Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Pharmacol Rep ; 71(5): 870-878, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31408786

RESUMEN

BACKGROUND: Several studies strongly support the role of the dopamine D2-like and glutamate mGlu5 receptors in psychostimulant reward and relapse. METHODS: The present study employed cocaine or MDMA self-administration with yoked-triad procedure in rats to explore whether extinction training affects the drug-seeking behavior and the D2-like and mGlu5 receptor Bmax and Kd values in several regions of the animal brain. RESULTS: Both cocaine and MDMA rats developed maintenance of self-administration, but MDMA evoked lower response rates and speed of self-administration acquisition. During reinstatement tests, cocaine or MDMA seeking behavior was produced by either exposure to the drug-associated cues or drug-priming injections. The extinction training after cocaine self-administration did not alter significantly D2-like receptor expression the in the limbic and subcortical brain areas, while MDMA yoked rats showed a decrease of the D2-like binding density in the nucleus accumbens and increase in the hippocampus and a rise of affinity in the striatum and hippocampus. Interestingly, in the prefrontal cortex a reduction in the mGlu5 receptor density in cocaine- or MDMA-abstinent rats was demonstrated, with significant effects being observed after previous MDMA exposure. Moreover, rats self-administered cocaine showed a rise in the density of mGlu5 receptor for the nucleus accumbens. CONCLUSION: This study first time shows that abstinence followed extinction training after cocaine or MDMA self- or passive-injections changes the D2-like and mGlu5 density and affinity. The observed changes in the expression of both receptors are brain-region specific and related to either pharmacological and/or motivational features of cocaine or MDMA.


Asunto(s)
Encéfalo/efectos de los fármacos , Cocaína/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Extinción Psicológica , N-Metil-3,4-metilenodioxianfetamina/farmacología , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Dopamina D2/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Cocaína/administración & dosificación , Masculino , N-Metil-3,4-metilenodioxianfetamina/administración & dosificación , Ratas Wistar , Esquema de Refuerzo , Autoadministración
3.
J Alzheimers Dis ; 61(1): 389-399, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29154284

RESUMEN

BACKGROUND: Based on the pivotal role of astrocytes in brain homeostasis and the strong metabolic cooperation existing between neurons and astrocytes, it has been suggested that astrocytic dysfunctions might cause and/or contribute to neuroinflammation and neurodegenerative processes. Therapeutic approaches aimed at both neuroprotection and neuroinflammation reduction may prove particularly effective in slowing the progression of these diseases. The endogenous lipid mediator palmitoylethanolamide (PEA) displayed neuroprotective and anti(neuro)inflammatory properties, and demonstrated interesting potential as a novel treatment for Alzheimer's disease. OBJECTIVE AND METHODS: We firstly evaluated whether astrocytes could participate in regulating the Aß42-induced neuronal damage, by using primary mouse astrocytes cell cultures and mixed astrocytes-neurons cultures. Furthermore, the possible protective effects of PEA against Aß42-induced neuronal toxicity have also been investigated by evaluating neuronal viability, apoptosis, and morphometric parameters. RESULTS: The presence of astrocytes pre-exposed to Aß42 (0.5µM; 24 h) induced a reduction of neuronal viability in primary mouse astrocytes-neurons co-cultures. Furthermore, under these experimental conditions, an increase in the number of neuronal apoptotic nuclei and a decrease in the number of MAP-2 positive neurons were observed. Finally, astrocytic Aß42 pre-exposure induced an increase in the number of neurite aggregations/100µm as compared to control (i.e., untreated) astrocytes-neurons co-cultures. These effects were not observed in neurons cultured in the presence of astrocytes pre-exposed to PEA (0.1µM), applied 1 h before and maintained during Aß42 treatment. CONCLUSION: Astrocytes contribute to Aß42-induced neurotoxicity and PEA, by blunting Aß42-induced astrocyte activation, improved neuronal survival in mouse astrocyte-neuron co-cultures.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Antiinflamatorios no Esteroideos/farmacología , Astrocitos/efectos de los fármacos , Etanolaminas/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ácidos Palmíticos/farmacología , Fragmentos de Péptidos/farmacología , Amidas , Análisis de Varianza , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Técnicas de Cocultivo , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Factores de Tiempo
4.
Cell Signal ; 40: 116-124, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28923416

RESUMEN

The effects of nanomolar cocaine concentrations, possibly not blocking the dopamine transporter activity, on striatal D2-σ1 heteroreceptor complexes and their inhibitory signaling over Gi/o, have been tested in rat striatal synaptosomes and HEK293T cells. Furthermore, the possible role of σ1 receptors (σ1Rs) in the cocaine-provoked amplification of D2 receptor (D2R)-induced reduction of K+-evoked [3H]-DA and glutamate release from rat striatal synaptosomes, has also been investigated. The dopamine D2-likeR agonist quinpirole (10nM-1µM), concentration-dependently reduced K+-evoked [3H]-DA and glutamate release from rat striatal synaptosomes. The σ1R antagonist BD1063 (100nM), amplified the effects of quinpirole (10 and 100nM) on K+-evoked [3H]-DA, but not glutamate, release. Nanomolar cocaine concentrations significantly enhanced the quinpirole (100nM)-induced decrease of K+-evoked [3H]-DA and glutamate release from rat striatal synaptosomes. In the presence of BD1063 (10nM), cocaine failed to amplify the quinpirole (100nM)-induced effects. In cotransfected σ1R and D2LR HEK293T cells, quinpirole had a reduced potency to inhibit the CREB signal versus D2LR singly transfected cells. In the presence of cocaine (100nM), the potency of quinpirole to inhibit the CREB signal was restored. In D2L singly transfected cells cocaine (100nM and 10µM) exerted no modulatory effects on the inhibitory potency of quinpirole to bring down the CREB signal. These results led us to hypothesize the existence of functional D2-σ1R complexes on the rat striatal DA and glutamate nerve terminals and functional D2-σ1R-DA transporter complexes on the striatal DA terminals. Nanomolar cocaine concentrations appear to alter the allosteric receptor-receptor interactions in such complexes leading to enhancement of Gi/o mediated D2R signaling.


Asunto(s)
Cocaína/administración & dosificación , Cuerpo Estriado/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores sigma/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Dopamina/metabolismo , Agonistas de Dopamina/administración & dosificación , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Complejos Multiproteicos/efectos de los fármacos , Complejos Multiproteicos/genética , Terminaciones Nerviosas/efectos de los fármacos , Terminaciones Nerviosas/metabolismo , Quinpirol/administración & dosificación , Ratas , Receptores de Dopamina D2/química , Receptores de Dopamina D2/genética , Receptores sigma/química , Receptores sigma/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Receptor Sigma-1
5.
Neurobiol Learn Mem ; 139: 135-143, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28104530

RESUMEN

The long-lasting effects of gestational cannabinoids exposure on the adult brain of the offspring are still controversial. It has already been shown that pre- or perinatal cannabinoids exposure induces learning and memory disruption in rat adult offspring, associated with permanent alterations of cortical glutamatergic neurotransmission and cognitive deficits. In the present study, the risk of long-term consequences induced by perinatal exposure to cannabinoids on rat hippocampal GABAergic system of the offspring, has been explored. To this purpose, pregnant rats were treated daily with Delta9-tetrahydrocannabinol (Δ9-THC; 5mg/kg) or its vehicle. Perinatal exposure to Δ9-THC induced a significant reduction (p<0.05) in basal and K+-evoked [3H]-GABA outflow of 90-day-old rat hippocampal slices. These effects were associated with a reduction of hippocampal [3H]-GABA uptake compared to vehicle exposed group. Perinatal exposure to Δ9-THC induced a significant reduction of CB1 receptor binding (Bmax) in the hippocampus of 90-day-old rats. However, a pharmacological challenge with either Δ9-THC (0.1µM) or WIN55,212-2 (2µM), similarly reduced K+-evoked [3H]-GABA outflow in both experimental groups. These reductions were significantly blocked by adding the selective CB1 receptor antagonist SR141716A. These findings suggest that maternal exposure to cannabinoids induces long-term alterations of hippocampal GABAergic system. Interestingly, previous behavioral studies demonstrated that, under the same experimental conditions as in the present study, perinatal cannabinoids exposure induced cognitive impairments in adult rats, thus resembling some effects observed in humans. Although it is difficult and sometimes misleading to extrapolate findings obtained from animal models to humans, the possibility that an alteration of hippocampus aminoacidergic transmission might underlie, at least in part, some of the cognitive deficits affecting the offspring of marijuana users, is supported.


Asunto(s)
Dronabinol/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Hipocampo/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptor Cannabinoide CB1/metabolismo , Transmisión Sináptica/efectos de los fármacos , Animales , Benzoxazinas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Femenino , Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Masculino , Morfolinas/farmacología , Naftalenos/farmacología , Embarazo , Unión Proteica/efectos de los fármacos , Ratas , Transmisión Sináptica/fisiología
6.
J Neurochem ; 138(2): 254-64, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27127992

RESUMEN

In this study, the functional role of individual striatal receptors for adenosine (A2AR), dopamine (D2R), and the metabotropic glutamate receptor mGlu5R in regulating rat basal ganglia activity was characterized in vivo using dual-probe microdialysis in freely moving rats. In particular, intrastriatal perfusion with the D2R agonist quinpirole (10 µM, 60 min) decreased ipsilateral pallidal GABA and glutamate levels, whereas intrastriatal CGS21680 (A2AR agonist; 1 µM, 60 min) was ineffective on either pallidal GABA and glutamate levels or the quinpirole-induced effects. Intrastriatal perfusion with the mGlu5R agonist (RS)-2-chloro-5-hydroxyphenylglycine (600 µM, 60 min), by itself ineffective on pallidal GABA and glutamate levels, partially counteracted the effects of quinpirole. When combined with CGS21680 (1 µM, 60 min), (RS)-2-chloro-5-hydroxyphenylglycine (CHPG; 600 µM, 60 min) fully counteracted the quinpirole (10 µM, 60 min)-induced reduction in ipsilateral pallidal GABA and glutamate levels. These effects were fully counteracted by local perfusion with the mGlu5R antagonist MPEP (300 µM) or the A2AR antagonist ZM 241385 (100 nM). These results suggest that A2ARs and mGlu5Rs interact synergistically in modulating the D2R-mediated control of striatopallidal GABA neurons. Using dual-probe microdialysis, we characterized the functional role of striatal adenosine A2A receptor (A2AR), dopamine D2 receptor (D2R), and metabotropic glutamate receptor 5 (mGluR5) interactions in regulating rat basal ganglia activity. The results suggest the possible usefulness of using an A2AR antagonist and mGluR5 antagonist combination in the treatment of Parkinson's disease to increase the inhibitory D2 signaling on striatopallidal GABA neurons.


Asunto(s)
Cuerpo Estriado/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Dopamina D2/metabolismo , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Dopamina/farmacología , Agonistas de Dopamina/farmacología , Ácido Glutámico/farmacología , Masculino , Microdiálisis/métodos , Neostriado/metabolismo , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5/efectos de los fármacos , Receptores de Dopamina D2/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
7.
J Alzheimers Dis ; 46(2): 407-21, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25765918

RESUMEN

BACKGROUND: Considering the heterogeneity of pathological changes occurring in Alzheimer's disease (AD), a therapeutic approach aimed both to neuroprotection and to neuroinflammation reduction may prove effective. Palmitoylethanolamide (PEA) has attracted attention for its anti-inflammatory/neuroprotective properties observed in AD animal models. OBJECTIVE AND METHODS: We evaluated the protective role of PEA against amyloid-ß42 (Aß42) toxicity on cell viability and glutamatergic transmission in primary cultures of cerebral cortex neurons and astrocytes from the triple-transgenic murine model of AD (3xTg-AD) and their wild-type littermates (non-Tg) mice. RESULTS: Aß42 (0.5 µM; 24 h) affects the cell viability in cultured cortical neurons and astrocytes from non-Tg mice, but not in those from 3xTg-AD mice. These effects were counteracted by the pretreatment with PEA (0.1 µM). Basal glutamate levels in cultured neurons and astrocytes from 3xTg-AD mice were lower than those observed in cultured cells from non-Tg mice. Aß42-exposure reduced and increased glutamate levels in non-Tg mouse cortical neurons and astrocytes, respectively. These effects were counteracted by the pretreatment with PEA. By itself, PEA did not affect cell viability and glutamate levels in cultured cortical neurons and astrocytes from non-Tg or 3xTg-AD mice. CONCLUSION: The exposure to Aß42 induced toxic effects on cultured cortical neurons and astrocytes from non-Tg mice, but not in those from 3xTg-AD mice. Furthermore, PEA exerts differential effects against Aß42-induced toxicity in primary cultures of cortical neurons and astrocytes from non-Tg and 3xTg-AD mice. In particular, PEA displays protective properties in non-Tg but not in 3xTg-AD mouse neuronal cultured cells overexpressing Aß.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Astrocitos/efectos de los fármacos , Etanolaminas/farmacología , Neuronas/efectos de los fármacos , Ácidos Palmíticos/farmacología , Amidas , Animales , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Ácido Glutámico/sangre , Ratones , Ratones Transgénicos , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA