Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 183(1): 18-24, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17599477

ABSTRACT

Complex interactions between pain perception, anxiety and depressive symptoms have repeatedly been described. However, pathophysiological or biochemical mechanisms underlying the alterations of pain perception in patients suffering from anxiety or depression still remain a matter of debate. Thus, we aimed to perform an investigation on pain perception in an animal model of extremes in anxiety-related behaviour, which might provide a tool for future studies. Here, thermal pain thresholds were obtained from rats with a genetic predisposition to high anxiety-related behaviour (HAB), including signs of comorbid depression-like behaviour and from controls (low-anxiety rats (LAB); cross-bred HAB and LAB rats; Wistar rats). Furthermore, the effect of eight-week antidepressive treatment using citalopram and of short-term anxiolytic treatment with diazepam on pain-related behaviour was assessed. Simultaneously, anxiety-related behaviour was monitored. At baseline, HAB animals showed 35% higher thresholds for thermal pain than controls. These were normalized to control levels after eight weeks of continuous citalopram treatment paralleled by a reduction of anxiety-related behaviour, but also acutely after diazepam administration. Overall, thermal pain thresholds in HAB animals are shifted in a similar fashion as seen in patients suffering from major depressive disorder. Antidepressive, as well as anxiolytic treatments, attenuated these differences. As the relative importance of the factors anxiety and depression cannot be derived from this study with certainty, extending these investigations to additional animal models might represent a valuable tool for future investigations concerning the interrelations between anxiety, depression, and pain at a molecular level.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents, Second-Generation/pharmacology , Anxiety/drug therapy , Citalopram/pharmacology , Diazepam/pharmacology , Pain Threshold/drug effects , Analysis of Variance , Animals , Anxiety/genetics , Crosses, Genetic , Disease Models, Animal , Hot Temperature , Male , Rats , Rats, Inbred Strains , Rats, Wistar , Reaction Time/drug effects , Species Specificity , Statistics, Nonparametric
2.
Biol Psychiatry ; 61(6): 782-96, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17224133

ABSTRACT

BACKGROUND: The enhanced depression-like behavior in the forced swim test displayed by rats selectively bred for high anxiety-related behavior (HAB) as compared with their low anxiety counterparts (LAB) is abolished by chronic paroxetine treatment. The aim of the present study was to identify neuronal substrates underlying this treatment response in HABs. METHODS: The HAB rats received paroxetine (10 mg/kg/day) for 24 days via drinking water, and drug-induced modulation of neuronal activation patterns in response to forced swimming was mapped with the expression of the immediate early gene c-Fos as marker. RESULTS: Chronic paroxetine treatment reduced the immobility scores during forced swimming, confirming the previously observed antidepressant-like effect in these animals, and attenuated the forced swim-induced c-Fos response in a restricted set (11 of 70) of brain areas. These included limbic areas such as the prelimbic cortex, parts of the amygdala, the bed nucleus of the stria terminalis, dorsal hippocampus, dorsal lateral septum as well as hypothalamic and hindbrain areas (dorsolateral periaqueductal gray [PAG], locus coeruleus). Untreated LAB rats, which displayed low depression-like behavior comparable to that of treated HABs, also showed low swim stress-induced c-Fos response in most of these same areas, further supporting an association of attenuated neuronal excitability in the identified areas with attenuated depression-like behavior. CONCLUSIONS: These findings indicate that modulation of neuronal activation in a restricted set of defined, mainly limbic as well as selected hypothalamic and hindbrain areas by paroxetine treatment is associated with the reduction of enhanced depression-like behavior in a psychopathological animal model.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Anxiety/metabolism , Brain/metabolism , Depression/metabolism , Paroxetine/pharmacology , Stress, Psychological/metabolism , Analysis of Variance , Animals , Anxiety/complications , Anxiety/drug therapy , Brain/drug effects , Brain/physiopathology , Depression/etiology , Depression/prevention & control , Immobility Response, Tonic/drug effects , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Statistics, Nonparametric , Stress, Psychological/complications , Stress, Psychological/drug therapy
3.
Neuropharmacology ; 50(8): 1048-58, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16620881

ABSTRACT

We reported recently that two rat lines bred for either high (HAB) or low (LAB) anxiety-related behavior display differential Fos expression in restricted parts of the fear/anxiety circuitry when exposed to mild anxiety evoked in exploratory anxiety tests. Since different forms of anxiety are thought to activate different parts of the anxiety circuitry, we investigated now whether (1) an aversive stimulus which elicits escape behavior (airjet) and (2) the anxiogenic/panicogenic drug FG-7142 would reveal further differences in Fos expression as a marker of neuronal activation between HAB and LAB rats. Both airjet exposure and FG-7142 induced Fos expression in both lines in various anxiety-related brain areas. HAB rats, which displayed exaggerated escape responses during airjet exposure, exhibited increased Fos expression in brain areas including the hypothalamus, periaqueductal gray and locus coeruleus, as well as blunted Fos activation in the cingulate cortex in response to airjet and/or FG-7142. The results corroborate previous findings showing that trait anxiety affects neuronal excitability in hypothalamic and medial prefrontal areas. Furthermore, by using airjet as well as FG-7142, we now reveal that enhanced trait anxiety is also associated with neuronal hyperexcitability in the locus coeruleus and the periaqueductal gray, suggesting that investigation of an array of different anxiogenic stimuli is important for the detection of altered neuronal processing in trait anxiety.


Subject(s)
Air , Anxiety , Carbolines/administration & dosage , GABA Antagonists/administration & dosage , Gene Expression/drug effects , Oncogene Proteins v-fos/metabolism , Animals , Anxiety/etiology , Anxiety/genetics , Anxiety/metabolism , Behavior, Animal , Brain/drug effects , Brain/metabolism , Brain/pathology , Dopamine beta-Hydroxylase/metabolism , Escape Reaction/drug effects , Immunohistochemistry/methods , Male , Rats , Rats, Wistar , Statistics, Nonparametric
4.
Behav Neurosci ; 120(1): 60-71, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16492117

ABSTRACT

Genetic background may influence an individual's susceptibility to, and subsequent coping strategy for, an acute stressor. When exposed to social defeat (SD), rats bred for high (HAB) or low (LAB) trait anxiety, which also differ in depression-like behavior, showed highly divergent passive and active coping behaviors, respectively. HABs spent more time freezing and emitted more ultrasound vocalization calls during SD than LABs, which spent more time rearing and grooming. Although the behavioral data confirmed the prediction that heightened trait anxiety would make rats more prone to experience stress, adrenocorticotropin and corticosterone were secreted to a higher extent in LABs than in HABs. In the latter, Fos expression upon SD was enhanced in the amygdala and hypothalamic areas compared with LABs, whereas it was diminished in prefrontal and brainstem areas.


Subject(s)
Adaptation, Psychological , Adrenocorticotropic Hormone/metabolism , Anxiety/genetics , Corticosterone/metabolism , Genes, fos/genetics , Neurons/physiology , Social Behavior , Adrenocorticotropic Hormone/blood , Amygdala/metabolism , Animals , Brain Stem/metabolism , Corticosterone/blood , Genetic Predisposition to Disease , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Pituitary-Adrenal System/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Vocalization, Animal
5.
Neuropsychopharmacology ; 31(5): 925-32, 2006 May.
Article in English | MEDLINE | ID: mdl-16192979

ABSTRACT

In depressed patients as well as healthy controls, a positive relationship between hippocampal volume and trait anxiety has been reported. This study sought to explore the possible inter-relation between hippocampal volume and trait anxiety further. Magnetic resonance imaging at 7 T was used to measure hippocampal volumes in a rat model of extremes in trait anxiety (experiment 1) and in a Wistar population with normal anxiety-related behavior (experiment 2). In addition to anxiety-related behavior, potentially confounding factors (depression-like, exploratory, and locomotor behavior) were assessed. Experiment 1 globally supported the hypothesis of a positive relationship between hippocampus volume and trait anxiety but did not allow for ruling out possible confounds arising from cosegregation of other behavioral traits. Experiment 2 yielded strong evidence for a negative relationship which was specific for trait anxiety. Thus, the relationship between hippocampal volume and anxiety may be more complex than expected. Interestingly, anxiety-related behavior in experiment 2 had a stronger influence on hippocampal volume than depression-like behavior. In the light of hippocampal volume loss in anxiety disorder and frequent comorbidity of anxiety and depression, this finding suggests that further research into the relationship between anxiety and hippocampal volume may be critical for understanding hippocampal contributions to normal and pathological behavior.


Subject(s)
Anxiety Disorders/diagnosis , Anxiety Disorders/physiopathology , Atrophy/diagnosis , Atrophy/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Animals , Anxiety Disorders/complications , Atrophy/etiology , Depressive Disorder/complications , Depressive Disorder/diagnosis , Depressive Disorder/physiopathology , Disease Models, Animal , Magnetic Resonance Imaging , Male , Neural Pathways/pathology , Neural Pathways/physiopathology , Rats
6.
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
7.
Neuroimage ; 23(1): 382-91, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15325386

ABSTRACT

The neural basis of trait anxiety is poorly understood. In genetically selected hyperanxious (high anxiety-related behavior; HAB) rats, diazepam induces a stronger anxiolytic response than in hypoanxious (low anxiety-related behavior; LAB) rats. A screen for neuronal response differences to diazepam between HAB and LAB rats using pharmacologic fMRI (phMRI) at 7 T revealed a blunted diazepam-induced neuronal deactivation in the dorsomedial prefrontal cortex (dmPFC) of HABs. This was not due to reduced benzodiazepine (BDZ) receptor densities in this region. Instead, dmPFC tissue oxygenation at baseline was found to be significantly lower in HABs. This suggests a tonic relative hypoactivity under the highly stressful phMRI conditions, offering an explanation for the reduced responsivity to the neural depressant effect of diazepam in the sense of a floor effect. Subsequently, Fos immunoreactivity (Fos-IR) showed that ethologically relevant stressors also cause less dmPFC activation in HABs. In the context of an anxiety-inhibiting role of the dmPFC, we propose that failure to sufficiently activate this region in stressful situations may contribute to high trait anxiety.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/physiopathology , Arousal/physiology , Diazepam/pharmacology , Fear/physiology , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Oxygen/blood , Prefrontal Cortex/physiology , Proto-Oncogene Proteins c-fos/analysis , Stress, Psychological/complications , Animals , Arousal/drug effects , Autoradiography , Fear/drug effects , Male , Neural Inhibition/drug effects , Neural Inhibition/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Rats , Rats, Inbred Strains , Receptors, Neurotransmitter/drug effects , Receptors, Neurotransmitter/physiology , Stress, Psychological/pathology , Stress, Psychological/physiopathology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
8.
Biol Psychiatry ; 55(7): 715-23, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15039000

ABSTRACT

BACKGROUND: Two Wistar rat lines selectively bred for either high (HAB) or low (LAB) anxiety-related behavior were used to identify neurobiological correlates of trait anxiety. METHODS: We used Fos expression for mapping of neuronal activation patterns in response to mild anxiety-provoking challenges. RESULTS: In both lines, exposure to an open field (OF) or the open arm (OA) of an elevated plus-maze induced Fos expression in several brain areas of the anxiety/fear circuitry. Rats of the HAB type, which showed signs of a hyperanxious phenotype and a hyperreactive hypothalamic-pituitary-adrenal axis compared with LAB rats, exhibited a higher number of Fos-positive cells in the paraventricular nucleus of the hypothalamus, the lateral and anterior hypothalamic area, and the medial preoptic area in response to both OA and OF. Less Fos expression was induced in the cingulate cortex in HAB than in LAB rats. Differential Fos expression in response to either OA or OF was observed in few brain regions, including the thalamus and hippocampus. CONCLUSIONS: The present data indicate that the divergent anxiety-related behavioral response of HAB versus LAB rats to OF and OA exposures is associated with differential neuronal activation in restricted parts of the anxiety/fear circuitry. Distinct hypothalamic regions displayed hyperexcitability, and the cingulate cortex showed hypoexcitability, which suggests that they are main candidate mediators of dysfunctional brain activation in pathologic anxiety.


Subject(s)
Anxiety/genetics , Arousal/genetics , Brain/metabolism , Fear/physiology , Proto-Oncogene Proteins c-fos/genetics , Adrenocorticotropic Hormone/blood , Animals , Anterior Hypothalamic Nucleus/metabolism , Anxiety/psychology , Brain Mapping , Gene Expression/physiology , Gyrus Cinguli/metabolism , Hippocampus/metabolism , Hypothalamic Area, Lateral/metabolism , Hypothalamo-Hypophyseal System/metabolism , Immunoenzyme Techniques , Male , Nerve Net/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Phenotype , Preoptic Area/metabolism , Rats , Rats, Wistar , Selection, Genetic , Thalamus/metabolism
9.
Neuropsychopharmacology ; 29(1): 1-14, 2004 Jan.
Article in English | MEDLINE | ID: mdl-12942143

ABSTRACT

To model aspects of trait anxiety/depression, Wistar rats were bred for extremes in either hyper (HAB)- or hypo(LAB)-anxiety as measured on the elevated plus-maze and in a variety of additional behavioral tests. Similar to psychiatric patients, HAB rats prefer passive stress-coping strategies, indicative of depression-like behavior, show hyper-reactivity of the hypothalamo-pituitary-adrenal axis, and a pathological response to the dexamethasone/corticotropin-releasing hormone (CRH) challenge test. Here we tested central mRNA expression, release patterns, and receptor binding of neuropeptides critically involved in the regulation of both anxiety-related behavior and the HPA axis. Thus, CRH, arginine-8-vasopressin (AVP), and oxytocin (OXT) were studied in brains of HAB and LAB males both under basal conditions and after exposure to a mild emotional stressor. In HAB rats, CRH mRNA was decreased in the bed nucleus of the stria terminalis only. While no significant difference in CRH1-receptor binding was found in any brain area, CRH2-receptor binding was elevated in the hypothalamic paraventricular nucleus (PVN), the ventromedial hypothalamus, and the central amygdala of HABs compared to LABs. AVP, but not OXT, mRNA expression as well as release of the neuropeptide, were higher in the PVN of HABs, whereas AVP V1a-receptor binding failed to show significant differences in any brain region studied. Remarkably, intra-PVN treatment of HABs with the AVP V1-receptor antagonist d (CH(2))(5) Tyr (Me) AVP resulted in a decrease in anxiety/depression-related behavior. The elevated expression and release of AVP within the PVN of HAB rats together with the behavioral effects of the AVP V1-receptor antagonist suggest a critical involvement of this neuropeptide in neuroendocrine and behavioral phenomena associated with trait anxiety/depression.


Subject(s)
Anxiety/metabolism , Central Nervous System/metabolism , Neuropeptides/metabolism , Receptors, Neuropeptide/metabolism , Vasopressins/metabolism , Analysis of Variance , Animals , Antidiuretic Hormone Receptor Antagonists , Anxiety/genetics , Autoradiography/methods , Behavior, Animal , Binding Sites , Breeding , Central Nervous System/anatomy & histology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Disease Models, Animal , Exploratory Behavior , Gene Expression , Genetics, Behavioral , In Situ Hybridization , Male , Maze Learning , Microdialysis/methods , Neuropeptides/genetics , Oxytocin/genetics , Oxytocin/metabolism , Protein Binding , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains/genetics , Rats, Wistar , Reaction Time , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Neuropeptide/genetics , Stress, Physiological/metabolism , Swimming , Time Factors , Vasopressins/genetics , Vasopressins/physiology
10.
Eur J Neurosci ; 18(2): 403-11, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12887422

ABSTRACT

This study explores the effects of enhancing vasopressin V1a receptor expression in the septum using viral vector-mediated gene transfer on social discrimination and social interactions. Bilateral infusion of an adeno-associated viral vector containing the prairie vole V1a receptor gene (V1aR-AAV) regulated by a neuron-specific enolase promoter resulted in a stable increase in V1a receptor binding density in the rat septum without affecting oxytocin receptor density. Control animals were infused with a vector expressing the lacZ gene. In a social discrimination paradigm, only V1aR-AAV-treated animals succeeded in discriminating a previously encountered from a novel juvenile after an interexposure interval (IEI) of more than 2 h, demonstrating the functional incorporation of the vole V1a receptor in the rat septal circuits underlying short-term memory processes. Microdialysis administration of synthetic vasopressin during the first juvenile exposure, used to mimic intraseptal release patterns of the neuropeptide, produced similar prolongations in recognition (up to an IEI of 24 h) in both V1aR-AAV and control animals. Septal microdialysis administration of a selective V1a, but not oxytocin, receptor antagonist in both groups prevented discrimination even after an IEI of as short as 0.5 h, confirming the specificity of the vole V1a receptor involvement in social discrimination abilities. In addition, active social interactions were found to be increased among V1aR-AAV rats compared to controls. Viral vector-mediated gene transfer provides a valuable tool for studies on the role of localized gene expression on behavioural parameters.


Subject(s)
Gene Transfer Techniques , Receptors, Vasopressin/genetics , Septum of Brain/physiology , Social Behavior , Animals , Antidiuretic Hormone Receptor Antagonists , Arvicolinae , Autoradiography , Dependovirus/genetics , Discrimination, Psychological/drug effects , Genetic Vectors , Lac Operon , Male , Microdialysis , Rats , Rats, Wistar , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacology , Vasopressins/administration & dosage , Vasopressins/pharmacology
11.
Neuropsychopharmacology ; 28(2): 235-43, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589376

ABSTRACT

The neuroendocrine and behavioral effects of chronic paroxetine treatment were investigated in two rat lines selectively bred for high anxiety-related behavior (HAB) or low anxiety-related behavior (LAB) emotionality. In addition to a characteristic behavioral phenotype with markedly passive stress-coping strategies, HAB rats show a hypothalamic vasopressinergic hyperdrive that is causally related to hypothalamic-pituitary-adrenocortical dysregulation as demonstrated in the combined dexamethasone (DEX)/corticotropin-releasing hormone (CRH) test. A total of 8 weeks of chronic paroxetine treatment induced a more active coping strategy in the forced swim test in HAB rats only. In contrast, paroxetine-treated LAB rats did not change their swimming behavior. To investigate the neuroendocrine alterations linked to these behavioral changes, a combined DEX/CRH test was performed. In HAB rats, the paroxetine-induced behavioral changes towards more active coping strategies were accompanied by a normalization of the CRH-stimulated increase in corticotropin (ACTH) and corticosterone secretion. Concomitantly, the hypothalamic vasopressinergic hyperdrive was found to be reduced in HAB but not LAB rats, as indicated by a decrease in vasopressin mRNA expression, whereas vasopressin 1a receptor binding was unaffected. These findings provide the first evidence that the vasopressinergic system is likely to be critically involved in the behavioral and neuroendocrine effects of antidepressant drugs. This novel mechanism of action of paroxetine on vasopressin gene regulation renders vasopressinergic neuronal circuits a promising target for the development of more causal antidepressant treatment strategies.


Subject(s)
Anxiety Disorders/metabolism , Disease Models, Animal , Hypothalamus/drug effects , Neurosecretory Systems/drug effects , Paroxetine/administration & dosage , Vasopressins/biosynthesis , Animals , Anxiety Disorders/drug therapy , Hypothalamus/metabolism , Male , Neurosecretory Systems/metabolism , RNA, Messenger/biosynthesis , Rats , Species Specificity , Vasopressins/antagonists & inhibitors
12.
Behav Genet ; 32(5): 301-14, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12405513

ABSTRACT

In addition to their robust difference in trait anxiety, as illustrated by a variety of behavioral tests, HAB and LAB rats differ in their stress coping strategies, the former being more susceptible and vulnerable to stressor exposure and preferring more passive strategies. HAB rats of either gender show signs of a hyper-reactive hypothalamic-pituitary-adrenocortical (HPA) axis, thus resembling psychiatric patients. As shown by in situ hybridization and microdialysis in freely behaving animals, both the expression and release of vasopressin in the hypothalamic paraventricular nucleus are higher in HAB than in LAB rats, thus contributing to the HPA axis hyperdrive. Accordingly, in HAB animals, administration of a V1 receptor antagonist normalized the pathological outcome of the dexamethasone/corticotropin-releasing hormone test and triggered behavioral changes toward reduced anxiety and active stress coping. Pharmacological validation has revealed signs of depressive-like behavior, as HAB but not LAB rats have shown more active stress coping behavior and a normalized HPA axis after treatment with paroxetine. Of interest, this antidepressant reduced the hypothalamic overexpression of vasopressin; this novel mechanism of action is likely to contribute to paroxetine effects on both behavioral and neuroendocrine parameters. Cross-mating and cross-fostering paradigms showed that the divergent emotionality in HAB vs. LAB rats is determined genetically, rather than postnatally through maternal behavior. As the behavioral and neuroendocrine phenotyping pointed to the vasopressin gene as a candidate gene critically involved in anxiety, preliminary genetic approaches have been focused on this gene, revealing single nucleotide polymorphisms (SNPs) in the promotor area of the vasopressin gene in HAB, but not LAB rats. HAB/LAB rats are thus proving to be a unique animal model to identify and characterize neurobiological, neuroendocrine, and genetic correlates of trait anxiety, and perhaps depression, in humans.


Subject(s)
Anxiety/genetics , Genetics, Behavioral , Animals , Anxiety/physiopathology , Anxiety/psychology , Behavior, Animal/physiology , Brain/metabolism , Cognition/physiology , Gene Expression , Models, Animal , Neurosecretory Systems/physiology , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Rats , Rats, Inbred Strains , Rats, Wistar , Species Specificity , Stress, Physiological/physiopathology
13.
Behav Brain Res ; 136(1): 227-37, 2002 Oct 17.
Article in English | MEDLINE | ID: mdl-12385809

ABSTRACT

The reliability of behavioural data constitutes a major concern in the neuroscience field. Indeed, discrepancies in the behavioural patterns of mice or rats in the same anxiety tests performed in different laboratories have been reported recently. The question raised by such data addressed, in particular, the selection and breeding of two lines of rats on the basis of their high (HAB) and low (LAB) anxiety-related behaviour in the elevated plus-maze test at the Max Planck Institute of Psychiatry in Munich (Germany). As the majority of the behavioural data in these animals has been derived from research carried out in this institute, the aims of the present study were: (1) to test the reliability of the differences in anxiety-related behaviour of these rats in two other laboratories (Villeneuve d'Ascq, France and Innsbruck, Austria); and (2) to determine how the different behavioural traits were associated in both HAB and LAB rats by a principal component analysis. Results were in agreement with the studies performed in Munich, as the divergence in anxiety-related behaviour of the two lines was highly consistent in all tests performed in Villeneuve d'Ascq and Innsbruck. Moreover, the most important parameters to discriminate the two lines were similar to those found in a previous study. Finally, the principal component analysis again confirmed that the selection of HAB and LAB rats is based on anxiety-related behaviour rather than locomotor activity.


Subject(s)
Anxiety/psychology , Behavior, Animal/physiology , Environment , Animals , Data Interpretation, Statistical , Discriminant Analysis , Male , Mice , Motor Activity/physiology , Multivariate Analysis , Rats , Rats, Wistar , Reproducibility of Results , Swimming/psychology
14.
J Psychiatr Res ; 36(4): 197-208, 2002.
Article in English | MEDLINE | ID: mdl-12191624

ABSTRACT

Excessive corticotropin-releasing hormone (CRH) secretion in limbic and prefrontal brain areas has been postulated to underly stress-related clinical conditions. Studies in mice with deleted or pharmacologically compromised CRH type 1 receptors (CRH-R1) point to a key role of the CRH/CRH-R1 signaling cascade as a potential drug target. Therefore, we compared the effect of a selective high affinity CRH-R1 antagonist (R121919) on sleep-wake behavior in two rat lines selectively bred for either high or low innate anxiety. We found that the subcutaneous injection of the solvent of R121919, a citrate buffer solution, transiently increased circulating levels of the stress hormones ACTH and corticosterone and reduced sleep, especially in high-anxiety animals. When R121919 was added to the solvent, hormone levels and sleep patterns returned to baseline and were indistinguishable between the rat lines. This finding is in accord with previous observations from a clinical trial in depressed patients and studies in rats with high innate anxiety that suggested major effects of CRH-R1 antagonism in the presence of a pathological (i.e. CRH hypersecretion) condition only.


Subject(s)
Arousal/drug effects , Pyrimidines/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Sleep Stages/drug effects , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Hypothalamo-Hypophyseal System/drug effects , Limbic System/drug effects , Male , Pituitary-Adrenal System/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Wakefulness/drug effects
15.
Neuropsychopharmacology ; 26(6): 729-36, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12007743

ABSTRACT

According to the tension reduction hypothesis, individuals with an elevated anxiety level may be more sensitive to the anxiolytic effects of alcohol and may, therefore, have a higher predisposition to consume alcohol. To examine this hypothesis, we studied the drinking behavior as well as the sensitivity to the anxiolytic effect of alcohol in two rat lines that were bred and selected for differences in anxiety-related behavior on the elevated plus-maze: the extremely anxious HAB (high anxiety-related behavior) and the non-anxious LAB (low anxiety-related behavior) lines. Alcohol self-administration and the occurrence of an alcohol deprivation effect were studied in female and male HAB and LAB rats in a free-choice, 4-bottle home cage paradigm. The sensitivity of HAB and LAB rats to the anxiolytic effect of alcohol was assessed by testing their behavior on the elevated plus-maze after an acute application of ethanol. During the first days of voluntary ethanol drinking, the ethanol intake and preference of female LABs was significantly higher than that of female HABs. Although not statistically significant, the same trend could be seen in male LABs. Moreover, male as well as female LAB but not HAB rats showed a significant alcohol deprivation effect after abstinence. There were no differences when saccharin was presented to naive animals, indicating that the different ethanol drinking behavior of HAB and LAB rats does not represent a general difference in the consumption of new liquids. Application of ethanol resulted in an anxiolytic effect in HAB but not in LAB rats on the elevated plus-maze. In summary, increased inborn anxiety and voluntary ethanol consumption of HAB and LAB rats were correlated to some extent; however, this relationship was a negative one. It is concluded that, although such a relationship might exist in some individuals, increased levels of inborn anxiety and alcohol consumption are not necessarily related.


Subject(s)
Alcohol Drinking/genetics , Alcohol Drinking/psychology , Anxiety/genetics , Anxiety/psychology , Animals , Breeding/methods , Female , Male , Maze Learning/drug effects , Rats , Rats, Wistar , Self Administration/methods , Self Administration/psychology , Species Specificity
16.
Neuropsychopharmacology ; 26(1): 94-105, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11751036

ABSTRACT

To investigate the neuroendocrine alterations linked to inborn emotionality in two Wistar rat lines selectively bred for either high (HAB) or low (LAB) anxiety-related behavior, we administered the combined dexamethasone (DEX)/corticotropin-releasing hormone (CRH) test. DEX (12:00 M. (noon); 30 microg/kg) resulted in a significantly less efficient suppression of the diurnal increase in the circulating corticotropin (ACTH) levels in the male HAB rats than in the male LAB rats. In addition, plasma ACTH and corticosterone responses to subsequent CRH (7:30 P.M.; 50 ng/kg) were significantly higher in male HAB rats. The rise in ACTH after CRH in the DEX-pretreated male HAB rats points toward an enhanced activity and involvement of endogenous vasopressin synthesized in the hypothalamic paraventricular nucleus (PVN) and acting at pituitary corticotrope cells. We tested this hypothesis by in situ hybridization and in vivo microdialysis, and found an increase in both basal synthesis and release of vasopressin within the PVN of the male HAB rats. As expected, pretreatment with a selective vasopressin type 1 receptor antagonist abolished the CRH-stimulated increase in ACTH secretion in the DEX-pretreated male HAB rats. The results indicate that vasopressin-mediated effects are critically involved in the profound disturbance of the hypothalamic-pituitary-adrenocortical system in male HAB rats, thus revealing striking parallels to the neuroendocrine situation in human depression.


Subject(s)
Anxiety/psychology , Corticotropin-Releasing Hormone , Dexamethasone , Glucocorticoids , Mood Disorders/physiopathology , Vasopressins/physiology , Adrenalectomy , Adrenocorticotropic Hormone/pharmacology , Animals , Antidiuretic Hormone Receptor Antagonists , Anxiety/genetics , Binding Sites , Corticosterone/blood , Feedback/physiology , Immunohistochemistry , In Situ Hybridization , Male , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/biosynthesis , Radioimmunoassay , Rats , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/metabolism , Sex Characteristics
SELECTION OF CITATIONS
SEARCH DETAIL
...