Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...