Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










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