Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Mol Psychiatry ; 23(3): 533-543, 2018 03.
Article in English | MEDLINE | ID: mdl-28696432

ABSTRACT

Single-nucleotide polymorphisms (SNPs) in CACNA1C, the α1C subunit of the voltage-gated L-type calcium channel Cav1.2, rank among the most consistent and replicable genetics findings in psychiatry and have been associated with schizophrenia, bipolar disorder and major depression. However, genetic variants of complex diseases often only confer a marginal increase in disease risk, which is additionally influenced by the environment. Here we show that embryonic deletion of Cacna1c in forebrain glutamatergic neurons promotes the manifestation of endophenotypes related to psychiatric disorders including cognitive decline, impaired synaptic plasticity, reduced sociability, hyperactivity and increased anxiety. Additional analyses revealed that depletion of Cacna1c during embryonic development also increases the susceptibility to chronic stress, which suggest that Cav1.2 interacts with the environment to shape disease vulnerability. Remarkably, this was not observed when Cacna1c was deleted in glutamatergic neurons during adulthood, where the later deletion even improved cognitive flexibility, strengthened synaptic plasticity and induced stress resilience. In a parallel gene × environment design in humans, we additionally demonstrate that SNPs in CACNA1C significantly interact with adverse life events to alter the risk to develop symptoms of psychiatric disorders. Overall, our results further validate Cacna1c as a cross-disorder risk gene in mice and humans, and additionally suggest a differential role for Cav1.2 during development and adulthood in shaping cognition, sociability, emotional behavior and stress susceptibility. This may prompt the consideration for pharmacological manipulation of Cav1.2 in neuropsychiatric disorders with developmental and/or stress-related origins.


Subject(s)
Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/physiology , Mental Disorders/genetics , Adult , Black or African American , Animals , Bipolar Disorder/genetics , Calcium Channels/genetics , Depressive Disorder, Major/genetics , Disease Models, Animal , Female , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Humans , Male , Mice/embryology , Mice, Transgenic/genetics , Neurons/metabolism , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics
2.
Hippocampus ; 27(1): 28-35, 2017 01.
Article in English | MEDLINE | ID: mdl-27699923

ABSTRACT

Even in the absence of neurodegenerative diseases, progressing age often coincides with cognitive decline and morphological changes. However, longitudinal studies that directly link these two processes are missing. In this proof-of-concept study we therefore performed repeated within-subject testing of healthy male R26R mice in a spatial learning task in combination with manganese-enhanced volumetric MRI analyses at the ages of 8, 16, and 24 months. We grouped the mice into good and poor performers (n = 6, each), based on their spatial learning abilities at the age of 24 months. Using this stratification, we failed to detect a priori volume differences, but observed a significant decrease in total hippocampal volume over time for both groups. Interestingly, this volume decrease was specific for the dorsal hippocampus and significantly accelerated in poor performers between 16 and 24 months of age. This is the first time that individual changes in hippocampal volume were traced alongside cognitive performance within the same subjects over 1½ years. Our study points to a causal link between volume loss of the dorsal hippocampus and cognitive impairments. In addition, it suggests accelerated degenerative processes rather than a priori volume differences as determining trajectories of age-related cognitive decline. Despite the relatively small sample sizes, the strong behavioral and moderate morphological alterations demonstrate the general feasibility of longitudinal studies of age-related decline in cognition and hippocampus integrity. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cognitive Aging , Cognitive Dysfunction/diagnostic imaging , Hippocampus/diagnostic imaging , Animals , Cognitive Dysfunction/etiology , Contrast Media , Longitudinal Studies , Magnetic Resonance Imaging , Male , Manganese , Maze Learning , Mice, Transgenic , Neuropsychological Tests , Organ Size , Proof of Concept Study , Spatial Memory
3.
Hippocampus ; 26(10): 1250-64, 2016 10.
Article in English | MEDLINE | ID: mdl-27101945

ABSTRACT

Expression of the lacZ-sequence is a widely used reporter-tool to assess the transgenic and/or transfection efficacy of a target gene in mice. Once activated, lacZ is permanently expressed. However, protein accumulation is one of the hallmarks of neurodegenerative diseases. Furthermore, the protein product of the bacterial lacZ gene is ß-galactosidase, an analog to the mammalian senescence-associated ß-galactosidase, a molecular marker for aging. Therefore we studied the behavioral, structural and molecular consequences of lacZ expression in distinct neuronal sub-populations. lacZ expression in cortical glutamatergic neurons resulted in severe impairments in hippocampus-dependent memory accompanied by marked structural alterations throughout the CNS. In contrast, GFP expression or the expression of the ChR2/YFP fusion product in the same cell populations did not result in either cognitive or structural deficits. GABAergic lacZ expression caused significantly decreased hyper-arousal and mild cognitive deficits. Attenuated structural and behavioral consequences of lacZ expression could also be induced in adulthood, and lacZ transfection in neuronal cell cultures significantly decreased their viability. Our findings provide a strong caveat against the use of lacZ reporter mice for phenotyping studies and point to a particular sensitivity of the hippocampus formation to detrimental consequences of lacZ expression. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hippocampus/metabolism , Lac Operon , Memory/physiology , Neurons/metabolism , beta-Galactosidase/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Survival/physiology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Gene Expression , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/diagnostic imaging , Hippocampus/pathology , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Recombinant Fusion Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
4.
Neurobiol Dis ; 77: 238-45, 2015 May.
Article in English | MEDLINE | ID: mdl-25796567

ABSTRACT

Endocannabinoids, including 2-arachidonoylglycerol (2-AG), activate presynaptic cannabinoid type 1 receptors (CB1R) on inhibitory and excitatory neurons, resulting in a decreased release of neurotransmitters. The event-specific activation of the endocannabinoid system by inhibition of the endocannabinoid degrading enzymes may offer a promising strategy to selectively activate CB1Rs at the site of excessive neuronal activation with the overall goal to prevent the development epilepsy. The aim of this study was to investigate the impact of monoacylglycerol lipase (MAGL) inhibition on the development and progression of epileptic seizures in the kindling model of temporal lobe epilepsy. Therefore, we selectively blocked MAGL by JZL184 (8mg/kg, i.p.) in mice to analyze the effects of increased 2-AG levels on kindling acquisition and to exclude an anticonvulsive potential. Our results showed that JZL184 treatment significantly delayed the development of generalized seizures (p=0.0066) and decreased seizure (p<0.0001) and afterdischarge duration (p<0.001) in the kindling model of temporal lobe epilepsy, but caused only modest effects in fully kindled mice. Moreover, we proved that JZL184 treatment had no effects in conditional CB1R knockout mice lacking expression of the receptor in principle neurons of the forebrain. In conclusion, the data demonstrate that indirect CB1R agonism delays the development of generalized epileptic seizures but has no relevant acute anticonvulsive effects. Furthermore, we confirmed that the effects of JZL184 on kindling progression are CB1R mediated. Thus, the data indicate that the endocannabinoid 2-AG might be a promising target for an anti-epileptogenic approach.


Subject(s)
Benzodioxoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Kindling, Neurologic/genetics , Monoacylglycerol Lipases/metabolism , Piperidines/therapeutic use , Receptor, Cannabinoid, CB1/metabolism , Amygdala/metabolism , Analysis of Variance , Animals , Disease Models, Animal , Epilepsy/etiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Reaction Time/drug effects , Receptor, Cannabinoid, CB1/genetics
5.
Neurobiol Dis ; 73: 334-47, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25123336

ABSTRACT

The endocannabinoid system serves as a retrograde negative feedback mechanism. It is thought to control neuronal activity in an epileptic neuronal network. The purpose of this study was to evaluate the impact of the endocannabinoid and endovanilloid systems on both epileptogenesis and ictogenesis. Therefore, we modulated the endocannabinoid and endovanilloid systems genetically and pharmacologically, and analyzed the subsequent impact on seizure progression in the kindling model of temporal lobe epilepsy in mice. In addition, the impact of seizures on associated cellular alterations was evaluated. Our principal results revealed that the endocannabinoid system affects seizure and afterdischarge duration dependent on the neuronal subpopulation being modulated. Genetic deletion of CB1-receptors (CB1Rs) from principal neurons of the forebrain and pharmacological antagonism with rimonabant (5 mg/kg) caused longer seizure duration. Deletion of CB1R from GABAergic forebrain neurons resulted in the opposite effect. Along with these findings, the CB1R density was elevated in animals with repetitively induced seizures. However, neither genetic nor pharmacological interventions had any impact on the development of generalized seizures. Other than CB1, genetic deletion or pharmacological blockade with SB366791 (1 mg/kg) of transient receptor potential vanilloid receptor 1 (TRPV1) had no effect on the duration of behavioral or electrographic seizure activity in the kindling model. In conclusion, we demonstrate that endocannabinoid, but not endovanilloid, signaling affects termination of seizure activity, without influencing seizure severity over time. These effects are dependent on the neuronal subpopulation. Thus, the data argue that the endocannabinoid system plays an active role in seizure termination but does not regulate epileptogenesis.


Subject(s)
Epilepsy/genetics , Epilepsy/pathology , Kindling, Neurologic/genetics , Neurons/metabolism , Prosencephalon/pathology , Receptor, Cannabinoid, CB1/deficiency , Anilides/pharmacology , Animals , Bromodeoxyuridine , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cannabinoid Receptor Antagonists/adverse effects , Cinnamates/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Knockout , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Piperidines/adverse effects , Pyrazoles/adverse effects , Rimonabant , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , gamma-Aminobutyric Acid/metabolism
6.
Eur J Neurosci ; 41(9): 1139-48, 2015 May.
Article in English | MEDLINE | ID: mdl-25720329

ABSTRACT

This study measured changes in brain extracellular norepinephrine (NE) and free corticosterone (CORT) levels in a mouse model of post-traumatic stress disorder and related them to hyperarousal and fear memory retention. To this end, microdialysis in the medial prefrontal cortex (mPFC) and the hippocampus (HPC) of male C57BL/6NCrl mice was performed during an acoustic startle response (ASR) and following an electric foot shock (FS), as well as during an ASR and recall of contextual fear (CF) 1 day later. Changes in ASR-stimulated NE levels in the mPFC corresponded to ASR 34 days after FS. Changes in basal and ASR-stimulated extracellular NE levels in the HPC, in contrast, were related to expression of early (day 2) and late (day 34) CF after FS. The increase in extracellular NE levels correlated in a U-shape manner with arousal levels and CF, thus suggesting a non-direct relationship. Stress of different modalities/strength (ASR, FS and CF) caused a similar relative increase in free CORT levels both in the mPFC and the HPC. One day after FS, ASR-induced increases in the CORT content in the mPFC tended to correlate with the FS-potentiated ASR in a U-shape manner. Taken together, these data show that the intracerebral increase in free CORT was likely related to an immediate response to stress, whereas NE neurotransmission in the forebrain predicted arousal and CF 1 month after trauma.


Subject(s)
Corticosterone/metabolism , Hippocampus/metabolism , Norepinephrine/metabolism , Prefrontal Cortex/metabolism , Stress Disorders, Post-Traumatic/metabolism , Animals , Arousal , Extracellular Space/metabolism , Fear , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Prefrontal Cortex/physiopathology , Reflex, Startle , Stress Disorders, Post-Traumatic/physiopathology
7.
NMR Biomed ; 26(5): 542-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23168745

ABSTRACT

Manganese-enhanced MRI has recently become a valuable tool for the assessment of in vivo functional cerebral activity in animal models. As a result of the toxicity of manganese at higher dosages, fractionated application schemes have been proposed to reduce the toxic side effects by using lower concentrations per injection. Here, we present data on regional-specific manganese accumulation during a fractionated application scheme over 8 days of 30 mg/kg MnCl2 , as well as on the clearance of manganese chloride over the course of several weeks after the termination of the whole application protocol supplying an accumulative dose of 240 mg/kg MnCl2 . Our data show most rapid accumulation in the superior and inferior colliculi, amygdala, bed nucleus of the stria terminalis, cornu ammonis of the hippocampus and globus pallidus. The data suggest that no ceiling effects occur in any region using the proposed application protocol. Therefore, a comparison of basal neuronal activity differences in different animal groups based on locally specific manganese accumulation is possible using fractionated application. Half-life times of manganese clearance varied between 5 and 7 days, and were longest in the periaqueductal gray, amygdala and entorhinal cortex. As the hippocampal formation shows one of the highest T1 -weighted signal intensities after manganese application, and manganese-induced memory impairment has been suggested, we assessed hippocampus-dependent learning as well as possible manganese-induced atrophy of the hippocampal volume. No interference of manganese application on learning was detected after 4 days of Mn(2+) application or 2 weeks after the application protocol. In addition, no volumetric changes induced by manganese application were found for the hippocampus at any of the measured time points. For longitudinal measurements (i.e. repeated manganese applications), a minimum of at least 8 weeks should be considered using the proposed protocol to allow for sufficient clearance of the paramagnetic ion from cerebral tissue.


Subject(s)
Brain/metabolism , Chlorides/pharmacokinetics , Image Enhancement , Magnetic Resonance Imaging/methods , Manganese Compounds/pharmacokinetics , Animals , Brain/drug effects , Chlorides/toxicity , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL
8.
Mol Psychiatry ; 13(11): 1028-42, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18475271

ABSTRACT

Hypersecretion of central corticotropin-releasing hormone (CRH) has been implicated in the pathophysiology of affective disorders. Both, basic and clinical studies suggested that disrupting CRH signaling through CRH type 1 receptors (CRH-R1) can ameliorate stress-related clinical conditions. To study the effects of CRH-R1 blockade upon CRH-elicited behavioral and neurochemical changes we created different mouse lines overexpressing CRH in distinct spatially restricted patterns. CRH overexpression in the entire central nervous system, but not when overexpressed in specific forebrain regions, resulted in stress-induced hypersecretion of stress hormones and increased active stress-coping behavior reflected by reduced immobility in the forced swim test and tail suspension test. These changes were related to acute effects of overexpressed CRH as they were normalized by CRH-R1 antagonist treatment and recapitulated the effect of stress-induced activation of the endogenous CRH system. Moreover, we identified enhanced noradrenergic activity as potential molecular mechanism underlying increased active stress-coping behavior observed in these animals. Thus, these transgenic mouse lines may serve as animal models for stress-elicited pathologies and treatments that target the central CRH system.


Subject(s)
Central Nervous System/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Stress, Physiological/genetics , Stress, Psychological/genetics , Adaptation, Psychological/drug effects , Adaptation, Psychological/physiology , Analysis of Variance , Animals , Brain Chemistry/drug effects , Central Nervous System/anatomy & histology , Central Nervous System/drug effects , Corticotropin-Releasing Hormone/antagonists & inhibitors , Exploratory Behavior , Female , Fenclonine/administration & dosage , Fenclonine/analogs & derivatives , Hindlimb Suspension , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Intermediate Filament Proteins/genetics , Male , Methyltyrosines/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Proteins/genetics , Pyrazoles/pharmacology , RNA, Untranslated , Radioimmunoassay/methods , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/etiology , Swimming , Triazines/pharmacology
9.
Pharmacogenomics J ; 8(3): 196-208, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17684478

ABSTRACT

Dysregulation of the endocannabinoid system is known to interfere with emotional processing of stressful events. Here, we studied the role of cannabinoid receptor type 1 (CB1) signaling in stress-coping behaviors using the forced swim test (FST) with repeated exposures. We compared effects of genetic inactivation with pharmacological blockade of CB1 receptors both in male and female mice. In addition, we investigated potential interactions of the endocannabinoid system with monoaminergic and neurotrophin systems of the brain. Naive CB1 receptor-deficient mice (CB1-/-) showed increased passive stress-coping behaviors as compared to wild-type littermates (CB1+/+) in the FST, independent of sex. These findings were partially reproduced in C57BL/6N animals and fully reproduced in female CB1+/+ mice by pharmacological blockade of CB1 receptors with the CB1 receptor antagonist SR141716. The specificity of SR141716 was confirmed in female CB1-/- mice, where it failed to affect behavioral performance. Sensitivity to the antidepressants desipramine and paroxetine was preserved, but slightly altered in female CB1-/- mice. There were no genotype differences between CB1+/+ and CB1-/- mice in monoamine oxidase A and B activities under basal conditions, nor in monoamine content of hippocampal tissue after FST exposure. mRNA expression of vesicular glutamate transporter type 1 was unaffected in CB1-/- mice, but mRNA expression of brain-derived neurotrophic factor (BDNF) was reduced in the hippocampus. Our results suggest that impaired CB1 receptor function promotes passive stress-coping behavior, which, at least in part, might relate to alterations in BDNF function.


Subject(s)
Adaptation, Psychological , Receptor, Cannabinoid, CB1/physiology , Signal Transduction/physiology , Stress, Psychological/psychology , Animals , Biogenic Monoamines/analysis , Brain-Derived Neurotrophic Factor/genetics , Desipramine/pharmacology , Female , Hippocampus/chemistry , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/analysis , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Rimonabant , Swimming , Vesicular Glutamate Transport Protein 1/genetics
10.
Genes Brain Behav ; 5 Suppl 2: 5-24, 2006.
Article in English | MEDLINE | ID: mdl-16681797

ABSTRACT

Genes and environment are both essential and interdependent determinants of behavioral responses. Behavioral genetics focuses on the role of genes on behavior. In this article, we aim to provide a succinct, but comprehensive, overview of the different means through which behavioral analysis may be performed in rodents. We give general recommendations for planning and performing behavioral experiments in rats and mice, followed by brief descriptions of experimental paradigms most commonly used for the analysis of reflexes, sensory function, motor function and exploratory, social, emotional and cognitive behavior. We end with a discussion of some of the shortcomings of current concepts of genetic determinism and argue that the genetic basis of behavior should be analyzed in the context of environmental factors.


Subject(s)
Behavior, Animal/physiology , Genetics, Behavioral/methods , Learning/physiology , Motor Activity/genetics , Animals , Cognition/physiology , Emotions/physiology , Environment , Exploratory Behavior/physiology , Mice , Rats , Research Design/standards , Social Behavior , Stereotyped Behavior/physiology
11.
Handb Exp Pharmacol ; (169): 1-34, 2005.
Article in English | MEDLINE | ID: mdl-16594253

ABSTRACT

Learning and memory processes are thought to underlie a variety of human psychiatric disorders, including generalised anxiety disorder and post-traumatic stress disorder. Basic research performed in laboratory animals may help to elucidate the aetiology of the respective diseases. This chapter gives a short introduction into theoretical and practical aspects of animal experiments aimed at investigating acquisition, consolidation and extinction of aversive memories. It describes the behavioural paradigms most commonly used as well as neuroanatomical, cellular and molecular correlates of aversive memories. Finally, it discusses clinical implications of the results obtained in animal experiments in respect to the development of novel pharmacotherapeutic strategies for the treatment of human patients.


Subject(s)
Learning , Memory , Animals , Avoidance Learning , Humans , Models, Animal
12.
Neurosci Biobehav Rev ; 20(3): 341-58, 1996.
Article in English | MEDLINE | ID: mdl-8880728

ABSTRACT

Since the pioneering work of David de Wied and his colleagues, the neuropeptides arginine vasopressin and oxytocin have been thought to play a pivotal role in behavioral regulation in general, and in learning and memory in particular. The present review focuses on the behavioral effects of intracerebral arginine vasopressin and oxytocin, with particular emphasis on the role of these neuropeptides as signals in interneuronal communication. We also discuss several methodological approaches that have been used to reveal the importance of these intracerebral neuropeptides as signals within signaling cascades. The literature suggests that arginine vasopressin improves, and oxytocin impairs, learning and memory. However, a critical analysis of the subject indicates the necessity for a revision of this generalized concept. We suggest that, depending on the behavioral test and the brain area under study, these endogenous neuropeptides are differentially involved in behavioral regulation; thus, generalizations derived from a single behavioral task should be avoided. In particular, recent studies on rodents indicate that socially relevant behaviors triggered by olfactory stimuli and paradigms in which the animals have to cope with an intense stressor (e.g., foot-shock motivated active or passive avoidance) are controlled by both arginine vasopressin and oxytocin released intracerebrally.


Subject(s)
Behavior, Animal/physiology , Learning/physiology , Memory/physiology , Oxytocin/physiology , Vasopressins/physiology , Animals
13.
Neuroscience ; 85(4): 1209-22, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9681958

ABSTRACT

To investigate the effects of an ethologically-relevant stressor on central and peripheral release of arginine vasopressin and oxytocin, we forced adult male Wistar rats to swim for 10 min and simultaneously measured the release of the two peptides (i) within the hypothalamic supraoptic and paraventricular nuclei (by means of the microdialysis technique) and (ii) into the blood (by chronically-implanted jugular venous catheters). Forced swimming caused a significant rise in the release of arginine vasopressin and oxytocin within both the supraoptic nuclei (four-fold and three-fold, respectively) and the paraventricular nuclei (three-fold and four- to five-fold, respectively). Release patterns measured before, during and after repeated stress exposure on three consecutive days indicated that, at the level of the hypothalamus, the two neuropeptides are critically involved in the rats' stress response in a peptide-, locus- and stress-specific manner. Particularly, despite a general reduction of the recovery of the microdialysis probes over the time, the release of arginine vasopressin within the paraventricular nuclei and of oxytocin within the supraoptic nuclei tended to increase upon repeated stress exposure. Measurement of plasma peptide concentrations revealed that the central release of oxytocin was accompanied by a secretion of this peptide into the systemic circulation. In contrast, arginine vasopressin, assayed in the same plasma samples, failed to respond to the stressor. The latter finding is consistent with a dissociated release of the neuropeptide from different parts of a single neuron (soma/dendrites vs axon terminals). It provides evidence that under physiological conditions plasma hormone levels do not necessarily reflect the secretory activity of central components of the respective neuropeptidergic system.


Subject(s)
Central Nervous System/physiology , Neurons/physiology , Neuropeptides/physiology , Oxytocin/metabolism , Peripheral Nerves/physiology , Stress, Psychological/metabolism , Vasopressins/metabolism , Animals , Central Nervous System/cytology , Central Nervous System/metabolism , Corticosterone/blood , Corticotropin-Releasing Hormone/blood , Lactic Acid/blood , Male , Microdialysis , Neurons/metabolism , Osmolar Concentration , Peripheral Nerves/cytology , Peripheral Nerves/metabolism , Rats , Rats, Wistar , Swimming
14.
J Endocrinol ; 164(2): 197-205, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10657855

ABSTRACT

The ageing process in animals and humans is thought to be accompanied by a gradual impairment of corticosteroid receptor function, which is reflected by increased pituitary-adrenocortical hormone secretion at baseline and a number of aberrant neuroendocrine function test results. The latter include the ACTH and corticosteroid responses to a combined dexamethasone (DEX)/corticotropin-releasing hormone (CRH) challenge. The excessive hormonal response to this test among aged individuals has been taken as indirect evidence of enhanced endogenous arginine vasopressin (AVP) release, which - together with peripherally administered CRH - is capable of overriding DEX-induced ACTH suppression. The current study was designed to explore the role of endogenous AVP in mediating excessive hypothalamic-pituitary-adrenocortical (HPA) activity in ageing. The combined DEX/CRH test was administered to aged (22-24 months old) Wistar rats and the effect of the AVP type 1 (V1) receptor antagonist, d(CH(2))(5)Tyr(Me)AVP, on ACTH release was studied. Infusion of the V1 receptor antagonist after DEX pretreatment and before CRH administration prevented the CRH-induced rise in ACTH secretion in comparison with vehicle-treated aged rats (area under the concentration-time curve: 699+/-479 versus 2896+/-759; P<0.01). This difference was absent in young (3 months old) control rats. In situ hybridization showed an increased number of AVP mRNA-expressing neurons in the parvocellular but not the magnocellular, portion of the hypothalamic paraventricular nucleus in DEX-pretreated aged rats. The number and synthetic activity of parvocellular neurons expressing CRH mRNA was also increased. We have concluded that the increased HPA activity in aged rats involves enhanced synthesis and release of AVP from parvocellular neurons, possibly secondary to impaired corticosteroid receptor function.


Subject(s)
Aging/physiology , Hypothalamo-Hypophyseal System/physiology , Vasopressins/physiology , Adrenocorticotropic Hormone/metabolism , Animals , Blood Pressure/drug effects , Corticosterone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Dexamethasone/pharmacology , Heart Rate/drug effects , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar
15.
Prog Brain Res ; 119: 201-20, 1998.
Article in English | MEDLINE | ID: mdl-10074790

ABSTRACT

In addition to its peripheral secretion from the neurohypophysis, the neuropeptide vasopressin (VP) is released within the mammalian brain from probably all parts of the neuronal membrane. In particular the development of brain microdialysis in vivo together with blood microdialysis or blood sampling provides the advantage of being able to reliably compare the dynamic release patterns into different compartments of the organism. The central VP release within hypothalamic (e.g., supraoptic, paraventricular and suprachiasmatic nuclei) and limbic (e.g., septum, amygdala) rat brain areas is stimulated by a variety of substances and stressors, including interleukin-1 beta, social defeat and forced swimming. Furthermore, it is characterized by positive and negative feedback mechanisms and the capacity of the VP system for co-ordinated or independent release, the latter being observed, for example, during social defeat. This emotional stressor, in contrast to exposure to a novel cage, increased VP release within the supraoptic nucleus, but not into plasma. This failure to release VP peripherally could be observed also during forced swimming, despite a dramatic rise in plasma osmolality and a markedly stimulated central release. In another series of experiments we studied the effects of centrally-released VP on cognitive and emotional aspects of behavior using reverse microdialysis for antagonist administration during the behavioral tests and antisense targeting to downregulate either VP or its local V1 receptor subtype. In this way, centrally (in particular septally) released VP could be shown to be causally involved in short-term memory and anxiety-related behavior. Furthermore, VP release within the hypothalamic paraventricular nucleus is likely to provide a negative tonus on the activity of the hypothalamic-pituitary-adrenocortical axis. This neuroendocrine effect together with cognitive, emotional and immunological effects of centrally released VP is thought to be essential to ensure adequate behavior of the animal during challenging situations and to contribute to the development of efficient coping strategies.


Subject(s)
Behavior, Animal/physiology , Brain Chemistry/physiology , Neurosecretory Systems/metabolism , Vasopressins/metabolism , Animals , Rats
16.
J Neuroendocrinol ; 6(2): 121-5, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8049710

ABSTRACT

The purpose of this study was to downregulate the transcriptional message of arginine vasopressin (AVP) by antisense treatment. A complete phosphorothioate antisense oligodesoxynucleotide corresponding to the beginning of the coding region of rat AVP mRNA was constructed and injected into the lateral ventricle of rats. Within 3-6 h animals exhibited a temporary diabetes insipidus, which lasted up to 9 h. Accordingly, vasopressin immunoreactivity in the hypothalamic nuclei was reduced. Our results demonstrate that a specific and reversible inhibition of neuropeptide expression can be accomplished in the intact hypothalamo-neurohypophysial system by antisense treatment, thus providing a novel tool for studies on stimulus-secretion coupling in vivo.


Subject(s)
Arginine Vasopressin/biosynthesis , Diabetes Insipidus/chemically induced , Oligonucleotides, Antisense/pharmacology , Animals , Base Sequence , Diabetes Insipidus/urine , Down-Regulation/drug effects , Injections, Intraventricular , Male , Molecular Sequence Data , Oligonucleotides, Antisense/administration & dosage , Osmolar Concentration , Rats , Rats, Wistar
17.
Neuroreport ; 7(12): 1933-6, 1996 Aug 12.
Article in English | MEDLINE | ID: mdl-8905696

ABSTRACT

The release of arginine vasopressin (AVP) within the suprachiasmatic nucleus (SCN) of anaesthetized male rats was monitored by microdialysis. Dialysis of the nucleus with 56 mM K+ solution (to 400%) as well as with 1 M NaCl-hypertonic medium (to 530%) triggered a significant increase in intra-SCN release of AVP (p < 0.01). To investigate whether endogenous AVP influences its own release, we administered in a second experiment the combined V1/V2 receptor antagonist d(CH2)5[D-Tyr(Et)2,Val4]AVP directly into the dialysed area. Compared with vehicle-treated controls, the antagonist had no effects on intranuclear release of AVP during basal or stimulated conditions. Thus, in contrast to recent observations in the supraoptic nucleus, we found no indications for autoregulatory mechanisms of AVP release within the SCN.


Subject(s)
Antidiuretic Hormone Receptor Antagonists , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism , Vasopressins/metabolism , Animals , Male , Microdialysis , Rats , Rats, Wistar
18.
Neuroreport ; 5(10): 1181-4, 1994 Jun 02.
Article in English | MEDLINE | ID: mdl-7919160

ABSTRACT

A combined microdialysis/microinfusion technique was used to investigate whether arginine vasopressin (AVP) is involved in the regulation of its own release into the extracellular fluid of the supraoptic nucleus in vivo. While intranuclear neuropeptide release was monitored, 0.33 microliter of either vehicle, lysine vasopressin (LVP; 10 ng microliters -1) or a V1/V2 AVP receptor antagonist (100 ng microliters -1) was infused into the supraoptic nucleus of adult male Wistar rats before and during direct osmotic stimulation of the nucleus via the microdialysis probe. Administration of LVP increased basal AVP release, whereas administration of the V1/V2 receptor antagonist attenuated the increase in AVP release during osmotic stimulation observed in vehicle-treated controls. Taken together, these results indicate a receptor-mediated positive feedback action of endogenous AVP on its own release within the supraoptic nucleus.


Subject(s)
Arginine Vasopressin/physiology , Supraoptic Nucleus/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists , Arginine Vasopressin/antagonists & inhibitors , Arginine Vasopressin/metabolism , Feedback/physiology , Lypressin/pharmacology , Male , Microdialysis , Neurons, Afferent/metabolism , Neurons, Efferent/metabolism , Osmolar Concentration , Rats , Rats, Wistar
20.
Regul Pept ; 59(2): 229-39, 1995 Oct 20.
Article in English | MEDLINE | ID: mdl-8584759

ABSTRACT

We studied the role of central amygdala CRH receptors in behavioral responses to an anxiogenic stimulus. An antisense oligodeoxynucleotide corresponding to the rat CRH1 receptor mRNA was infused chronically into the central amygdaloid nucleus of male rats via osmotic minipumps (0.25 micrograms/0.5 microliters/h). Control groups received infusions of either a scrambled sequence oligodeoxynucleotide or vehicle. On the 4th day of treatment, rats were subjected to 10 min of social defeat and immediately afterwards tested on the elevated plus-maze. Antisense oligodeoxynucleotide-treated rats spent significantly more time exploring the open arms of the plus-maze than scrambled sequence- and vehicle-treated animals, both of which did not differ from each other. The social discrimination test, on the other hand, revealed no difference in juvenile recognition abilities among the treatment groups. Using in situ hybridization and receptor autoradiography, we were not able to detect clear signals of CRH1 receptor mRNA and CRH binding sites in the central amygdaloid nucleus of either group, confirming the reportedly low expression and density of CRH receptors in this brain area. The present data support the view that CRH receptors in the central nucleus of the amygdala are involved in the mediation and expression of anxiety-related behavior, but simultaneously raise questions as to the mechanisms of antisense oligodeoxynucleotide action.


Subject(s)
Amygdala/drug effects , Anxiety/drug therapy , Behavior, Animal/drug effects , Oligonucleotides, Antisense/pharmacology , Receptors, Corticotropin-Releasing Hormone/genetics , Social Behavior , Animals , Anxiety/psychology , Autoradiography , Base Sequence , Discrimination Learning/drug effects , Functional Laterality , In Situ Hybridization , Infusions, Parenteral , Male , Maze Learning/drug effects , Molecular Sequence Data , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL