Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Pharmacol ; 173(12): 1925-38, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26844810

RESUMEN

BACKGROUND AND PURPOSE: Avoiding danger and finding food are closely related behaviours that are essential for surviving in a natural environment. Growing evidence supports an important role of gut-brain peptides in modulating energy homeostasis and emotional-affective behaviour. For instance, postprandial release of pancreatic polypeptide (PP) reduced food intake and altered stress-induced motor activity and anxiety by activating central Y4 receptors. EXPERIMENTAL APPROACH: We characterized [K(30) (PEG2)]hPP2-36 as long-acting Y4 receptor agonist and injected it peripherally into wildtype and Y4 receptor knockout (Y4KO) C57Bl/6NCrl mice to investigate the role of Y4 receptors in fear conditioning. Extinction and relapse after extinction was measured by spontaneous recovery and renewal. KEY RESULTS: The Y4KO mice showed impaired cued and context fear extinction without affecting acquisition, consolidation or recall of fear. Correspondingly, peripheral injection of [K(30) (PEG2)]hPP2-36 facilitated extinction learning upon fasting, an effect that was long-lasting and generalized. Furthermore, peripherally applied [K(30) (PEG2)]hPP2-36 before extinction inhibited the activation of orexin-expressing neurons in the lateral hypothalamus in WT, but not in Y4KO mice. CONCLUSIONS AND IMPLICATIONS: Our findings suggests suppression of excessive arousal as a possible mechanism for the extinction-promoting effect of central Y4 receptors and provide strong evidence that fear extinction requires integration of vegetative stimuli with cortical and subcortical information, a process crucially depending on Y4 receptors. Importantly, in the lateral hypothalamus two peptide systems, PP and orexin, interact to generate an emotional response adapted to the current homeostatic state. Detailed investigations of feeding-relevant genes may thus deliver multiple intervention points for treating anxiety-related disorders.


Asunto(s)
Señales (Psicología) , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Polipéptido Pancreático/farmacología , Receptores de Neuropéptido Y/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Neuropéptido Y/deficiencia
2.
Neuropeptides ; 55: 111-26, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26444585

RESUMEN

While anxiety disorders are the brain disorders with the highest prevalence and constitute a major burden for society, a considerable number of affected people are still treated insufficiently. Thus, in an attempt to identify potential new anxiolytic drug targets, neuropeptides have gained considerable attention in recent years. Compared to classical neurotransmitters they often have a regionally restricted distribution and may bind to several distinct receptor subtypes. Neuropeptide Y (NPY) is a highly conserved neuropeptide that is specifically concentrated in limbic brain areas and signals via at least 5 different G-protein-coupled receptors. It is involved in a variety of physiological processes including the modulation of emotional-affective behaviors. An anxiolytic and stress-reducing property of NPY is supported by many preclinical studies. Whether NPY may also interact with processing of learned fear and fear extinction is comparatively unknown. However, this has considerable relevance since pathological, inappropriate and generalized fear expression and impaired fear extinction are hallmarks of human post-traumatic stress disorder and a major reason for its treatment-resistance. Recent evidence from different laboratories emphasizes a fear-reducing role of NPY, predominantly mediated by exogenous NPY acting on Y1 receptors. Since a reduction of fear expression was also observed in Y1 receptor knockout mice, other Y receptors may be equally important. By acting on Y2 receptors, NPY promotes fear extinction and generates a long-term suppression of fear, two important preconditions that could support cognitive behavioral therapies in human patients. A similar effect has been demonstrated for the closely related pancreatic polypeptide (PP) when acting on Y4 receptors. Preliminary evidence suggests that NPY modulates fear in particular by activation of Y1 and Y2 receptors in the basolateral and central amygdala, respectively. In the basolateral amygdala, NPY signaling activates inhibitory G protein-coupled inwardly-rectifying potassium channels or suppresses hyperpolarization-induced I(h) currents in a Y1 receptor-dependent fashion, favoring a general suppression of neuronal activity. A more complex situation has been described for the central extended amygdala, where NPY reduces the frequency of inhibitory and excitatory postsynaptic currents. In particular the inhibition of long-range central amygdala output neurons may result in a Y2 receptor-dependent suppression of fear. The role of NPY in processes of learned fear and fear extinction is, however, only beginning to emerge, and multiple questions regarding the relevance of endogenous NPY and different receptor subtypes remain elusive. Y2 receptors may be of particular interest for future studies, since they are the most prominent Y receptor subtype in the human brain and thus among the most promising therapeutic drug targets when translating preclinical evidence to potential new therapies for human anxiety disorders.


Asunto(s)
Encéfalo/metabolismo , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Animales , Humanos , Neuropéptidos/metabolismo
3.
Brain Struct Funct ; 221(7): 3373-91, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26365505

RESUMEN

The amygdala is essential for generating emotional-affective behaviors. It consists of several nuclei with highly selective, elaborate functions. In particular, the central extended amygdala, consisting of the central amygdala (CEA) and the bed nucleus of the stria terminalis (BNST) is an essential component actively controlling efferent connections to downstream effectors like hypothalamus and brain stem. Both, CEA and BNST contain high amounts of different neuropeptides that significantly contribute to synaptic transmission. Among these, neuropeptide Y (NPY) has emerged as an important anxiolytic and fear-reducing neuromodulator. Here, we characterized the expression, connectivity and electrophysiological function of NPY and Y2 receptors within the CEA. We identified several NPY-expressing neuronal populations, including somatostatin- and calretinin-expressing neurons. Furthermore, in the main intercalated nucleus, NPY is expressed primarily in dopamine D1 receptor-expressing neurons but also in interspersed somatostatin-expressing neurons. Interestingly, NPY neurons did not co-localize with the Y2 receptor. Retrograde tract tracing experiments revealed that NPY neurons reciprocally connect the CEA and BNST. Functionally, the Y2 receptor agonist PYY3-36, reduced both, inhibitory as well as excitatory synaptic transmission in the centromedial amygdala (CEm). However, we also provide evidence that lack of NPY or Y2 receptors results in increased GABA release specifically at inhibitory synapses in the CEm. Taken together, our findings suggest that NPY expressed by distinct populations of neurons can modulate afferent and efferent projections of the CEA via presynaptic Y2 receptors located at inhibitory and excitatory synapses.


Asunto(s)
Amígdala del Cerebelo/citología , Amígdala del Cerebelo/fisiología , Potenciales Postsinápticos Excitadores , Potenciales Postsinápticos Inhibidores , Neuronas/fisiología , Neuropéptido Y/fisiología , Receptores de Neuropéptido Y/fisiología , Amígdala del Cerebelo/metabolismo , Animales , Calbindina 2/metabolismo , Núcleo Amigdalino Central/citología , Núcleo Amigdalino Central/metabolismo , Núcleo Amigdalino Central/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Neuropéptido Y/metabolismo , Núcleos Septales/citología , Núcleos Septales/metabolismo , Núcleos Septales/fisiología , Somatostatina/metabolismo
4.
Neuropharmacology ; 99: 665-74, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26314208

RESUMEN

The amygdala is fundamental for associative fear and extinction learning. Recently, also the central nucleus of the amygdala (CEA) has emerged as a site of plasticity actively controlling efferent connections to downstream effector brain areas. Although synaptic transmission is primarily mediated by glutamate and GABA, neuropeptides critically influence the overall response. While neuropeptide Y (NPY) acting via postsynaptic Y1 receptors exerts an important anxiolytic and fear-reducing action, the role of the predominantly presynaptic Y2 receptors is less defined. To investigate the role of Y2 receptors in the CEA we employed viral-vector mediated over-expression of the Y2 selective agonist NPY3-36 in fear conditioning and extinction experiments. NPY3-36 over-expression in the CEA resulted in reduced fear expression during fear acquisition and recall. Interestingly, this effect was blocked by intraperitoneal injection of a brain-penetrant Y2 receptor antagonist. Furthermore, over-expression of NPY3-36 in the CEA also reduced fear expression during fear extinction of CS-induced but not context-related fear. Again, fear extinction appeared delayed by peripheral injection of a Y2 receptor antagonist JNJ-31020028. Importantly, mice with over-expression of NPY3-36 in the CEA also displayed reduced spontaneous recovery and reinstatement, suggesting that Y2 receptor activation supports a permanent suppression of fear. Local deletion of Y2 receptors in the CEA, on the other hand, increased the expression of CS-induced freezing during fear recall and fear extinction. Thus, NPY inhibits fear learning and promotes cued extinction by reducing fear expression also via activation of presynaptic Y2 receptors on CEA neurons.


Asunto(s)
Núcleo Amigdalino Central/metabolismo , Miedo/fisiología , Receptores de Neuropéptido Y/metabolismo , Animales , Benzamidas/administración & dosificación , Núcleo Amigdalino Central/efectos de los fármacos , Fármacos del Sistema Nervioso Central/administración & dosificación , Señales (Psicología) , Dependovirus/genética , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Miedo/efectos de los fármacos , Vectores Genéticos , Masculino , Recuerdo Mental/efectos de los fármacos , Recuerdo Mental/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptido Y/administración & dosificación , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Pruebas Neuropsicológicas , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Piperazinas/administración & dosificación , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/genética
5.
Neuroscience ; 236: 345-72, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23337532

RESUMEN

The GABAA receptor is the main inhibitory receptor in the brain and its subunits originate from different genes or gene families (α1-α6, ß1-ß3, γ1-γ3, δ, ε, θ, π, or ρ1-3). In the mouse brain the anatomical distribution of GABAA receptor subunit mRNAs so far investigated is restricted to subunits forming benzodiazepine-sensitive receptor complexes (α1-α3, α5, ß2, ß3 and γ2) in the forebrain and midbrain as assessed by in situ hybridization (ISH). In the present study the anatomical distribution of the GABAA receptor subunits α1-α6, ß1-ß3, γ1-γ2 and δ was analyzed in the mouse brain (excluding brain stem) by ISH and immunohistochemistry (IHC). In several brain areas such as hippocampus, cerebellum, bulbus olfactorius and habenula we observed that mRNA levels did not reflect protein levels, indicating that the protein is located far distantly from the cell body. We also compared the distribution of these 12 subunit mRNAs and proteins with that reported in the rat brain. Although in general there is a considerable correspondence in the distribution between mouse and rat brains, several species-specific differences were observed.


Asunto(s)
Encéfalo/metabolismo , Receptores de GABA-A/análisis , Receptores de GABA-A/biosíntesis , Animales , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Ratas , Especificidad de la Especie
6.
Br J Pharmacol ; 166(4): 1461-73, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22289084

RESUMEN

BACKGROUND AND PURPOSE: Neuropeptide Y (NPY) and its receptors have been implicated in the control of emotional-affective processing, but the mechanism is unclear. While it is increasingly evident that stimulation of Y1 and inhibition of Y2 receptors produce prominent anxiolytic and antidepressant effects, the contribution of the individual NPY receptor subtypes in the acquisition and extinction of learned fear are unknown. EXPERIMENTAL APPROACH: Here we performed Pavlovian fear conditioning and extinction in NPY knockout (KO) and in NPY receptor KO mice. KEY RESULTS: NPY KO mice display a dramatically accelerated acquisition of conditioned fear. Deletion of Y1 receptors revealed only a moderately accelerated acquisition of conditioned fear, while lack of Y2 receptors was without any effect on fear learning. However, the strong phenotype seen in NPY KO mice was reproduced in mice lacking both Y1 and Y2 receptors. In addition, NPY KO mice showed excessive recall of conditioned fear and impaired fear extinction. This behaviour was replicated only after deletion of both Y1 and Y2 receptors. In Y1 receptor single KO mice, fear extinction was delayed and was unchanged in Y2 receptor KO mice. Deletion of NPY and particularly Y2 receptors resulted in a generalization of conditioned fear. CONCLUSIONS AND IMPLICATIONS: Our data demonstrate that NPY delays the acquisition, reduces the expression of conditioned fear while promoting fear extinction. Although these effects appear to be primarily mediated by Y1 receptors, the pronounced phenotype of Y1Y2 receptor double KO mice suggests a synergistic role of Y2 receptors in fear acquisition and in fear extinction.


Asunto(s)
Condicionamiento Clásico , Discriminación en Psicología , Extinción Psicológica , Miedo , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Trastornos de Ansiedad/metabolismo , Conducta Animal , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptido Y/genética , Receptores de Neuropéptido Y/genética , Transducción de Señal
7.
Neuroscience ; 194: 208-19, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21840377

RESUMEN

Traumatic brain injury (TBI) is a risk factor for the development of epilepsy, which can occur months to years after the insult. The hippocampus is particularly vulnerable to the pathophysiological effects of TBI. Here, we determined whether there are long-term changes in inhibition in the dentate gyrus that could contribute to the progressive susceptibility to seizures after TBI. We used severe lateral-fluid percussion brain injury to induce TBI in rats. In this model, spontaneous seizure activity, which involves the hippocampus, appears after a long latent period, resembling the human condition. We demonstrate that synaptic GABA(A) receptor-mediated inhibition is profoundly reduced in ipsilateral dentate granule cells 1 month after TBI. Moreover, synaptic inhibition decreases over time, and by 6 months after TBI, it is also significantly decreased contralaterally. Progressive loss of synaptic inhibition is paralleled by a decline in the number of parvalbumin-positive interneurons, but, in contrast to status epilepticus models, GABA(A) receptor subunit expression is largely unaltered. At both time points, the magnitude of tonic GABA(A) receptor-mediated currents after TBI is maintained, indicating a preservation of the inhibitory constraint of granule cells through tonic inhibition. Our results extend the time window during which strategies to target epileptogenesis may be effective.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Giro Dentado/fisiopatología , Epilepsia del Lóbulo Temporal/fisiopatología , Interneuronas/fisiología , Inhibición Neural/fisiología , Receptores de GABA-A/fisiología , Animales , Lesiones Encefálicas/complicaciones , Giro Dentado/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epilepsia del Lóbulo Temporal/etiología , Epilepsia del Lóbulo Temporal/patología , Interneuronas/efectos de los fármacos , Interneuronas/patología , Masculino , Inhibición Neural/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos
8.
Neuroscience ; 189: 316-29, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21616128

RESUMEN

The subiculum is the major output area of the hippocampus. It is closely interconnected with the entorhinal cortex and other parahippocampal areas. In animal models of temporal lobe epilepsy (TLE) and in TLE patients it exerts increased network excitability and may crucially contribute to the propagation of limbic seizures. Using immunohistochemistry and in situ-hybridization we now investigated neuropathological changes affecting parvalbumin and calretinin containing neurons in the subiculum and other parahippocampal areas after kainic acid-induced status epilepticus. We observed prominent losses in parvalbumin containing interneurons in the subiculum and entorhinal cortex, and in the principal cell layers of the pre- and parasubiculum. Degeneration of parvalbumin-positive neurons was associated with significant precipitation of parvalbumin-immunoreactive debris 24 h after kainic acid injection. In the subiculum the superficial portion of the pyramidal cell layer was more severely affected than its deep part. In the entorhinal cortex, the deep layers were more severely affected than the superficial ones. The decrease in number of parvalbumin-positive neurons in the subiculum and entorhinal cortex correlated with the number of spontaneous seizures subsequently experienced by the rats. The loss of parvalbumin neurons thus may contribute to the development of spontaneous seizures. On the other hand, surviving parvalbumin neurons revealed markedly increased expression of parvalbumin mRNA notably in the pyramidal cell layer of the subiculum and in all layers of the entorhinal cortex. This indicates increased activity of these neurons aiming to compensate for the partial loss of this functionally important neuron population. Furthermore, calretinin-positive fibers terminating in the molecular layer of the subiculum, in sector CA1 of the hippocampus proper and in the entorhinal cortex degenerated together with their presumed perikarya in the thalamic nucleus reuniens. In addition, a significant loss of calretinin containing interneurons was observed in the subiculum. Notably, the loss in parvalbumin positive neurons in the subiculum equaled that in human TLE. It may result in marked impairment of feed-forward inhibition of the temporo-ammonic pathway and may significantly contribute to epileptogenesis. Similarly, the loss of calretinin-positive fiber tracts originating from the nucleus reuniens thalami significantly contributes to the rearrangement of neuronal circuitries in the subiculum and entorhinal cortex during epileptogenesis.


Asunto(s)
Hipocampo/patología , Interneuronas/metabolismo , Fibras Nerviosas/metabolismo , Parvalbúminas/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Convulsiones/patología , Núcleos Talámicos/patología , Animales , Calbindina 2 , Corteza Entorrinal/patología , Ácido Kaínico , Masculino , Parvalbúminas/genética , ARN Mensajero/metabolismo , Ratas , Convulsiones/inducido químicamente , Núcleos Talámicos/metabolismo
9.
Neuroscience ; 183: 71-80, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21458543

RESUMEN

Anxiety disorders are the most prevalent central nervous system diseases imposing a high social burden to our society. Emotional processing is particularly controlled by GABA-ergic transmission in the amygdala. Using in situ hybridization and immunohistochemistry we now investigated changes in the expression of GABA synthesizing enzymes (GAD65 and GAD67), GABA(A) (α1-5, ß1-3, γ1-2) and GABA(B) receptor subunits (GBBR1, GBBR2) in amygdaloid nuclei of high anxiety-related behavior (HAB) mice in comparison to mice selected for normal anxiety-related behavior (NAB). Levels of GAD65 and GAD67 mRNAs and protein, as well as those of GABA were increased in the amygdala of HAB mice. Relative to NAB controls, mRNA expression of the GABA(A) receptor subunits ß1, ß2 and γ2 was specifically increased in the basolateral amygdala of HAB mice while transcription of α5 and γ1 subunits was reduced in the central and medial amygdala. On the protein level, increases in ß2 and γ2 subunit immunoreactivities were evident in the basolateral amygdala of HAB mice. No change in GABA(B) receptor expression was observed. These findings point towards an imbalanced GABA-ergic neurotransmission in the amygdala of HAB mice. On the other hand, FosB, a marker for neuronal activity, was increased in principal neurons of the basolateral amygdala in HAB mice, reflecting activation of excitatory neurons, possibly as a consequence of reduced GABA-ergic tonic inhibition through α5 and γ1 containing receptors. Ultimately these mechanisms may lead to the compensatory activation of GABA transmission, as indicated by the increased expression of GAD65/67 in HAB mice.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Regulación de la Expresión Génica/fisiología , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Análisis de Varianza , Animales , Ansiedad/patología , Modelos Animales de Enfermedad , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Estadísticas no Paramétricas
10.
Neuroscience ; 176: 420-30, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21055451

RESUMEN

Neuropeptide Y (NPY) has been extensively studied in relation to anxiety and depression but of the seven NPY receptors known to date, it is not yet clear which one is mainly involved in mediating its effects in emotional behavior. Mice lacking the NPY-Y2 receptors were previously shown to be less anxious due to their improved ability to cope with stressful situations. In the present study, the behavioral phenotype including the response to challenges was analyzed in NPY-Y2 knockout (KO) mice backcrossed in to congenic C57BL/6 background. In the elevated plus-maze (EPM) and the forced swim test (FST), the anxiolytic-like or antidepressant-like phenotype of the NPY-Y2 KO mice could not be confirmed, although this study differs from the previous one only with regard to the genetic background of the mice. In addition, no differences in response to acute stress or to the antidepressant desipramine in the FST were detected between wild type (WT) and NPY-Y2 KO animals. These results suggest that the genetic background of the animals appears to have a strong influence on the behavioral phenotype of NPY-Y2 KO mice. Additionally, to further characterize the animals by their biochemical response to a challenge, the neurochemical changes induced by the anxiogenic compound yohimbine were measured in the medial prefrontal cortex (mPFC) of NPY-Y2 KO and compared to WT mice. Dopamine (DA) levels were significantly increased by yohimbine in the WT but unaffected in the KO mice, suggesting that NPY-Y2 receptor exerts a direct control over both the tonic and phasic release of DA and that, although the anxiety-like behavior of these NPY-Y2 KO mice is unaltered, there are clear modifications of DA dynamics. However, yohimbine led to a significant increase in noradrenaline (NA) concentration and a slight reduction in serotonin concentration that were identical for both phenotypes.


Asunto(s)
Ansiedad/genética , Conducta Animal/fisiología , Encéfalo/metabolismo , Receptores de Neuropéptido Y/genética , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Ansiedad/metabolismo , Encéfalo/efectos de los fármacos , Química Encefálica , Dopamina/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Norepinefrina/metabolismo , Fenotipo , Receptores de Neuropéptido Y/deficiencia , Receptores de Neuropéptido Y/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Yohimbina/farmacología
11.
Gene Ther ; 17(5): 643-52, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20220782

RESUMEN

Neuropeptide Y (NPY) is an endogenous peptide with powerful anticonvulsant properties. Its overexpression in the rat hippocampus, mediated by the local application of recombinant adeno-associated viral (rAAV) vectors carrying the human NPY gene, results in significant reduction of seizures in acute and chronic seizure models. In this study, we characterized a more efficient rAAV-NPY vector to improve cell transfection in the injected area. The changes included pseudotyping with the AAV vector serotype 1 (rAAV1), and using the strong constitutive hybrid CBA promoter, which contains a cytomegalovirus enhancer and chicken beta-actin promoter sequences. We compared NPY expression and the associated anticonvulsant effects of this new vector, with those mediated by the former rAAV vector with chimeric serotype 1/2 (rAAV1/2). In addition, we investigated whether rAAV serotype 1 vector-mediated chronic NPY overexpression causes behavioural deficits that may detract from the clinical utility of this therapeutic approach. We report that rAAV-NPY serotype 1 vector has significantly improved anticonvulsant activity when compared with serotype 1/2 vector, as assessed by measuring EEG seizure activity in kainic acid treated rats. rAAV1-mediated NPY overexpression in naive rats did not result in alterations of physiological functions such as learning and memory, anxiety and locomotor activity. In addition, we did not observe glia activation, or humoral immune responses against serotype 1 vector, which could inactivate gene expression. Our findings show that rAAV1-NPY vector with the CBA promoter mediates powerful anticonvulsant effects and seems to be safe in rodents, thus it may be considered a vector of choice for possible clinical applications.


Asunto(s)
Epilepsia del Lóbulo Temporal/terapia , Terapia Genética/métodos , Hipocampo/metabolismo , Neuropéptido Y/genética , Convulsiones/terapia , Transducción Genética/métodos , Actinas/genética , Animales , Dependovirus , Epilepsia del Lóbulo Temporal/fisiopatología , Vectores Genéticos , Inmunidad Humoral , Ácido Kaínico/efectos adversos , Aprendizaje , Masculino , Memoria , Actividad Motora , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Convulsiones/fisiopatología
12.
J Psychopharmacol ; 24(10): 1541-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19351805

RESUMEN

Neuropeptide-Y acting through Y1 receptors reduces anxiety and stress sensitivity in rodents. In Y1 receptor knockout (Y1⁻/⁻) mice, however, anxiety-related behaviour is altered only in a context-dependent manner. Here, we investigated whether stress causes a delayed change in the emotional-affective behaviour of female Y1⁻/⁻ mice. Locomotor and anxiety-related behaviour was assessed with the elevated plus-maze (EPM) test and depression-like behaviour with the forced swim test (FST). These behavioural tests were also used as experimental stress paradigms. Locomotion and anxiety-like behaviour did not differ between naïve control and Y1⁻/⁻ mice. One week after the FST, locomotion was reduced in control animals but unchanged in Y1⁻/⁻ mice, whereas anxiety-like behaviour remained unaltered in both genotypes. Depression-like behaviour (immobility) was identical in naïve control and Y1⁻/⁻ mice but, 1 week after the EPM test, was attenuated in Y1⁻/⁻ mice relative to control animals. Our data show that naïve female Y1⁻/⁻ mice do not grossly differ from female control animals in their locomotor and depression-like behaviour. Exposure to the stress associated with behavioural testing, however, leads to delayed genotype-dependent differences in locomotion and depression-like behaviour. These findings attest to a role of Y1 receptor signalling in the control of stress coping and/or adaptation.


Asunto(s)
Depresión/fisiopatología , Actividad Motora , Neuropéptido Y/fisiología , Receptores de Neuropéptido Y/fisiología , Transducción de Señal , Estrés Fisiológico , Estrés Psicológico , Analgesia , Animales , Ansiedad , Conducta Animal , Corticosterona/sangre , Depresión/sangre , Depresión/psicología , Femenino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Dimensión del Dolor , Tiempo de Reacción , Receptores de Neuropéptido Y/genética , Estrés Psicológico/sangre , Factores de Tiempo
13.
Neuroscience ; 158(4): 1717-30, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19121371

RESUMEN

There is growing evidence that neuropeptide Y (NPY) acting through Y1 and Y2 receptors has a prominent role in modulating anxiety- and depression-like behavior in rodents. However, a role of other Y-receptors like that of Y4 receptors in this process is poorly understood. We now investigated male Y2, Y4 single and Y2/Y4 double knockout mice in behavioral paradigms for changes in motor activity, anxiety and depression-like behavior. Motor activity was increased in Y2, Y4 and Y2/Y4 knockout mice under changing and stressful conditions, but not altered in a familiar environment. Y4 and Y2 knockout mice revealed an anxiolytic phenotype in the light/dark test, marble burying test and in stress-induced hyperthermia, and reduced depression-like behavior in the forced swim and tail suspension tests. In Y2/Y4 double knockout mice, the response in the light/dark test and in the forced swim test was further enhanced compared with Y4 and Y2 knockout mice, respectively. High levels of Y4 binding sites were observed in brain stem nuclei including nucleus of solitary tract and area postrema. Lower levels were found in the medial amygdala and hypothalamus. Peripheral administration of pancreatic polypeptide (PP) induced Y4 receptor-dependent c-Fos expression in brain stem, hypothalamus and amygdala. PP released peripherally from the pancreas in response to food intake, may act not only as a satiety signal but also modulate anxiety-related locomotion.


Asunto(s)
Depresión/genética , Depresión/fisiopatología , Conducta Exploratoria/fisiología , Actividad Motora/genética , Receptores de Neuropéptido Y/deficiencia , Amígdala del Cerebelo/metabolismo , Animales , Autorradiografía/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Suspensión Trasera/métodos , Hipertermia Inducida , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polipéptido Pancreático/farmacología , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología , Natación/fisiología
14.
Neuroscience ; 136(4): 1097-107, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16216428

RESUMEN

Vagal afferents signal gastric acid challenge to the nucleus tractus solitarii of the rat brainstem. This study investigated whether nucleus tractus solitarii neurons in the mouse also respond to gastric acid challenge and whether this chemonociceptive input is modified by neuropeptide Y acting via neuropeptide Y receptors of type Y2 or Y4. The gastric mucosa of female mice was exposed to different concentrations of HCl or saline, excitation of neurons in the nucleus tractus solitarii visualized by c-Fos immunohistochemistry, gastric emptying deduced from the gastric volume recovery, and gastric lesion formation evaluated by planimetry. Relative to saline, intragastric HCl (0.15-0.35 M) increased the number of c-Fos-expressing cells in the nucleus tractus solitarii in a concentration-dependent manner, inhibited gastric emptying but failed to cause significant hemorrhagic injury in the stomach. Mice in which the Y2 or Y4 receptor gene had been deleted responded to gastric acid challenge with a significantly higher expression of c-Fos in the nucleus tractus solitarii, the increases amounting to 39 and 31%, respectively. The HCl-induced inhibition of gastric emptying was not altered by deletion of the Y2 or Y4 receptor gene. BIIE0246 ((S)-N2-[[1-[2-[4-[(R,S)-5,11-dihydro-6(6H)-oxodibenz[b,e] azepin-11-yl]-1-piperazinyl]-2-oxoethyl]cyclopentyl] acetyl]-N-[2-[1,2-dihydro-3,5 (4H)-dioxo-1,2-diphenyl-3H-1,2,4-triazol-4-yl]ethyl]-argininamide; 0.03 mmol/kg s.c.), a Y2 receptor antagonist which does not cross the blood-brain barrier, did not modify the c-Fos response to gastric acid challenge. The Y2 receptor agonist peptide YY-(3-36) (0.1 mg/kg intraperitoneally) likewise failed to alter the gastric HCl-evoked expression of c-Fos in the nucleus tractus solitarii. BIIE0246, however, prevented the effect of peptide YY-(3-36) to inhibit gastric acid secretion as deduced from measurement of intragastric pH. The current data indicate that gastric challenge with acid concentrations that do not induce overt injury but inhibit gastric emptying is signaled to the mouse nucleus tractus solitarii. Endogenous neuropeptide Y acting via Y2 and Y4 receptors depresses the afferent input to the nucleus tractus solitarii by a presumably central site of action.


Asunto(s)
Tronco Encefálico/fisiología , Ácido Gástrico/fisiología , Mucosa Gástrica/inervación , Neuropéptido Y/fisiología , Receptores de Neuropéptido Y/fisiología , Análisis de Varianza , Animales , Arginina/análogos & derivados , Arginina/farmacología , Benzazepinas/farmacología , Tronco Encefálico/citología , Recuento de Células , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Transportador 3 de Aminoácidos Excitadores/metabolismo , Femenino , Vaciamiento Gástrico , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Ácido Clorhídrico/farmacología , Concentración de Iones de Hidrógeno , Inmunohistoquímica/métodos , Ratones , Ratones Noqueados , Neuronas/fisiología , Fragmentos de Péptidos , Péptido YY/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/deficiencia
15.
Neuroscience ; 134(2): 691-704, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15951123

RESUMEN

Epilepsy may result from altered transmission of the principal inhibitory transmitter GABA in the brain. Using in situ hybridization in two animal models of epileptogenesis, we investigated changes in the expression of nine major GABA(A) receptor subunits (alpha1, alpha2, alpha4, alpha5, beta1-beta3, gamma2 and delta) and of the GABA(B) receptor species GABA(B)R1a, GABA(B)R1b and GABA(B)R2 in 1) hippocampal kindling and 2) epilepsy following electrically-induced status epilepticus (SE). Hippocampal kindling triggers a decrease in seizure threshold without producing spontaneous seizures and hippocampal damage, whereas the SE model is characterized by spontaneous seizures and hippocampal damage. Changes in the expression of GABA(A) and GABA(B) receptor mRNAs were observed in both models, and compared with those seen in other models and in human temporal lobe epilepsy. The most prominent changes were a relatively fast (24 h after kindling and electrically-induced SE) and lasting (7 and 30 days after termination of kindling and SE, respectively) reduction of GABA(A) receptor subunit delta mRNA levels (by 43-78%) in dentate granule cells, accompanied by increases in mRNA levels of all three beta-subunits (by 8-79%) and subunit gamma2 (by 11-43%). Levels of the minor subunit alpha4 were increased by up to 60% in dentate granule cells in both animal models, whereas those of subunit alpha5 were decreased 24 h and 30 days after SE, but not after kindling. In cornu ammonis 3 pyramidal cells, downregulation of subunits alpha2, alpha4, alpha5, and beta1-3 was observed in the ventral hippocampus and of alpha2, alpha5, beta3 and gamma2 in its dorsal extension 24 h after SE. Similar but less pronounced changes were seen in sector cornu ammonis 1. Persistent decreases in subunit alpha2, alpha4 and beta2 transcript levels were presumably related to SE-induced cell loss. GABA(B) receptor expression was characterized by increases in GABA(B)R2 mRNA levels at all intervals after kindling and SE. The observed changes suggest substantial and cell specific rearrangement of GABA receptors. Lasting downregulation of subunits delta and alpha5 in granule cells and transient decreases in subunit alpha2 and beta1-3 mRNA levels in cornu ammonis 3 pyramidal cells are suggestive of impaired GABA(A) receptor-mediated inhibition. Persistent upregulation of subunits beta1-3 and gamma2 of the GABA(A) receptor and of GABA(B)R2 mRNA in granule cells, however, may result in activation of compensatory anticonvulsant mechanisms.


Asunto(s)
Hipocampo/fisiopatología , Excitación Neurológica/fisiología , ARN Mensajero/genética , Receptores de GABA-A/genética , Receptores de GABA-B/genética , Estado Epiléptico/genética , Animales , Secuencia de Bases , Sondas de ADN , Electrochoque , Hibridación in Situ , Excitación Neurológica/patología , Datos de Secuencia Molecular , Subunidades de Proteína/genética , Ratas , Ratas Sprague-Dawley , Convulsiones/clasificación , Convulsiones/fisiopatología , Estado Epiléptico/etiología
16.
Curr Top Med Chem ; 2(8): 795-816, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12171572

RESUMEN

GABA(A) receptors are the major inhibitory neurotransmitter receptors in the brain and are the site of action of many clinically important drugs. These receptors are composed of five subunits that can belong to eight different subunit classes. Depending on their subunit composition, these receptors exhibit distinct pharmacological and electrophysiological properties. Recent studies on recombinant and native GABA(A) receptors suggest the existence of far more receptor subtypes than previously assumed. Thus, receptors composed of one, two, three, four, or five different subunits might exist in the brain. Studies on the regional, cellular and subcellular distribution of GABA(A) receptor subunits, and on the co-localization of these subunits at the light and electron microscopic level for the first time provide information on the distribution of GABA(A) receptor subtypes in the brain. These studies will have to be complemented by electrophysiological and pharmacological studies on the respective recombinant and native receptors to finally identify the receptor subtypes present in the brain. The distinct cellular and subcellular location of individual receptor subtypes suggests that they exhibit specific functions in the brain that can be selectively modulated by subtype specific drugs. This conclusion is supported by the recent demonstration that different GABA(A) receptor subtypes mediate different effects of benzodiazepines. Together, these results should cause a revival of GABA(A) receptor research and strongly stimulate the development of drugs with a higher selectivity for alpha2-, alpha3-, or alpha5-subunit-containing receptor subtypes. Such drugs might exhibit quite selective clinical effects.


Asunto(s)
Receptores de GABA-A , Animales , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Especificidad de Órganos , Subunidades de Proteína , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Receptores de GABA-A/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
18.
Neuroscience ; 110(2): 237-43, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11958866

RESUMEN

Functional studies in epileptic tissue indicate that neuropeptide Y and some of its peptide analogs potently inhibit seizure activity. We investigated seizure susceptibility in transgenic rats overexpressing the rat neuropeptide Y gene under the control of its natural promoter. Seizures were induced in adult transgenic male rats and their wild-type littermates by i.c.v. injection of 0.3 microg kainic acid or by electrical kindling of the dorsal hippocampus. Transgenic rats showed a significant reduction in the number and duration of electroencephalographic seizures induced by kainate by 30% and 55% respectively (P<0.05 and 0.01). Transgenic rats were also less susceptible to epileptogenesis than wild-type littermates as demonstrated by a 65% increase in the number of electrical stimuli required to induce stage 5 seizures (P<0.01). This phenotype was associated with a strong and specific expression of neuropeptide Y mRNA in area CA1, a brain area involved in the seizure network. We conclude that endogenous neuropeptide Y overexpression in the rat hippocampus is associated with inhibition of seizures and epileptogenesis suggesting that this system may be a valuable target for developing novel antiepileptic treatments.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia/genética , Predisposición Genética a la Enfermedad/genética , Hipocampo/metabolismo , Neuronas/metabolismo , Neuropéptido Y/genética , Regulación hacia Arriba/genética , Animales , Animales Modificados Genéticamente , Estimulación Eléctrica , Electroencefalografía/efectos de los fármacos , Epilepsia/inducido químicamente , Epilepsia/fisiopatología , Epilepsia del Lóbulo Temporal/fisiopatología , Agonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Excitación Neurológica/efectos de los fármacos , Excitación Neurológica/genética , Masculino , Neuronas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Brain Res Mol Brain Res ; 97(1): 1-6, 2001 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-11744156

RESUMEN

Ingestion of trimethyltin (TMT) produces mental confusion and temporal lobe seizures in humans. In rats, it causes increased seizure susceptibility, hyperactivity, aggression, learning impairment, and neuronal loss especially of hippocampal CA3c pyramidal cells and in the piriform cortex. As some of these symptoms may be due to impaired inhibitory neurotransmission, mRNA levels of the nine major GABA(A) receptor subunits, of GABA(B) receptors 1 and 2, and the 65- and 67-kD glutamate decarboxylase (GAD) variants were investigated by in situ hybridization 2, 5, and 16 days after TMT administration. GAD-65 mRNA levels were enhanced in hippocampal interneurons by up to 46% 5 days after TMT application, suggesting increased activity of respective neurons. In the granule cell layer, only the GABA(A) receptor subunit delta mRNA was altered (decreased by 48%). In the hippocampal sector CA3c and in the piriform cortex, mRNA levels of GABA(A) receptor subunits alpha1, alpha5, beta1, beta2, beta3, gamma2 and of both GABA(B) receptors declined (by 46-72%) after 5-16 days, being consistent with the extensive cell loss. In contrast, subunit alpha2 mRNA levels decreased already after 2 days at an extent exceeding the cell loss in CA3. Subunit alpha4 mRNA levels increased (about two-fold) in surviving CA3 neurons. In sector CA1, mRNA levels of subunits alpha1, alpha5, beta2, beta3, and gamma2 decreased by 35-54% in spite of only a minor (9%) cell loss. The data indicate neurodegeneration related decreases in mRNA levels in sector CA3 and piriform cortex, whereas decreases in sector CA1 may be a consequence of impaired excitatory input to this area.


Asunto(s)
Agresión/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glutamato Descarboxilasa/biosíntesis , Hipocampo/efectos de los fármacos , Hipercinesia/inducido químicamente , Isoenzimas/biosíntesis , Degeneración Nerviosa/inducido químicamente , Proteínas del Tejido Nervioso/biosíntesis , Neurotoxinas/farmacología , Receptores de GABA-A/biosíntesis , Compuestos de Trimetilestaño/farmacología , Ácido gamma-Aminobutírico/fisiología , Animales , Muerte Celular , Inducción Enzimática/efectos de los fármacos , Glutamato Descarboxilasa/genética , Hipocampo/patología , Hipercinesia/metabolismo , Hipercinesia/patología , Hibridación in Situ , Isoenzimas/genética , Masculino , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Neurotoxinas/toxicidad , Subunidades de Proteína , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/genética , Receptores de GABA-B/biosíntesis , Receptores de GABA-B/genética , Compuestos de Trimetilestaño/toxicidad , Regulación hacia Arriba/efectos de los fármacos
20.
Ann Neurol ; 50(2): 216-26, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11506405

RESUMEN

Chromogranins are polypeptides which are widely expressed in the central nervous system. They are stored in dense core vesicles of nerve terminals, from where they are released upon stimulation. Using immunocytochemistry, we investigated the distribution of chromogranin A, chromogranin B, secretoneurin, and, for comparison, dynorphin in hippocampal specimens removed at routine surgery from patients with drug-resistant mesial temporal lobe epilepsy and in autopsy tissues from nonneurologically deceased subjects. In post mortem controls (n = 21), immunoreactivity for all 4 peptides (most prominently for chromogranin B and dynorphin) was observed in the terminal field of mossy fibers. For chromogranins, staining was observed also in sectors CA1 to CA3 and in the subiculum. Chromogranin B immunoreactivity was found in the inner molecular layer of the dentate gyrus, the area of terminating associational-commissural fibers. Secretoneurin and dynorphin immunoreactivity labeled the outer molecular layer and the stratum lacunosum moleculare of sectors CA1 to CA3, where projections from the entorhinal cortex terminate. In specimens with Ammon's horn sclerosis (n = 25), staining for all 3 chromogranins and for dynorphin was reduced in the hilus of the dentate gyrus. Instead, intense staining was observed in the inner molecular layer, presumably delineating terminals of sprouted mossy fibers. Specimens obtained from temporal lobe epilepsy patients without Ammon's horn sclerosis (n = 4) lacked this pronounced rearrangement of mossy fibers. In the stratum lacunosum moleculare of sector CA1, secretoneurin and dynorphin immunoreactivity was reduced in sclerotic, but not in nonsclerotic, specimens, paralleling the partial loss of fibers arising from the entorhinal cortex. Instead, presumably sprouted secretoneurin-immunoreactive fibers were found in the outer dentate molecular layer in sclerotic specimens. These changes in staining patterns for chromogranins and dynorphin mark profound plastic and functional rearrangement of hippocampal circuitry in temporal lobe epilepsy.


Asunto(s)
Biomarcadores/análisis , Cromograninas/análisis , Epilepsia del Lóbulo Temporal/patología , Hipocampo/patología , Adulto , Dinorfinas/análisis , Femenino , Humanos , Inmunohistoquímica , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...