Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
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.
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
6.
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
7.
Neurosci Biobehav Rev ; 46 Pt 3: 418-28, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24726577

ABSTRACT

The Transient Receptor Potential Vanilloid Type-1 (TRPV1) was first characterized in primary afferent fibers as a receptor for capsaicin (the pungent ingredient of chili peppers). Later on, this cation-permeable ion channel was also described in the central nervous system, where its main putative endogenous ligand is N-arachidonoyl ethanolamide (an endocannabinoid, also known as anandamide). Recent results employing genetic, pharmacological and histochemical techniques indicate that TRPV1 tonically modulate anxiety, fear and panic responses in brain regions related to defensive responses, such as the dorsal periaqueductal gray, the hippocampus and the medial prefrontal cortex. Genetic deletion or antagonism of this ion channel induces anxiolytic-like effects in several animal models. The main mechanism responsible for TRPV1-mediated effects on anxiety seems to involve facilitation of glutamatergic neurotransmission. In addition, there is evidence for interactions with other neurotransmitter systems, such as nitric oxide and endocannabinoids.


Subject(s)
Brain/metabolism , Defense Mechanisms , TRPV Cation Channels/physiology , Animals , Anxiety/drug therapy , Anxiety/pathology , Disease Models, Animal , Humans , Models, Biological
8.
Brain Res ; 1527: 47-56, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23791919

ABSTRACT

Both the serotonergic and the endocannabinoid system play a major role in mediating fear and anxiety. In the basolateral amygdala (BLA) it has been shown that the cannabinoid receptor 1 (CB1) is highly co-expressed with 5-HT3 receptors on GABAergic interneurons suggesting that 5-HT3 receptor activity modulates CB1-mediated effects on inhibitory synaptic transmission. In the present study, we investigated the possible interactions of CB1 and 5-HT3-mediated neuronal processes in the BLA using electrophysiological and behavioural approaches. Whole-cell patch-clamp recordings were performed in coronal brain slices of mice. Electric stimuli were delivered to the lateral amygdala to evoke GABAA receptor-mediated inhibitory postsynaptic currents (GABAA-eIPSCs) in the BLA. The induction of LTDi, a CB1-mediated depression of inhibitory synaptic transmission, was neither affected by the 5-HT3 antagonists ondansetron (OND; 20 µM) and tropisetron (Trop; 50 nM) nor by the 5-HT3 agonists SR57227A (10 µM). In auditory fear conditioning tests, mice treated with SR57227A (3.0mg/kg i.p.) showed sustained freezing, whereas treatment with Trop (1.0 mg/kg i.p.) decreased the expression of conditioned fear. These effects were overruled by the CB1 antagonist rimonabant (RIM; 3.0 mg/kg), which caused increased freezing with or without co-treatment with Trop. In summary, these experiments do not support a functional interaction between CB1 and 5-HT3 receptors at the level of GABA neurotransmission in the BLA nor in terms of fear regulation.


Subject(s)
Amygdala/metabolism , Fear/physiology , Receptor, Cannabinoid, CB1/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Synaptic Transmission/physiology , Animals , Conditioning, Classical , Electric Stimulation , Endocannabinoids/metabolism , Immunohistochemistry , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neural Inhibition/physiology , Neural Pathways/metabolism , Patch-Clamp Techniques , gamma-Aminobutyric Acid/metabolism
9.
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
10.
Neuroscience ; 204: 159-85, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22173015

ABSTRACT

The endocannabinoid system seems to play very specific roles in fear extinction, which can only be described within a well-defined model of the various fear relief processes. We, therefore, seek to clarify the current conceptual framework of fear relief within classical and operant fear conditioning paradigms as well as propose new clarifications within this framework where necessary. Based on these revisions as well as previous research involving the endocannabinoid system and fear relief, we are able to pinpoint the processes in which endocannabinoids seem to play a significant role. Following auditory-cued fear conditioning, this applies in particular to habituation and its involvement in acute and long-lasting fear relief. Following contextual conditioning, in contrast, endocannabinoids seem to affect relearning processes as well. Furthermore, we describe how the involvement of the endocannabinoid system develops over the course of the fear relief process and what this may imply for the clinical use of pharmacotherapies targeting the endocannabinoid system in treating fear and anxiety disorders.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Conditioning, Classical/physiology , Endocannabinoids , Extinction, Psychological/physiology , Fear/physiology , Receptor, Cannabinoid, CB1/metabolism , Animals , Behavior, Animal/physiology
11.
Neuroscience ; 204: 186-92, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21906661

ABSTRACT

The transient receptor potential vanilloid type 1 channel (TRPV1; originally vanilloid receptor VR1) is activated in peripheral terminals of nociceptive fibers by noxious heat, low pH, and natural products such as capsaicin, the pungent ingredient of red-hot chilli peppers. Evidence has been accumulating that TRPV1 is expressed also in the brain, where it seems to be involved in antinociception, locomotor control, and regulation of affective behaviors. This ion channel might be activated by arachidonoyl ethanolamide (anandamide), the endogenous agonist of the cannabinoid type 1 (CB(1)) receptor. However, while CB(1) activation leads to a decrease in intracellular calcium and attenuation of synaptic transmission, anandamide binding to TRPV1 results in elevated calcium levels and potentiated synaptic transmission. This suggests a tripartite regulatory system with antagonistic effects of CB(1) and TRPV1, which are tied together by the same endogenous ligand. Such a system may have important implication for the modulation of behavioral responses. The present commentary elaborates on this interplay between CB(1) receptors and TRPV1 channels in the context of fear- and anxiety-related behaviors.


Subject(s)
Anxiety/metabolism , Fear/physiology , Receptor, Cannabinoid, CB1/metabolism , TRPV Cation Channels/metabolism , Animals , Signal Transduction/physiology , Stress, Psychological/metabolism
12.
Genes Brain Behav ; 10(2): 137-48, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20846178

ABSTRACT

The role of the cyclic nucleotide-gated (CNG) channel CNGA3 is well established in cone photoreceptors and guanylyl cyclase-D-expressing olfactory neurons. To assess a potential function of CNGA3 in the mouse amygdala and hippocampus, we examined synaptic plasticity and performed a comparative analysis of spatial learning, fear conditioning and step-down avoidance in wild-type mice and CNGA3 null mutants (CNGA3(-/-) ). CNGA3(-/-) mice showed normal basal synaptic transmission in the amygdala and the hippocampus. However, cornu Ammonis (CA1) hippocampal long-term potentiation (LTP) induced by a strong tetanus was significantly enhanced in CNGA3(-/-) mice as compared with their wild-type littermates. Unlike in the hippocampus, LTP was not significantly altered in the amygdala of CNGA3(-/-) mice. Enhanced hippocampal LTP did not coincide with changes in hippocampus-dependent learning, as both wild-type and mutant mice showed a similar performance in water maze tasks and contextual fear conditioning, except for a trend toward higher step-down latencies in a passive avoidance task. In contrast, CNGA3(-/-) mice showed markedly reduced freezing to the conditioned tone in the amygdala-dependent cued fear conditioning task. In conclusion, our study adds a new entry on the list of physiological functions of the CNGA3 channel. Despite the dissociation between physiological and behavioral parameters, our data describe a so far unrecognized role of CNGA3 in modulation of hippocampal plasticity and amygdala-dependent fear memory.


Subject(s)
Behavior, Animal/physiology , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Synapses/physiology , Acoustic Stimulation , Amygdala/metabolism , Animals , Avoidance Learning/physiology , Blotting, Western , Cyclic Nucleotide-Gated Cation Channels/metabolism , Depression/genetics , Depression/psychology , Discrimination Learning/physiology , Excitatory Postsynaptic Potentials/physiology , Fear/psychology , Hippocampus/metabolism , Immunohistochemistry , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reflex, Startle/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sensation/physiology , Swimming/psychology
13.
J Psychiatr Res ; 45(3): 354-60, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20655545

ABSTRACT

Long-lasting presence of avoidance and emotional numbing are reliable behavioral markers for PTSD, but little is known about its psychological and biological underpinnings. We employed our recently established mouse model of PTSD (i) to study the emergence of avoidance behavior in the aftermath of a trauma, (ii) to disentangle the impact of context generalization vs. lack of motivation vs. novelty fear and (iii) to assess the therapeutic value of benzodiazepines and selective serotonin reuptake inhibitors (SSRIs). Specific conditioned avoidance to shock-paired odor turned into generalized avoidance after 28 days of fear incubation. Combination of habituation to the novel environment and extinction of contextual fear abolished both generalized and specific avoidance behavior. Chronic fluoxetine treatment partially reversed the phenotype, whereas acute treatment with diazepam did not. Our animal model may help understanding the mechanisms underlying psychological and biological mechanisms of PTSD for the benefit of developing pharmacotherapeutic strategies, which specifically address generalized avoidance.


Subject(s)
Avoidance Learning , Fear , Animals , Antidepressive Agents, Second-Generation , Avoidance Learning/drug effects , Behavior, Animal , Conditioning, Classical/drug effects , Diazepam/pharmacology , Electroshock/adverse effects , Extinction, Psychological/drug effects , Fear/drug effects , Fluoxetine/pharmacology , Habituation, Psychophysiologic , Hypnotics and Sedatives/pharmacology , Male , Mice , Mice, Inbred C57BL , Odorants , Time Factors
14.
Genes Brain Behav ; 9(8): 947-57, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20659172

ABSTRACT

Environmental factors may unleash genetically determined susceptibility to psychopathology. Great effort has been spent in identifying both the genetic basis and environmental sources of exaggerated fear in animal models of anxiety disorders. Here, we show that the origin of inbred mice, probably via subtle differences in breeding and rearing conditions, may have large consequences specifically on acquisition and retention of fear memories, while leaving anxiety-related behaviours unaffected. These effects could be seen in BALB/cAnN (BALB), but not in C57BL/6N (C57BL/6) mice, thus suggesting their dependency on the genetic background. Increased susceptibility for developing exaggerated fear responses was accompanied by decreased long-term depression and increased surface trafficking of the AMPA receptor GluR1 subunit at the level of the basolateral amygdala complex. Together, these data raise a novel caveat in the debate about the origins of variation in behavioural studies with experimental animals. Considering that there are currently no animal models which explicitly consider conceptual analogy to the specific gene-environment interactions observed in the aetiology of phobias, our study might suggest a novel approach and direction for further preclinical studies focusing on such aspects of phobic-like fears.


Subject(s)
Amygdala/metabolism , Neuronal Plasticity/genetics , Receptors, AMPA/genetics , Age Factors , Animals , Fear , Genetic Predisposition to Disease , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/genetics , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Phobic Disorders , Receptors, AMPA/metabolism , Social Environment , Species Specificity , Statistics, Nonparametric
15.
Neuroscience ; 169(3): 1216-26, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20576499

ABSTRACT

Only a small percentage of individuals develop posttraumatic stress disorder (PTSD) in the aftermath of a trauma. It is still largely unknown to what extent gene-environment interactions contribute to the inter-individual differences in PTSD susceptibility and resilience and what cellular processes may underlie long-term maintenance of the disorder. Here we employed a mouse model of PTSD to unravel the contribution of genetic background and maternal influences on long-lasting changes in kinase and transcription factor activities in PTSD-susceptible C57BL/6NCrl (B6N) and resilient C57BL/6JOlaHsd (B6JOla) mice. Mice received an inescapable foot shock and were tested for activity changes in the AKT/GSK-3beta/beta-catenin-pathway in specific brain structures 42 days later. To control for prenatal and postnatal environmental (i.e. maternal) factors part of the experiments were performed with animals originating from within-strain and between-strain embryo transfers. In PTSD-susceptible B6N mice, long-term maintenance of contextual and sensitized fear was accompanied by (i) increased levels of phosphorylated AKT within the dorsal hippocampus and (ii) higher levels of phosphorylated AKT and GSK-3beta and increased beta-catenin levels within the basolateral amygdala. In animals originating from embryo transfers, levels of phosphorylated GSK-3beta and of beta-catenin were decreased in the dorsal hippocampus, but increased in the basolateral amygdala of shocked B6N mice compared to shocked B6JOla mice. This was independent of the genotype of the recipient mothers. At the behavioural level, these differences coincided with sustained sensitized and more pronounced contextual fear of B6N compared to B6JOla mice. Taken together our study identifies lasting changes in the AKT/GSK-3beta/beta-catenin cascade within the hippocampus and amygdala as molecular correlates of genetically determined differences in the severity of PTSD-like symptoms.


Subject(s)
Amygdala/metabolism , Fear , Glycogen Synthase Kinase 3/physiology , Hippocampus/metabolism , Proto-Oncogene Proteins c-akt/physiology , Stress Disorders, Post-Traumatic/physiopathology , beta Catenin/physiology , Animals , Embryo Transfer , Female , Genetic Predisposition to Disease , Genotype , Glycogen Synthase Kinase 3 beta , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , Species Specificity , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/metabolism
16.
Genes Brain Behav ; 8(7): 685-98, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19563475

ABSTRACT

To investigate the impact of averseness, controllability and familiarity of a test situation on the involvement of the endocannabinoid system in the regulation of exploratory behaviour, we tested conventional and conditional cannabinoid receptor type 1 (CB1)-deficient mice in behavioural paradigms with different emotional load, which depended on the strength of illumination and the ability of the animals to avoid the light stimulus. Complete CB1 null-mutant mice (Total-CB1-KO) showed an anxiogenic-like phenotype under circumstances where they were able to avoid the bright light such as the elevated plus-maze and the light/dark avoidance task. Conditional mutant mice lacking CB1 expression specifically in cortical glutamatergic neurons (Glu-CB1-KO), in contrast, failed to show a similar phenotype under the same experimental conditions. However, both mutant lines showed increased avoidance of open arm exploration during a second exposure to the elevated plus-maze. If tested in situations where the fear eliciting light could not be avoided, Total-CB1-KO mice showed increased thigmotaxis in an open field, decreased social investigation and decreased novel object exploration under aversive light conditions, but not under non-aversive low light. This time, Glu-CB1-KO also showed decreased exploratory behaviour towards objects and conspecific juveniles and increased thigmotaxis in the open field. Taking into consideration that the behavioural performance of wild-type mice was only marginally affected by changes in light intensities, these data indicate that the endocannabinoid system renders exploratory behaviour largely independent of the test averseness. This process differentially involves endocannabinoid-controlled glutamatergic transmission, depending on the controllability of the test situation.


Subject(s)
Avoidance Learning/physiology , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Exploratory Behavior/physiology , Fear/physiology , Glutamic Acid/metabolism , Receptor, Cannabinoid, CB1/genetics , Animals , Behavior, Animal/physiology , Brain Chemistry/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropsychological Tests , Phenotype , Photophobia/genetics , Photophobia/metabolism , Photophobia/physiopathology , Synaptic Transmission/genetics
17.
Genes Brain Behav ; 8(2): 203-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19077175

ABSTRACT

Recent evidence showed that the endocannabinoid system plays an important role in the behavioral adaptation of stress and fear responses. In this study, we chose a behavioral paradigm that includes criteria of both fear and stress responses to assess whether the involvement of endocannabinoids in these two processes rely on common mechanisms. To this end, we delivered a footshock and measured the fear response to a subsequently presented novel tone stimulus. First, we exposed different groups of cannabinoid receptor type 1 (CB(1))-deficient mice (CB(1) (-/-)) and their wild-type littermates (CB(1) (+/+)) to footshocks of different intensities. Only application of an intense footshock resulted in a sustained fear response to the tone in CB(1) (-/-). Using the intense protocol, we next investigated whether endocannabinoids mediate their effects via an interplay with corticotropin-releasing hormone (CRH) signaling. Pharmacological blockade of CB(1) receptors by rimonabant in mice deficient for the CRH receptor type 1 (CRHR1(-/-)) or type 2 (CRHR2(-/-)), and in respective wild-type littermates, resulted in a sustained fear response in all genotypes. This suggests that CRH is not involved in the fear-alleviating effects of CB(1). As CRHR1(-/-) are known to be severely impaired in stress-induced corticosterone secretion, our observation also implicates that corticosterone is dispensable for CB(1)-mediated acute fear adaptation. Instead, conditional mutants with a specific deletion of CB(1) in principal neurons of the forebrain (CaMK-CB(1) (-/-)), or in cortical glutamatergic neurons (Glu-CB(1) (-/-)), showed a similar phenotype as CB(1) (-/-), thus indicating that endocannabinoid-controlled glutamatergic transmission plays an essential role in acute fear adaptation.


Subject(s)
Adaptation, Psychological/physiology , Cannabinoid Receptor Modulators/physiology , Corticotropin-Releasing Hormone/physiology , Endocannabinoids , Fear/physiology , Glutamates/physiology , Neurons/physiology , Acoustic Stimulation , Animals , Electroshock , Fear/psychology , Male , Mice , Mice, Knockout , Piperidines/pharmacology , Prosencephalon/cytology , Prosencephalon/physiology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/physiology , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/physiology , Receptors, Glutamate/genetics , Receptors, Glutamate/physiology , Rimonabant , Signal Transduction/physiology
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...