Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Sci Rep ; 8(1): 4158, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29515159

ABSTRACT

Resected hippocampal tissue from patients with drug-resistant epilepsy presents a unique possibility to test novel treatment strategies directly in target tissue. The post-resection time for testing and analysis however is normally limited. Acute tissue slices allow for electrophysiological recordings typically up to 12 hours. To enable longer time to test novel treatment strategies such as, e.g., gene-therapy, we developed a method for keeping acute human brain slices viable over a longer period. Our protocol keeps neurons viable well up to 48 hours. Using a dual-flow chamber, which allows for microscopic visualisation of individual neurons with a submerged objective for whole-cell patch-clamp recordings, we report stable electrophysiological properties, such as action potential amplitude and threshold during this time. We also demonstrate that epileptiform activity, monitored by individual dentate granule whole-cell recordings, can be consistently induced in these slices, underlying the usefulness of this methodology for testing and/or validating novel treatment strategies for epilepsy.


Subject(s)
Epilepsy, Temporal Lobe/physiopathology , Hippocampus/physiopathology , Neurons , Adolescent , Adult , Child , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Middle Aged , Patch-Clamp Techniques
2.
Psychopharmacology (Berl) ; 234(5): 845-855, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28070619

ABSTRACT

RATIONALE: The 5-choice serial reaction time task (5-CSRTT) is widely used to measure rodent attentional functions. In humans, many attention studies in healthy and clinical populations have used testing based on Bundesen's Theory of Visual Attention (TVA) to estimate visual processing speeds and other parameters of attentional capacity. OBJECTIVES: We aimed to bridge these research fields by modifying the 5-CSRTT's design and by mathematically modelling data to derive attentional parameters analogous to human TVA-based measures. METHODS: C57BL/6 mice were tested in two 1-h sessions on consecutive days with a version of the 5-CSRTT where stimulus duration (SD) probe length was varied based on information from previous TVA studies. Thereafter, a scopolamine hydrobromide (HBr; 0.125 or 0.25 mg/kg) pharmacological challenge was undertaken, using a Latin square design. Mean score values were modelled using a new three-parameter version of TVA to obtain estimates of visual processing speeds, visual thresholds and motor response baselines in each mouse. RESULTS: The parameter estimates for each animal were reliable across sessions, showing that the data were stable enough to support analysis on an individual level. Scopolamine HBr dose-dependently reduced 5-CSRTT attentional performance while also increasing reward collection latency at the highest dose. Upon TVA modelling, scopolamine HBr significantly reduced visual processing speed at both doses, while having less pronounced effects on visual thresholds and motor response baselines. CONCLUSIONS: This study shows for the first time how 5-CSRTT performance in mice can be mathematically modelled to yield estimates of attentional capacity that are directly comparable to estimates from human studies.


Subject(s)
Attention/physiology , Choice Behavior/physiology , Reaction Time/physiology , Visual Perception/physiology , Animals , Attention/drug effects , Behavior, Animal , Choice Behavior/drug effects , Cholinergic Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Models, Theoretical , Psychological Theory , Reaction Time/drug effects , Reward , Scopolamine/pharmacology , Visual Perception/drug effects
3.
Neuropeptides ; 55: 79-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26454711

ABSTRACT

High levels of NPY expression in brain regions important for learning and memory together with its neuromodulatory and neurotrophic effects suggest a regulatory role for NPY in memory processes. Therefore it is not surprising that an increasing number of studies have provided evidence for NPY acting as a modulator of neuroplasticity, neurotransmission, and memory. Here these results are presented in relation to the types of memory affected by NPY and its receptors. NPY can exert both inhibitory and stimulatory effects on memory, depending on memory type and phase, dose applied, brain region, and NPY receptor subtypes. Thus NPY act as a resilience factor by impairing associative implicit memory after stressful and aversive events, as evident in models of fear conditioning, presumably via Y1 receptors in the amygdala and prefrontal cortex. In addition, NPY impairs acquisition but enhances consolidation and retention in models depending on spatial and discriminative types of associative explicit memory, presumably involving Y2 receptor-mediated regulations of hippocampal excitatory transmission. Moreover, spatial memory training leads to increased hippocampal NPY gene expression that together with Y1 receptor-mediated neurogenesis could constitute necessary steps in consolidation and long-term retention of spatial memory. Altogether, NPY-induced effects on learning and memory seem to be biphasic, anatomically and temporally differential, and in support of a modulatory role of NPY at keeping the system in balance. Obtaining further insight into memory-related effects of NPY could inspire the engineering of new therapeutics targeting diseases where impaired learning and memory are central elements.


Subject(s)
Brain/metabolism , Learning/physiology , Memory/physiology , Neuropeptide Y/metabolism , Animals , Fear/physiology , Humans , Receptors, Neuropeptide Y/metabolism
4.
Neuropeptides ; 48(6): 335-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25267070

ABSTRACT

Neuropeptide Y (NPY) causes anxiolytic- and antidepressant-like effects after central administration in rodents. These effects could theoretically be utilized in future gene therapy for anxiety and depression using viral vectors for induction of overexpression of NPY in specific brain regions. Using a recombinant adeno-associated viral (rAAV) vector, we addressed this idea by testing effects on anxiolytic- and depression-like behaviours in adult mice after overexpression of NPY transgene in the amygdala and/or hippocampus, two brain regions implicated in emotional behaviours. In the amygdala, injections of rAAV-NPY caused significant anxiolytic-like effect in the open field, elevated plus maze, and light-dark transition tests. In the hippocampus, rAAV-NPY treatment was associated with anxiolytic-like effect only in the elevated plus maze. No additive effect was observed after combined rAAV-NPY injection into both the amygdala and hippocampus where anxiolytic-like effect was found in the elevated plus maze and light-dark transition tests. Antidepressant-like effects were not detected in any of the rAAV-NPY injected groups. Immobility was even increased in the tail suspension and forced swim tests after intra-amygdaloid rAAV-NPY. Taken together, the present data show that rAAV-NPY treatment may confer non-additive anxiolytic-like effect after injection into the amygdala or hippocampus, being most pronounced in the amygdala.


Subject(s)
Amygdala/metabolism , Anti-Anxiety Agents/administration & dosage , Anxiety/metabolism , Dependovirus/genetics , Genetic Vectors/administration & dosage , Hippocampus/metabolism , Neuropeptide Y/biosynthesis , Animals , Anxiety/genetics , Anxiety/virology , Depression/genetics , Depression/metabolism , Depression/virology , Male , Mice , Motor Activity , Neuropeptide Y/genetics
5.
Eur Neuropsychopharmacol ; 23(12): 1789-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23597878

ABSTRACT

Electroconvulsive therapy remains the most effective treatment for depression including a fast onset of action. However, this therapeutic approach suffers from some potential drawbacks. In the acute phase this includes amnesia. Electroconvulsive stimulation (ECS) has previously been shown to reverse a depression-like state in the chronic mild stress model of depression (CMS), but the effect of ECS on cognition has not previously been investigated. In this study the CMS model was used to induce a depressive-like condition in rats. The study was designed to investigate the acute effect of ECS treatment on working memory and the chronic effect of repeated ECS treatments on depression-like behavior and working memory. The results indicated that, in the acute phase, ECS treatment induced a working memory deficit in healthy controls unexposed to stress, while repeated treatments reversed stress-induced decline in working memory, as well as recovering rats submitted to the CMS paradigm from the anhedonic-like state. Like in the clinical setting, a single ECS exposure was ineffective in inducing remission from a depression-like state.


Subject(s)
Cognition Disorders/etiology , Cognition Disorders/therapy , Electroconvulsive Therapy , Stress, Psychological/complications , Analysis of Variance , Animals , Attention/physiology , Chronic Disease , Disease Models, Animal , Drinking/physiology , Food Preferences/physiology , Male , Rats , Rats, Wistar , Sucrose/administration & dosage , Sweetening Agents/administration & dosage
6.
Transl Psychiatry ; 2: e156, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22948381

ABSTRACT

A recent genome-wide association study in patients with panic disorder (PD) identified a risk haplotype consisting of two single-nucleotide polymorphisms (SNPs) (rs7309727 and rs11060369) located in intron 3 of TMEM132D to be associated with PD in three independent samples. Now we report a subsequent confirmation study using five additional PD case-control samples (n = 1670 cases and n = 2266 controls) assembled as part of the Panic Disorder International Consortium (PanIC) study for a total of 2678 cases and 3262 controls in the analysis. In the new independent samples of European ancestry (EA), the association of rs7309727 and the risk haplotype rs7309727-rs11060369 was, indeed, replicated, with the strongest signal coming from patients with primary PD, that is, patients without major psychiatric comorbidities (n = 1038 cases and n = 2411 controls). This finding was paralleled by the results of the meta-analysis across all samples, in which the risk haplotype and rs7309727 reached P-levels of P = 1.4e-8 and P = 1.1e-8, respectively, when restricting the samples to individuals of EA with primary PD. In the Japanese sample no associations with PD could be found. The present results support the initial finding that TMEM132D gene contributes to genetic susceptibility for PD in individuals of EA. Our results also indicate that patient ascertainment and genetic background could be important sources of heterogeneity modifying this association signal in different populations.


Subject(s)
Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Haplotypes/genetics , Membrane Proteins/genetics , Panic Disorder/genetics , Polymorphism, Single Nucleotide/genetics , Adult , Female , Humans , Male , White People/genetics
7.
Neuropeptides ; 46(2): 71-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22342800

ABSTRACT

Neuropeptide Y (NPY) has been implicated in anxiolytic- and antidepressant-like behaviour as well as seizure-suppressant effects in rodents. Although these effects appear to be predominantly mediated via other NPY receptors (Y1 and/or Y2), several studies have also indicated a role for Y5 receptors. Gene therapy using recombinant viral vectors to induce overexpression of NPY, Y1 or Y2 receptors in the hippocampus or amygdala has previously been shown to modulate emotional behaviour and seizures in rodents. The present study explored the potential effects of gene therapy with the Y5 receptor, by testing effects of recombinant adeno-associated viral vector (rAAV) encoding Y5 (rAAV-Y5) in anxiety- and depression-like behaviour as well as in kainate-induced seizures in adult mice. The rAAV-Y5 vector injected into the hippocampus and amygdala induced a pronounced and sustained increase in Y5 receptor mRNA expression and functional Y5 receptor binding, but no significant effects were found with regard to anxiety- and depression-like behaviours or seizure susceptibility. Instead, rAAV-mediated Y5 receptor transgene overexpression resulted in moderate hyperactivity in the open field test. These results do not support a potential role for single transgene overexpression of Y5 receptors for modulating anxiety-/depression-like behaviours or seizures in adult mice. Whether the induction of hyperactivity by rAAV-Y5 could be relevant for other conditions remains to be studied.


Subject(s)
Anxiety/physiopathology , Depression/physiopathology , Hyperkinesis/metabolism , Receptors, Neuropeptide Y/metabolism , Seizures/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/physiology , Genetic Therapy , Genetic Vectors/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred BALB C , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Receptors, Neuropeptide Y/agonists , Seizures/chemically induced , Seizures/therapy
8.
J Neurosci Res ; 90(2): 498-507, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21971867

ABSTRACT

Neuropeptide Y (NPY) exerts anxiolytic- and antidepressant-like effects in rodents that appear to be mediated via Y1 receptors. Gene therapy using recombinant viral vectors to induce overexpression of NPY in the hippocampus or amygdala has previously been shown to confer anxiolytic-like effect in rodents. The present study explored an alternative and more specific approach: overexpression of Y1 receptors. Using a recombinant adeno-associated viral vector (rAAV) encoding the Y1 gene (rAAV-Y1), we, for the first time, induced overexpression of functional transgene Y1 receptors in the hippocampus of adult mice and tested the animals in anxiety- and depression-like behavior. Hippocampal Y1 receptors have been suggested to mediate seizure-promoting effect, so the effects of rAAV-induced Y1 receptor overexpression were also tested in kainate-induced seizures. Y1 receptor transgene overexpression was found to be associated with modest anxiolytic-like effect in the open field and elevated plus maze tests, but no effect was seen on depression-like behavior using the tail suspension and forced swim tests. However, the rAAV-Y1 vector modestly aggravated kainate-induced seizures. These data indicate that rAAV-induced overexpression of Y1 receptors in the hippocampus could confer anxiolytic-like effect accompanied by a moderate proconvulsant adverse effect. Further studies are clearly needed to determine whether Y1 gene therapy might have a future role in the treatment of anxiety disorders.


Subject(s)
Anti-Anxiety Agents/administration & dosage , Convulsants/administration & dosage , Dependovirus/genetics , Gene Expression Regulation, Viral , Genetic Vectors/administration & dosage , Hippocampus/metabolism , Receptors, Neuropeptide Y/biosynthesis , Seizures/metabolism , Animals , Convulsants/toxicity , Genetic Vectors/toxicity , Male , Mice , Receptors, Neuropeptide Y/genetics , Recombinant Fusion Proteins/genetics , Seizures/genetics , Seizures/virology
9.
J Neural Transm (Vienna) ; 118(6): 969-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21416264

ABSTRACT

Panic disorder (PD) is an anxiety disorder characterized by recurrent panic attacks with a lifetime prevalence of 4.7%. Genetic factors are known to contribute to the development of the disorder. Several lines of evidence point towards a major role of the norepinephrine system in the pathogenesis of PD. The SLC6A2 gene is located on chromosome 16q12.2 and encodes the norepinephrine transporter (NET), responsible for the reuptake of norepinephrine into presynaptic nerve terminals. The aim of the present study was to analyze genetic variants located within the NET gene for association with PD. The case-control sample consisted of 449 patients with PD and 279 ethnically matched controls. All cases fulfilled the ICD-10 diagnostic criteria for PD. Genotyping was performed using the Sequenom platform (Sequenom, Inc, San Diego, USA). To test for allelic and haplotypic association, the PLINK software was used, and COMBASSOC was applied to test for gene-wise association. After quality control 29 single nucleotide polymorphisms (SNPs) spanning the gene-region were successfully analyzed. Seven SNPs located within the 5' end of the gene were significantly associated with PD. Furthermore, the NET gene showed overall evidence for association with the disease (P = 0.000035). In conclusion, the present study indicates that NET could be a susceptibility gene for PD.


Subject(s)
Genetic Predisposition to Disease/genetics , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine/metabolism , Panic Disorder/genetics , Panic Disorder/metabolism , Adult , Case-Control Studies , Female , Humans , Male , Middle Aged , Panic Disorder/physiopathology , Polymorphism, Single Nucleotide/genetics
10.
Behav Brain Res ; 216(2): 585-91, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20816900

ABSTRACT

Stressful life events and chronic stress are implicated in the development of depressive disorder in humans. Neuropeptide Y (NPY) and galanin have been shown to modulate the stress response, and exert antidepressant-like effects in rodents. To further investigate these neuropeptides in depression-like behaviour, NPY and galanin gene expression was studied in brains of mice subjected to chronic restraint stress (CRS) and concomitant treatment with the antidepressant fluoxetine (FLX). CRS caused a significant increase in depression-like behaviour that was associated with increased NPY mRNA levels in the medial amygdala. Concomitant FLX treatment reverted depression-like effects of CRS and led to significant increases in levels of NPY and galanin mRNA in the dentate gyrus, amygdala, and piriform cortex. These findings suggest that effects on NPY and galanin gene expression could play a role in the antidepressant effects of FLX.


Subject(s)
Brain/metabolism , Depression/metabolism , Fluoxetine/pharmacology , Galanin/metabolism , Neuropeptide Y/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Amygdala/drug effects , Amygdala/metabolism , Animals , Brain/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Depression/drug therapy , Depression/etiology , Galanin/drug effects , Galanin/genetics , Male , Mice , Neuropeptide Y/drug effects , Neuropeptide Y/genetics , Parahippocampal Gyrus/drug effects , Parahippocampal Gyrus/metabolism , RNA, Messenger/analysis , Restraint, Physical/physiology , Restraint, Physical/psychology , Temporal Lobe/drug effects , Temporal Lobe/metabolism
11.
J Neurosci Res ; 88(16): 3635-43, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20936701

ABSTRACT

Even though induction of seizures by electroconvulsive stimulation (ECS) is a treatment widely used for major depression in humans, the working mechanism of ECS remains uncertain. The antiepileptic effect of ECS has been suggested to be involved in mediating the therapeutic effect of ECS. The neuropeptide galanin exerts antiepileptic and antidepressant-like effects and has also been implicated in the pathophysiology of depression. To explore a potential role of galanin in working mechanisms of ECS, the present study examined effects of repeated ECS on the galanin system using QRT-PCR, in situ hybridization, and [(125) I]galanin receptor binding. ECS was administered to adult mice daily for 14 days, and this paradigm was confirmed to exert antidepressant-like effect in the tail suspension test. Prominent increases in galanin gene expression were found in several brain regions involved in regulation of epileptic activity and depression, including the piriform cortex, hippocampal dentate gyrus, and amygdala. Likewise, GalR2 gene expression was up-regulated in both the central and the medial amygdala, whereas GalR1 gene expression showed a modest down-regulation in the medial amygdala. [(125) I]galanin receptor binding in the piriform cortex, hippocampus, and amygdala was found to be significantly down-regulated. These data show that the galanin system is regulated by repeated ECS in a number of brain regions implicated in seizure regulation and depression. These changes may play a role in the therapeutic effect of ECS.


Subject(s)
Brain/metabolism , Depression/metabolism , Electroshock , Galanin/metabolism , Seizures/metabolism , Animals , Brain/physiopathology , Depression/therapy , Electroconvulsive Therapy , Gene Expression Regulation/physiology , Male , Mice , Tissue Distribution
12.
Acta Psychiatr Scand ; 120(4): 281-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19753663

ABSTRACT

OBJECTIVE: To identify whether a genetic variation (rs1800857; IVS1-5T>C) in the neuropeptide cholecystokinin-A receptor (CCKAR) gene is a risk factor in the pathogenesis of schizophrenia. METHOD: The variation was analysed in a case-control design comprising 508 patients with schizophrenia and 1619 control subjects. A possible functional impact of this variant on CCKAR protein synthesis through alterations in splicing was analysed in an exon-trapping assay. RESULTS: In males only, the risk variant, IVS1-5C, was associated with a significantly increased risk of schizophrenia. Carrying one risk allele was associated with an increased risk of 1.74 (Odds Ratio, OR) and homozygosity (CC) was associated with an OR of 3.19. The variation had no impact on protein synthesis of CCKAR. CONCLUSION: This is the first report associating the CCKAR gene variant with schizophrenia specifically in men. Our study strengthens the conclusion that a CCKAR dysfunction could be involved in the aetiology of schizophrenia.


Subject(s)
Gene Expression/genetics , Introns/genetics , Receptor, Cholecystokinin A/genetics , Schizophrenia/genetics , Adult , Case-Control Studies , Chromosomes, Human, Pair 4/genetics , Denmark/epidemiology , Diagnostic and Statistical Manual of Mental Disorders , Female , Humans , International Classification of Diseases , Male , Polymorphism, Single Nucleotide/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics , Schizophrenia/diagnosis , Schizophrenia/epidemiology , Severity of Illness Index , Sex Distribution
13.
J Neurosci Res ; 87(10): 2386-97, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19267419

ABSTRACT

Previous studies show that chronic ethanol treatment induces prominent changes in brain neuropeptide Y (NPY). The purpose of the present study was to explore ethanol effects at a deeper NPY-system level, measuring expression of NPY and its receptors (Y1, Y2, Y5) as well as NPY receptor binding and NPY-stimulated [(35)S]GTPgammaS functional binding. Rats received intragastric ethanol repeatedly for 4 days, and the NPY system was studied in the hippocampal dentate gyrus (DG), CA3, CA1, and piriform cortex (PirCx) and neocortex (NeoCx) during intoxication, peak withdrawal (16 hr), late withdrawal (3 days), and 1 week after last ethanol administration. NPY mRNA levels decreased during intoxication and at 16 hr in hippocampal regions but increased in the PirCx and NeoCx at 16 hr. NPY mRNA levels were increased at 3 days and returned to control levels in most regions at 1 week. Substantial changes also occurred at the receptor level. Thus Y1, Y2, and Y5 mRNA labelling decreased at 16 hr in most regions, returning to control levels at 3 days, except for PirCx Y2 mRNA, which increased at 3 days and 1 week. Conversely, increases in NPY receptor binding occurred in hippocampal regions during intoxication and in functional binding in the DG and NeoCx during intoxication and at 16 hr and in PirCx during intoxication and at 1 week. Thus this study shows that ethanol intoxication and withdrawal induce complex plastic changes in the NPY system, with decreased/increased gene expression or binding occurring in a time- and region-specific manner. These changes may play an important role in mediating ethanol-induced changes in neuronal excitability.


Subject(s)
Alcohol-Induced Disorders, Nervous System , Brain/pathology , Neuronal Plasticity/physiology , Neuropeptide Y/metabolism , Substance Withdrawal Syndrome , Alcohol-Induced Disorders, Nervous System/metabolism , Alcohol-Induced Disorders, Nervous System/pathology , Alcohol-Induced Disorders, Nervous System/physiopathology , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Neuropeptide Y/pharmacology , Protein Binding/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/physiopathology , Sulfur Isotopes/metabolism , Time Factors , Up-Regulation/drug effects , Up-Regulation/physiology
14.
J Neurosci Res ; 84(6): 1282-91, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16941487

ABSTRACT

Although electroconvulsive seizures (ECS) are widely used as a treatment for severe depression, the working mechanism of ECS remains unclear. Repeated ECS causes anticonvulsant effects that have been proposed to underlie the therapeutic effect of ECS, and neuropeptide Y (NPY) is a potential candidate for mediating this anticonvulsant effect. Repeated ECS results in prominent increases in NPY synthesis. In contrast, NPY-sensitive receptor binding is decreased, so it is unclear whether ECS causes a net increase in NPY signalling. Agonist-stimulated [35S]GTPgammaS binding is a method for detecting functional activation of G-protein-coupled receptors. The present study in mice examined the effects of daily ECS for 14 days on NPY-stimulated [35S]GTPgammaS functional binding and compared this with gene expression of NPY and NPY receptors as well as [125I]peptide YY (PYY) binding in hippocampus of the same animals. Significant increases in NPY mRNA and concomitant reductions in NPY-sensitive binding were found in the dentate gyrus, hippocampal CA1, and neocortex of ECS treated mice, which is consistent with previous rat data. These changes remained significant 1 week after repeated ECS. Significant increases in NPY Y1, Y2, and Y5 mRNA were found in the dentate gyrus after ECS. Surprisingly, unaltered levels of functional NPY receptor binding accompanied the decreased NPY-sensitive binding. This suggests that mechanisms coupling NPY receptor stimulation to G-protein activation could be augmented after repeated ECS. Thus increased synthesis of NPY after repeated ECS should result in a net increase in NPY signalling in spite of reduced levels of NPY-sensitive binding.


Subject(s)
Electroshock , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Neuropeptide Y/pharmacology , Seizures/physiopathology , Signal Transduction/drug effects , Animals , Brain Chemistry/drug effects , Female , Hippocampus/drug effects , Hippocampus/metabolism , In Situ Hybridization , Mice , Mice, Inbred Strains , RNA, Messenger/biosynthesis , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/genetics , Seizures/metabolism
15.
Acta Psychiatr Scand ; 113(1): 54-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16390370

ABSTRACT

OBJECTIVE: A polymorphism in the promoter region of the NPY gene at position -399 C > T was recently reported to be associated with schizophrenia in a Japanese population and with treatment refractory unipolar depression in a Swedish population. The objective of this study was to investigate potential associations between the polymorphism and three psychiatric disorders in a Danish population. METHOD: We investigated the occurrence of the polymorphism in patients with schizophrenia (n = 291), unipolar depression (n = 256) and panic disorder (n = 142) compared with controls (n = 716). RESULTS: We detected the polymorphism -399 C > T at a frequency of 48% in controls. No significant differences were found between genotype or allele frequencies in controls vs. the patient groups. CONCLUSION: The lack of association between the -399 C > T polymorphism and schizophrenia, unipolar depression or panic disorder, respectively, suggests that the polymorphism is not involved in the etiology of these disorders in the Danish population.


Subject(s)
Alleles , Depressive Disorder/ethnology , Depressive Disorder/genetics , Neuropeptide Y/genetics , Panic Disorder/ethnology , Panic Disorder/genetics , Polymorphism, Genetic/genetics , Schizophrenia/ethnology , Schizophrenia/genetics , Adult , Aged , Aged, 80 and over , DNA Primers/genetics , Denmark/epidemiology , Depressive Disorder/epidemiology , Female , Gene Frequency/genetics , Genotype , Humans , Incidence , Male , Middle Aged , Panic Disorder/epidemiology , Polymerase Chain Reaction , Prevalence , Schizophrenia/epidemiology
16.
Brain Res ; 1064(1-2): 161-5, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16309632

ABSTRACT

The temporal profile of Arc gene expression after acute and chronic electroconvulsive stimulations (ECS) was studied using semi-quantitative in situ hybridisation in the rat cortex. A single ECS strongly and temporarily increased Arc mRNA levels in dentate granular cells with maximal induction seen up to 4 h after the stimulus, but returned to baseline at 24 h. A single ECS also increased expression of Arc mRNA in the CA1 and the parietal cortex, but the expression peaked within 1 h and returned to baseline levels within 2 h. Repeated or chronic ECS is a model of electroconvulsive therapy and it would be predicted that gene products involved in antidepressant effects accumulate after repeated ECS. However, repeated ECS reduced Arc gene expression in the CA1 24 h after the last stimulus. These results indicate that Arc is an immediate early gene product regulated by an acute excitatory stimulus, but not accumulated by long term repetitive ECS and therefore not a molecular biomarker for antidepressant properties. More likely, Arc is likely a molecular link to the decline in memory consolidation seen in depressive patients subjected to electroconvulsive therapy.


Subject(s)
Cerebral Cortex/metabolism , Cytoskeletal Proteins/metabolism , Electroshock , Nerve Tissue Proteins/metabolism , Parietal Lobe/metabolism , RNA, Messenger/metabolism , Animals , Cytoskeletal Proteins/genetics , Depressive Disorder/metabolism , Depressive Disorder/therapy , Disease Models, Animal , Electroconvulsive Therapy , Gene Expression Regulation/physiology , Hippocampus/metabolism , Male , Memory/physiology , Nerve Tissue Proteins/genetics , Rats , Rats, Sprague-Dawley
17.
Neuropeptides ; 38(4): 253-60, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15337377

ABSTRACT

The endogenous NPY system in the brain is centrally involved in seizure regulation. The present paper reviews the evidence that exogenously applied NPY receptor ligands can inhibit epileptic seizures in various rodent in vitro and in vivo models. Agonists at Y2 and/or Y5 receptors and antagonists at Y1 receptors appear to inhibit seizures, depending on the seizure model studied. Although progress has been made, further studies are needed using transgenic animals as well as novel selective agonists and antagonists to firmly identify the NPY receptors mediating antiepileptic effects. This may lead to the development of future antiepileptic drug treatments targeting the NPY system.


Subject(s)
Anticonvulsants/metabolism , Neuropeptide Y , Receptors, Neuropeptide Y/metabolism , Seizures/metabolism , Animals , Brain/anatomy & histology , Brain/metabolism , Electric Stimulation , Ligands , Neuropeptide Y/agonists , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/metabolism , Seizures/chemically induced
18.
Neurosci Lett ; 333(2): 127-30, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12419497

ABSTRACT

Previous studies show that neuropeptide Y (NPY) inhibits in vitro seizures in rodent hippocampus. Here, we explored the effect of NPY application on epileptiform discharges induced by perfusion with magnesium-free solution in slices of entorhinal cortex from two different mouse strains. NPY significantly reduced the duration of epileptiform discharges with a peak effect of 36-50%. This is the first study showing anti-epileptiform effect of NPY in the entorhinal cortex and also the first evidence that NPY inhibits seizures in a cortical region in mice. The entorhinal cortex has a central role in transferring information between the hippocampus and the rest of the brain. Therefore our data further strengthen the concept of NPY and its receptors as widespread regulators of epileptiform activity and as a potential future target for antiepileptic therapy.


Subject(s)
Action Potentials/drug effects , Anticonvulsants/pharmacology , Entorhinal Cortex/physiology , Neuropeptide Y/pharmacology , Animals , Culture Media , Epilepsy/chemically induced , Magnesium Deficiency , Male , Mice , Organ Culture Techniques , Patch-Clamp Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...