Avp gene promoter explains gene expression differences in association with the observed phenotype, thus further strengthening the concept of the critical involvement of centrally released AVP in trait anxiety.


Subject(s)
Alleles , Anxiety/genetics , Arginine Vasopressin/genetics , Behavior, Animal/physiology , Animals , Anxiety/physiopathology , Arginine Vasopressin/metabolism , Depression/genetics , Depression/physiopathology , Female , Gene Expression Profiling , Humans , Male , Mice , Motor Activity/physiology , Neuropsychological Tests , Oligonucleotide Array Sequence Analysis , Oxytocin/genetics , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Supraoptic Nucleus/metabolism
12.
PLoS One ; 4(4): e5346, 2009.
Article in English | MEDLINE | ID: mdl-19399175

ABSTRACT

There is evidence for a disturbed perception and processing of emotional information in pathological anxiety. Using a rat model of trait anxiety generated by selective breeding, we previously revealed differences in challenge-induced neuronal activation in fear/anxiety-related brain areas between high (HAB) and low (LAB) anxiety rats. To confirm whether findings generalize to other species, we used the corresponding HAB/LAB mouse model and investigated c-Fos responses to elevated open arm exposure. Moreover, for the first time we included normal anxiety mice (NAB) for comparison. The results confirm that HAB mice show hyperanxious behavior compared to their LAB counterparts, with NAB mice displaying an intermediate anxiety phenotype. Open arm challenge revealed altered c-Fos response in prefrontal-cortical, limbic and hypothalamic areas in HAB mice as compared to LAB mice, and this was similar to the differences observed previously in the HAB/LAB rat lines. In mice, however, additional differential c-Fos response was observed in subregions of the amygdala, hypothalamus, nucleus accumbens, midbrain and pons. Most of these differences were also seen between HAB and NAB mice, indicating that it is predominately the HAB line showing altered neuronal processing. Hypothalamic hypoactivation detected in LAB versus NAB mice may be associated with their low-anxiety/high-novelty-seeking phenotype. The detection of similarly disturbed activation patterns in a key set of anxiety-related brain areas in two independent models reflecting psychopathological states of trait anxiety confirms the notion that the altered brain activation in HAB animals is indeed characteristic of enhanced (pathological) anxiety, providing information for potential targets of therapeutic intervention.


Subject(s)
Anxiety/physiopathology , Neurons/physiology , Animals , Anxiety/genetics , Anxiety/pathology , Brain/pathology , Brain/physiopathology , Breeding , Disease Models, Animal , Female , Male , Mice , Phenotype , Proto-Oncogene Proteins c-fos/metabolism , Rats , Species Specificity , Stress, Physiological , Tissue Distribution
13.
Psychoneuroendocrinology ; 33(6): 839-62, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18502051

ABSTRACT

Affective disorders such as major depression are among the most prevalent and costly diseases of the central nervous system, but the underlying mechanisms are still poorly understood. In recent years, it has become evident that alterations of the stress hormone system, in particular dysfunctions (hyper- or hypo-activity) of the hypothalamic-pituitary-adrenal (HPA) axis, play a prominent role in the development of major depressive disorders. Therefore, we aimed to generate a new animal model comprising these neuroendocrine core symptoms in order to unravel parameters underlying increased or decreased stress reactivity. Starting from a population of outbred mice (parental generation: 100 males and 100 females of the CD-1 strain), two breeding lines were established according to the outcome of a 'stress reactivity test' (SRT), consisting of a 15-min restraint period and tail blood samplings immediately before and after exposure to the stressor. Mice showing a very high or a very low secretion of corticosterone in the SRT, i.e. animals expressing a hyper- or a hypo-reactivity of the HPA axis, were selected for the 'high reactivity' (HR) and the 'low reactivity' (LR) breeding line, respectively. Additionally, a third breeding line was established consisting of animals with an 'intermediate reactivity' (IR) in the SRT. Already in the first generation, i.e. animals derived from breeding pairs selected from the parental generation, significant differences in the reactivity of the HPA axis between HR, IR, and LR mice were observed. Moreover, these differences were found across all subsequent generations and could be increased by selective breeding, which indicates a genetic basis of the respective phenotype. Repeated testing of individuals in the SRT furthermore proved that the observed differences in stress responsiveness are present already early in life and can be regarded as a robust genetic predisposition. Tests investigating the animal's emotionality including anxiety-related behavior, exploratory drive, locomotor activity, and depression-like behavior point to phenotypic similarities with behavioral changes observed in depressive patients. In general, HR males and females were 'hyperactive' in some behavioral paradigms, resembling symptoms of restlessness and agitation often seen in melancholic depression. LR mice, on the other hand, showed more passive-aggressive coping styles, corresponding to signs of retardation and retreat observed in atypical depression. Several morphometric and neuroendocrine findings further support this view. For example, monitoring the circadian rhythm of glucocorticoid secretion revealed clearly increased trough levels in HR mice, resulting in a flattened diurnal rhythm, again adding to the neuroendocrine similarities to patients suffering from melancholic depression. Taken together, our results suggest that distinct mechanisms influencing the function and regulation of the HPA axis are involved in the respective behavioral and neurobiological endophenotypes. Thus, the generated HR/IR/LR mouse lines can be a valuable model to elucidate molecular genetic, neuroendocrine, and behavioral parameters associated with altered stress reactivity, thereby improving our understanding of affective disorders, presumably including the symptomatology and pathophysiology of specific subtypes of major depression.


Subject(s)
Adaptation, Psychological/physiology , Breeding , Disease Models, Animal , Mice, Inbred Strains , Mood Disorders/physiopathology , Stress, Psychological/physiopathology , Animals , Corticosterone/blood , Female , Gene Expression Profiling , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Male , Mice , Mood Disorders/blood , Mood Disorders/genetics , Mood Disorders/pathology , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiology , Psychological Tests , Quantitative Trait, Heritable , Selection, Genetic , Stress, Psychological/genetics , Stress, Psychological/pathology
14.
Eur J Neurosci ; 26(10): 2857-64, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18001282

ABSTRACT

Following secretion from the posterior pituitary, the neuropeptide vasopressin (AVP) stimulates the kidney to retain water, and when released centrally it can contribute to anxiety- and depression-like behaviours. We hypothesized that CD1 mice bred for low trait anxiety (LAB) suffer from a deficit in AVP. Both osmotically stimulated peripheral secretion and intra-paraventricular nucleus (PVN) release of AVP were found decreased in LAB animals compared with normal anxiety (NAB) or high anxiety (HAB) controls. Consequently, in addition to their extreme non-anxiety, LAB mice showed signs of central diabetes insipidus (cDI), including increased fluid intake and reduced urine osmolality, as well as a pathological increase in plasma osmolality upon water deprivation. These cDI symptoms were attenuated by administration of a selective AVP V2 receptor agonist. A single nucleotide polymorphism (SNP) in exon 1 (C(+40)T) of the Avp gene of LAB animals causes an amino acid substitution in the signal peptide of the AVP precursor, and is likely to impair processing and trafficking of the precursor, as suggested by reduced axonal transport of AVP from the hypothalamic PVN, finally contributing to cDI symptoms and low trait anxiety. In an F2 panel, this SNP co-segregated with fluid intake and showed a partial contribution to low anxiety-related behaviour, indicated by its co-segregation with time spent on the open arms of the elevated plus-maze in a subset of F2 mice. Thus, the SNP-associated deficit in plasma and central AVP contributes to signs of cDI and, at least partially, to low trait anxiety, both features being typical of LAB animals.


Subject(s)
Anxiety/genetics , Arginine Vasopressin/deficiency , Diabetes Insipidus/genetics , Polymorphism, Single Nucleotide/genetics , Animals , Anxiety/metabolism , Anxiety/pathology , Behavior, Animal , Deamino Arginine Vasopressin/therapeutic use , Diabetes Insipidus/drug therapy , Diabetes Insipidus/metabolism , Diabetes Insipidus/pathology , Drinking/genetics , Exons , Exploratory Behavior/physiology , Hemostatics/therapeutic use , Male , Maze Learning/physiology , Mice , Osmolar Concentration , Paraventricular Hypothalamic Nucleus/metabolism , Plasma/physiology , Radioimmunoassay/methods , Urine/physiology , Water Deprivation/physiology
15.
J Neurosci ; 27(4): 832-9, 2007 Jan 24.
Article in English | MEDLINE | ID: mdl-17251423

ABSTRACT

The transient receptor potential vanilloid type 1 channel (TRPV1) (formerly called vanilloid receptor VR1) is known for its key role of functions in sensory nerves such as perception of inflammatory and thermal pain. Much less is known about the physiological significance of the TRPV1 expression in the brain. Here we demonstrate that TRPV1 knock-out mice (TRPV1-KO) show less anxiety-related behavior in the light-dark test and in the elevated plus maze than their wild-type littermates with no differences in locomotion. Furthermore, TRPV1-KO mice showed less freezing to a tone after auditory fear conditioning and stress sensitization. This reduction of conditioned and sensitized fear could not be explained by alterations in nociception. Also, tone perception per se was unaffected, as revealed by determination of auditory thresholds through auditory brainstem responses and distortion-product otoacoustic emissions. TRPV1-KO showed also less contextual fear if assessed 1 d or 1 month after strong conditioning protocols. These impairments in hippocampus-dependent learning were mirrored by a decrease in long-term potentiation in the Schaffer collateral-commissural pathway to CA1 hippocampal neurons. Our data provide first evidence for fear-promoting effects of TRPV1 with respect to both innate and conditioned fear and for a decisive role of this receptor in synaptic plasticity.


Subject(s)
Anxiety/physiopathology , Conditioning, Psychological/physiology , Fear/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , TRPV Cation Channels/physiology , Animals , Anxiety/genetics , Anxiety/psychology , Fear/psychology , Long-Term Potentiation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
16.
Neurosci Biobehav Rev ; 31(1): 89-102, 2007.
Article in English | MEDLINE | ID: mdl-16934871

ABSTRACT

Two animal models of trait anxiety, HAB/LAB rats and mice, are described, representing inborn extremes in anxiety-related behavior. The comprehensive phenotypical characterization included basal behavioral features, stress-coping strategies and neuroendocrine responses upon stressor exposure with HAB animals being hyper-anxious, preferring passive coping, emitting more stressor-induced ultrasonic vocalization calls and showing typical peculiarities of the hypothalamic-pituitary-adrenocortical axis and line-specific patterns of Fos expression in the brain indicative of differential neuronal activation. In most cases, unselected Wistar rats and CD1 mice, respectively, displayed intermediate behaviors. In both HAB/LAB rats and mice, the behavioral phenotype has been found to be significantly correlated with the expression of the neuropeptide arginine vasopressin (AVP) at the level of the hypothalamic paraventricular nucleus (PVN). Additional receptor antagonist approaches in HABs confirmed that intra-PVN release of AVP is likely to contribute to hyper-anxiety and depression-like behavior. As shown exemplarily in HAB rats and LAB mice, single nucleotide polymorphisms (SNPs) in regulatory structures of the AVP gene underlie AVP-mediated phenotypic phenomena; in HAB rats, a SNP in the promoter of the AVP gene leads to reduced binding of the transcriptional repressor CBF-A, thus causing AVP overexpression and overrelease. Conversely, in LAB mice, a SNP in the AVP gene seems to cause an amino acid exchange in the signal peptide, presumably leading to a deficit in bioavailable AVP likely to underlie the total hypo-anxiety of LAB mice in combination with signs of central diabetes insipidus. Another feature of LAB mice is overexpression of glyoxalase-I. The functional characterization of this enzyme will determine its involvement in anxiety-related behavior beyond that of a reliable biomarker. The further identification of quantitative trait loci, candidate genes (and their products) and SNPs will not only help to explain inter-individual variation in emotional behavior, but will also reveal novel targets for anxiolytic and antidepressive interventions.


Subject(s)
Anxiety/genetics , Arginine Vasopressin/genetics , Behavior, Animal/physiology , Lactoylglutathione Lyase/genetics , Animals , Anxiety/metabolism , Arginine Vasopressin/metabolism , Brain/metabolism , Gene Expression Regulation , Genetics, Behavioral/methods , Lactoylglutathione Lyase/metabolism , Mice , Mice, Inbred Strains , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/physiology , Rats , Rats, Wistar
17.
Mol Cell Proteomics ; 5(10): 1914-20, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16775081

ABSTRACT

Brain proteome analysis of mice selectively bred for either high or low anxiety-related behavior revealed quantitative and qualitative protein expression differences. The enzyme glyoxalase-I was consistently expressed to a higher extent in low anxiety as compared with high anxiety mice in several brain areas. The same phenotype-dependent difference was also found in red blood cells with normal and cross-mated animals showing intermediate expression profiles of glyoxalase-I. Another protein that showed a different mobility during two-dimensional gel electrophoresis was identified as enolase phosphatase. The presence of both protein markers in red or white blood cells, respectively, creates the opportunity to screen for their expression in clinical blood specimens from patients suffering from anxiety.


Subject(s)
Anxiety/metabolism , Blood Proteins/metabolism , Disease Models, Animal , Amygdala/chemistry , Amygdala/metabolism , Animals , Anxiety/chemically induced , Anxiety/diagnosis , Behavior, Animal , Biomarkers/analysis , Biomarkers/metabolism , Blood Proteins/analysis , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Lactoylglutathione Lyase/metabolism , Male , Mice , Phosphopyruvate Hydratase/metabolism
18.
J Neurosci ; 25(17): 4375-84, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15858064

ABSTRACT

For >15 generations, CD1 mice have been selectively and bidirectionally bred for either high-anxiety-related behavior (HAB-M) or low-anxiety-related behavior (LAB-M) on the elevated plus-maze. Independent of gender, HAB-M were more anxious than LAB-M animals in a variety of additional tests, including those reflecting risk assessment behaviors and ultrasound vocalization, with unselected CD1 "normal" control (NAB-M) and cross-mated (CM-M) mice displaying intermediate behavioral scores in most cases. Furthermore, in both the forced-swim and tail-suspension tests, LAB-M animals showed lower scores of immobility than did HAB-M and NAB-M animals, indicative of a reduced depression-like behavior. Using proteomic and microarray analyses, glyoxalase-I was identified as a protein marker, which is consistently expressed to a higher extent in LAB-M than in HAB-M mice in several brain areas. The same phenotype-dependent difference was found in red blood cells with NAB-M and CM-M animals showing intermediate expression profiles of glyoxalase-I. Additional studies will examine whether glyoxalase-I has an impact beyond that of a biomarker to predict the genetic predisposition to anxiety- and depression-like behavior.


Subject(s)
Anxiety Disorders/enzymology , Lactoylglutathione Lyase/metabolism , Analysis of Variance , Animals , Animals, Newborn , Anti-Anxiety Agents/administration & dosage , Anxiety Disorders/diagnosis , Anxiety Disorders/drug therapy , Anxiety Disorders/genetics , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal , Biomarkers/metabolism , Blotting, Western/methods , Brain/drug effects , Brain/metabolism , Breeding/methods , Diazepam/administration & dosage , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional/methods , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Hindlimb Suspension/physiology , Lactoylglutathione Lyase/isolation & purification , Locomotion/genetics , Male , Mass Spectrometry/methods , Mice , Microarray Analysis/methods , Phenotype , Predictive Value of Tests , Proteomics/methods , Reaction Time/physiology , Reproducibility of Results , Sex Factors , Spatial Behavior/drug effects , Spatial Behavior/physiology , Statistics as Topic , Swimming , Time Factors , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
19.
J Neurosci ; 24(35): 7762-70, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15342744

ABSTRACT

Two inbred rat lines have been developed that show either high (HAB) or low (LAB) anxiety-related behavior. The behavioral phenotype correlates with arginine vasopressin (AVP) expression at the level of the hypothalamic paraventricular nucleus (PVN), but not supraoptic nucleus, with HAB animals overexpressing the neuropeptide in both magnocellular and parvocellular subdivisions of the PVN. We detected a number of single nucleotide polymorphisms (SNPs) in the AVP locus that differ between the HAB and LAB animals, two of which were embedded in cis-regulatory elements. The HAB-specific allele of the AVP gene promoter occurs in 1.5% of outbred Wistar rats and is more transcriptionally active in vivo, as revealed by allele-specific transcription studies in cross-mated HAB/LAB F1 animals. Interestingly, one specific SNP [A(-1276)G] conferred reduced binding of the transcriptional repressor CArG binding factor A (CBF-A) in the HAB allele, the consequent differential regulation of the AVP promoter resulting in an overexpression of AVP in vitro and in vivo. Furthermore, CBF-A is highly coexpressed in AVP-containing neurons of the PVN supporting an important role for regulation of AVP gene expression in vivo. Taken together, our results demonstrate a role for an AVP gene polymorphism and CBF-A in elevated AVP expression in the PVN of HAB rats likely to contribute to their behavioral and neuroendocrine phenotype.


Subject(s)
Anxiety/genetics , Arginine Vasopressin/physiology , DNA-Binding Proteins/physiology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Repressor Proteins/physiology , Alleles , Animals , Arginine Vasopressin/biosynthesis , Arginine Vasopressin/genetics , Bone Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Exploratory Behavior , Fear , Gene Expression Regulation/genetics , Humans , Inbreeding , Maze Learning , Osteosarcoma/pathology , Oxytocin/biosynthesis , Oxytocin/genetics , Protein Binding , Protein Interaction Mapping , Rats , Rats, Wistar , Reaction Time , Recombinant Fusion Proteins/biosynthesis , Ribonucleoproteins , Transcription Factors , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...