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












Base de datos
Intervalo de año de publicación
1.
Eur J Neurosci ; 46(4): 2015-2025, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28677227

RESUMEN

Deficits in neuronal network synchrony in hippocampus and prefrontal cortex have been widely demonstrated in disorders of cognitive dysfunction, including schizophrenia and Alzheimer's disease. The atypical dopamine agonist SKF 83959 has been shown to increase brain-derived neurotrophic factor signalling and suppress activity of glycogen synthase kinase-3 in PFC, two processes important to learning and memory. The purpose of this study was to therefore evaluate the impact of SKF 83959 on oscillatory deficits in methylazoxymethanol acetate (MAM) rat model of schizophrenia. To achieve this, local field potentials were recorded simultaneously from the hippocampus and prefrontal cortex of anesthetized rats at 15 and 90 min following both acute and repeated administration of SKF 83959 (0.4 mg/kg). In MAM rats, but not controls, repeated SKF 83959 treatment increased signal amplitude in hippocampus and enhanced the spectral power of low frequency delta and theta oscillations in this region. In PFC, SKF 83959 increased delta, theta and gamma spectral power. Increased HIP-PFC theta coherence was also evident following acute and repeated SKF 83959. In apparent contradiction to these oscillatory effects, in MAM rats, SKF 83959 inhibited spatial learning and induced a significant increase in thigmotactic behaviour. These findings have uncovered a previously unknown role for SKF 83959 in the positive regulation of hippocampal-prefrontal cortical oscillatory network activity. As SKF 83959 is known to have affinity for a number of receptors, delineating the receptor mechanisms that mediate the positive drug effects on neuronal oscillations could have significant future implications in disorders associated with cognitive dysfunction.


Asunto(s)
2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , Disfunción Cognitiva/tratamiento farmacológico , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Hipocampo/efectos de los fármacos , Red Nerviosa/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/uso terapéutico , Animales , Disfunción Cognitiva/fisiopatología , Agonistas de Dopamina/uso terapéutico , Hipocampo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Red Nerviosa/fisiología , Corteza Prefrontal/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Dopaminérgicos/fisiología
2.
Front Pharmacol ; 8: 924, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29354053

RESUMEN

A significant subpopulation of neurons in rat nucleus accumbens (NAc) coexpress dopamine D1 and D2 receptors, which can form a D1-D2 receptor complex, but their relevance in addiction is not known. The existence of the D1-D2 heteromer in the striatum of rat and monkey was established using in situ PLA, in situ FRET and co-immunoprecipitation. In rat, D1-D2 receptor heteromer activation led to place aversion and abolished cocaine CPP and locomotor sensitization, cocaine intravenous self-administration and reinstatement of cocaine seeking, as well as inhibited sucrose preference and abolished the motivation to seek palatable food. Selective disruption of this heteromer by a specific interfering peptide induced reward-like effects and enhanced the above cocaine-induced effects, including at a subthreshold dose of cocaine. The D1-D2 heteromer activated Cdk5/Thr75-DARPP-32 and attenuated cocaine-induced pERK and ΔFosB accumulation, together with inhibition of cocaine-enhanced local field potentials in NAc, blocking thus the signaling pathway activated by cocaine: D1R/cAMP/PKA/Thr34-DARPP-32/pERK with ΔFosB accumulation. In conclusion, our results show that the D1-D2 heteromer exerted tonic inhibitory control of basal natural and cocaine reward, and therefore initiates a fundamental physiologic function that limits the liability to develop cocaine addiction.

3.
Front Aging Neurosci ; 9: 434, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29375364

RESUMEN

Glycogen synthase kinase-3 (GSK-3) plays a critical role in cognitive dysfunction associated with Alzheimer's disease (AD), yet the mechanism by which GSK-3 alters cognitive processes in other disorders, such as schizophrenia, remains unknown. In the present study, we demonstrated a role for GSK-3 in the direct regulation of neuronal oscillations in hippocampus (HIP) and prelimbic cortex (PL). A comparison of the GSK-3 inhibitors SB 216763 and lithium demonstrated disparate effects of the drugs on spatial memory and neural oscillatory activity in HIP and PL. SB 216763 administration improved spatial memory whereas lithium treatment had no effect. Analysis of neuronal local field potentials in anesthetized animals revealed that whereas both repeated SB 216763 (2.5 mg/kg) and lithium (100 mg/kg) induced a theta frequency spike in HIP at approximately 10 Hz, only SB 216763 treatment induced an overall increase in theta power (4-12 Hz) compared to vehicle. Acute administration of either drug suppressed slow (32-59 Hz) and fast (61-100 Hz) gamma power. In PL, both drugs induced an increase in theta power. Repeated SB 216763 increased HIP-PL coherence across all frequencies except delta, whereas lithium selectively suppressed delta coherence. These findings demonstrate that GSK-3 plays a direct role in the regulation of theta oscillations in regions critically involved in cognition, and highlight a potential mechanism by which GSK-3 may contribute to cognitive decline in disorders of cognitive dysfunction.

4.
Eur Neuropsychopharmacol ; 26(9): 1366-1377, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27480020

RESUMEN

Cocaine-induced increases in dopamine signaling in nucleus accumbens (NAc) play a significant role in cocaine seeking behavior. The majority of cocaine addiction research has focused on neuroanatomically segregated dopamine D1 and D2 receptor-expressing neurons, yet an involvement for those NAc neurons coexpressing D1 and D2 receptors in cocaine addiction has never been explored. In situ proximity ligation assay, confocal fluorescence resonance energy transfer and coimmunoprecipitation were used to show native D1 and D2 receptors formed a heteromeric complex in D1/D2 receptor-coexpressing neurons in rat and non-human primate NAc. D1-D2 heteromer expression was lower in NAc of adolescent rats compared to their adult counterparts. Functional disruption of the dopamine D1-D2 receptor heteromer, using a peptide targeting the site of interaction between the D1 and D2 receptor, induced conditioned place preference and increased NAc expression of ∆FosB. D1-D2 heteromer disruption also resulted in the promotion, exacerbation and acceleration of the locomotor activating and incentive motivational effects of cocaine in the self-administration paradigm. These findings support a model for tonic inhibition of basal and cocaine-induced reward processes by the D1-D2 heteromer thus highlighting its potential value as a novel target for drug discovery in cocaine addiction. Given that adolescents show increased drug abuse susceptibility, an involvement for reduced D1-D2 heteromer function in the heightened sensitivity to the rewarding effects of cocaine in adolescence is also implicated.


Asunto(s)
Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Neuronas/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Células Cultivadas , Cocaína/administración & dosificación , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Antagonistas de Dopamina/farmacología , Inhibidores de Captación de Dopamina/administración & dosificación , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Macaca fascicularis , Masculino , Motivación/efectos de los fármacos , Motivación/fisiología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuronas/citología , Neuronas/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/crecimiento & desarrollo , Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Autoadministración , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología
5.
Eur Neuropsychopharmacol ; 25(12): 2437-48, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26431907

RESUMEN

A role for the mesolimbic dopaminergic system in the pathophysiology of depression has become increasingly evident. Specifically, brain-derived neurotrophic factor (BDNF) has been shown to be elevated in the nucleus accumbens of depressed patients and to positively contribute to depression-like behaviour in rodents. The dopamine D1-D2 receptor heteromer exhibits significant expression in NAc and has also been shown to enhance BDNF expression and signalling in this region. We therefore examined the effects of D1-D2 heteromer stimulation in rats by SKF 83959, or its inactivation by a selective heteromer-disrupting TAT-D1 peptide on depression- and anxiety-like behaviours in non-stressed animals and in animals exposed to chronic unpredictable stress. SKF 83959 treatment significantly enhanced the latency to immobility in the forced swim test, increased the latency to drink condensed milk and reduced total milk consumption in the novelty-induced hypophagia test, and additionally reduced the total time spent in the open arms in the elevated plus maze test. These pro-depressant and anxiogenic effects of SKF 83959 were consistently abolished or attenuated by TAT-D1 peptide pre-treatment, signifying the behaviours were mediated by the D1-D2 heteromer. More importantly, in animals exposed to chronic unpredictable stress (CUS), TAT-D1 peptide treatment alone induced significant and rapid anxiolytic and antidepressant-like effects in two tests for CUS-induced anhedonia-like reactivity and in the novelty-suppressed feeding test. Together these findings indicate a positive role for the D1-D2 heteromer in mediating depression- and anxiety-like behaviours and suggest its possible value as a novel therapeutic target.


Asunto(s)
Ansiolíticos/uso terapéutico , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Estrés Psicológico/metabolismo , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Dopaminérgicos/farmacología , Relación Dosis-Respuesta a Droga , Conducta Alimentaria/efectos de los fármacos , Preferencias Alimentarias/efectos de los fármacos , Productos del Gen tat/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Sacarosa/administración & dosificación , Natación/psicología
6.
Pharmacol Biochem Behav ; 128: 33-40, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25444866

RESUMEN

A role for the dopamine D1-D2 receptor heteromer in the regulation of reward and addiction-related processes has been previously implicated. In the present study, we examined the effects of D1-D2 heteromer stimulation by the agonist SKF 83959 and its disruption by a selective TAT-D1 peptide on amphetamine-induced locomotor sensitization, a behavioral model widely used to study the neuroadaptations associated with psychostimulant addiction. D1-D2 heteromer activation by SKF 83959 did not alter the acute locomotor effects of amphetamine but significantly inhibited amphetamine-induced locomotor responding across the 5day treatment regimen. In addition, a single injection of SKF 83959 was sufficient to abolish the expression of locomotor sensitization induced by a priming injection of amphetamine after a 72-hour withdrawal. Conversely, inhibition of D1-D2 heteromer activity by the TAT-D1 peptide enhanced subchronic amphetamine-induced locomotion and the expression of amphetamine locomotor sensitization. Treatment solely with the TAT-D1 disrupting peptide during the initial 5day treatment phase was sufficient to induce a sensitized locomotor phenotype in response to the priming injection of amphetamine. Together these findings demonstrate that the dopamine D1-D2 receptor heteromer exerts a tonic inhibitory control on neurobiological processes involved in sensitization to amphetamine, indicating that the dopamine D1-D2 receptor heteromer may be a novel molecular substrate in addiction processes involving psychostimulants.


Asunto(s)
Anfetamina/farmacología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Receptores de Dopamina D1/fisiología , Receptores de Dopamina D2/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Trastornos Relacionados con Anfetaminas/fisiopatología , Trastornos Relacionados con Anfetaminas/psicología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Estimulantes del Sistema Nervioso Central/farmacología , Antagonistas de Dopamina/farmacología , Antagonistas de los Receptores de Dopamina D2/farmacología , Masculino , Complejos Multiproteicos/química , Complejos Multiproteicos/fisiología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/química , Receptores de Dopamina D2/química , Recompensa
7.
FASEB J ; 28(11): 4806-20, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25063849

RESUMEN

Although the dopamine D1-D2 receptor heteromer has emerging physiological relevance and a postulated role in different neuropsychiatric disorders, such as drug addiction, depression, and schizophrenia, there is a need for pharmacological tools that selectively target such receptor complexes in order to analyze their biological and pathophysiological functions. Since no selective antagonists for the D1-D2 heteromer are available, serial deletions and point mutations were used to precisely identify the amino acids involved in an interaction interface between the receptors, residing within the carboxyl tail of the D1 receptor that interacted with the D2 receptor to form the D1-D2 receptor heteromer. It was determined that D1 receptor carboxyl tail residues (404)Glu and (405)Glu were critical in mediating the interaction with the D2 receptor. Isolated mutation of these residues in the D1 receptor resulted in the loss of agonist activation of the calcium signaling pathway mediated through the D1-D2 receptor heteromer. The physical interaction between the D1 and D2 receptor could be disrupted, as shown by coimmunoprecipitation and BRET analysis, by a small peptide generated from the D1 receptor sequence that contained these amino acids, leading to a switch in G-protein affinities and loss of calcium signaling, resulting in the inhibition of D1-D2 heteromer function. The use of the D1-D2 heteromer-disrupting peptide in vivo revealed a pathophysiological role for the D1-D2 heteromer in the modulation of behavioral despair. This peptide may represent a novel pharmacological tool with potential therapeutic benefits in depression treatment.


Asunto(s)
Señalización del Calcio/fisiología , Neuronas/metabolismo , Multimerización de Proteína , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Animales , Encéfalo/metabolismo , Dopamina/metabolismo , Antagonistas de los Receptores de Dopamina D2/farmacología , Masculino , Neuronas/efectos de los fármacos , Péptidos/metabolismo , Ratas Sprague-Dawley , Receptores de Dopamina D1/antagonistas & inhibidores
8.
Biochem Biophys Res Commun ; 450(1): 906-11, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24976397

RESUMEN

The µ-δ opioid receptor heteromer activates the pertussis toxin-resistant Gαz GTP-binding protein following stimulation by the δ-agonist deltorphin-II whereas µ- and δ-receptors activate the pertussis toxin-sensitive Gαi3 protein following stimulation by µ- and δ-agonists, respectively. Although the regulation of the µ-δ heteromer is being investigated extensively in vitro, its physiological relevance remains elusive owing to a lack of available molecular tools. We investigated µ-δ heteromer signaling under basal conditions and following prolonged morphine treatment in rodent brain regions highly co-expressing µ- and δ-receptors and Gαz. Deltorphin-II induced Gαz activation in the striatum and hippocampus, demonstrating the presence of µ-δ heteromer signaling in these brain regions. Prolonged morphine treatment, which desensitizes µ- and δ-receptor function, had no effect on µ-δ heteromer signaling in the brain. Our data demonstrate that µ-δ heteromer signaling does not desensitize and is regulated differently from µ- and δ-receptor signaling following prolonged morphine treatment.


Asunto(s)
Cuerpo Estriado/metabolismo , Hipocampo/metabolismo , Morfina/farmacología , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal/fisiología , Analgésicos Opioides/farmacología , Animales , Cuerpo Estriado/efectos de los fármacos , Dimerización , Hipocampo/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Distribución Tisular/efectos de los fármacos
9.
Dev Neurosci ; 35(5): 384-95, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24021607

RESUMEN

Brain-derived neurotrophic factor (BDNF) signaling through its receptor, tropomyosin receptor kinase B (TrkB), plays a critical role in neural plasticity and its dysregulation in striatum and prefrontal cortex (PFC) has been implicated in the etiology of mental health disorders such schizophrenia and drug addiction. In the present study, we characterized age-dependent differences in BDNF signaling and TrkB expression within the nucleus accumbens (NAc), caudate putamen (CP) and PFC in rats and determined the effects of administration of the dopamine agonist, SKF 83959, which activates the Gq-coupled dopamine receptors, the dopamine D5 receptor and the D1-D2 receptor heteromer. As proBDNF binds with high affinity to the p75 neurotrophin receptor (p75NTR), expression levels of these proteins were also assessed. The present findings showed that juvenile rats (aged 26-28 days) exhibited significantly elevated basal BDNF expression and activation of full-length TrkB (TrkBfull) in NAc compared to their adult counterparts, as evidenced by increased TrkBfull phosphorylation. These changes were concomitant with an increase in the relative expression of TrkBfull compared to the truncated isoform, TrkB.T1, in NAc and CP. Conversely, in PFC the basal expression of BDNF in juvenile rats was significantly lower than in adult rats with an elevated relative expression of TrkBfull. Acute administration of SKF 83959 to juvenile rats abolished the age-dependent differences in BDNF expression in NAc and PFC, and in the relative expression of TrkBfull in NAc and CP. Together these findings indicate that the expression and/or signaling of BDNF and TrkB in striatum and PFC of juvenile rats is fundamentally different from that of adult rats, a finding that may have implications in neuropsychiatric disorders that exhibit age-dependent susceptibility such as schizophrenia and drug addiction.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas/metabolismo , Núcleo Accumbens/metabolismo , Receptor trkB/metabolismo , Transducción de Señal/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Factores de Edad , Animales , Núcleo Caudado/efectos de los fármacos , Núcleo Caudado/metabolismo , Agonistas de Dopamina/farmacología , Masculino , Neuronas/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Fosforilación/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Int J Dev Neurosci ; 31(3): 189-95, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23313435

RESUMEN

The goal of this study was to determine whether two stressors commonly used to model aspects of neuropsychiatric disease in rats have an additive effect on striatal dopamine type 2 receptor (D2R) expression, a key player in the etiology of neuropsychiatric disease. Animals subjected to early postnatal stress show alterations in function of the dopaminergic system thought to be mediated by stress-induced glucocorticoid release. Subsequent stress during puberty is known to further impact the dopaminergic system and result in dopaminergic hyperactivity analogous to schizophrenia. We exposed rats to maternal deprivation (MD) during the second postnatal week, a time of active striatal development. A subset of these animals were then subjected to pubertal stress induced by immobilization. Both procedures are know to induce glucocorticoid release. At the conclusion of the MD protocol, we observed upregulation in the expression of D2R and of dopamine- and cAMP-regulated phosphoprotein 32-KD (DARPP-32; PPP1R1B), but not of D1R, calcium/calmodulin-dependent protein kinase II beta (CaMKIIß), CaMKIIα or neurokinin B (NKB). Animals exposed to pubertal stress showed upregulation in expression of both D2R and CaMKIIß. Furthermore, rats previously exposed to MD showed a much greater upregulation in CaMKIIß expression, than animals only exposed to pubertal stress. These results support the two-hit hypothesis, indicating that such stressors have an additive effect. The main targets appear to be the D2R and the CaMKIIß, the latter being an important member of the DR signalling pathway, both of which are associated with schizophrenia.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cuerpo Estriado/metabolismo , Privación Materna , Receptores de Dopamina D2/metabolismo , Estrés Fisiológico/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Femenino , Expresión Génica , Masculino , Neuronas/metabolismo , Fosforilación , Ratas , Receptores de Dopamina D2/genética , Restricción Física , Regulación hacia Arriba
11.
Synapse ; 67(4): 179-88, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23184870

RESUMEN

Because abnormal development of striatal neurons is thought to be the part of pathology underlying major psychiatric illnesses, we studied the expression pattern of genes involved in striatal development and of genes comprising key striatal-specific pathways, during an active striatal maturation period, the first two postnatal weeks in rat. This period parallels human striatal development during the second trimester, when prenatal stress is though to lead to increased risk for neuropsychiatric disorders. To identify genes involved in this developmental process, we used subtractive hybridization, followed by quantitative real-time PCR, which allowed us to characterize the developmental expression of over 60 genes, many not previously known to play a role in neuromaturation. Of these 12 were novel transcripts, which did not match known genes, but which showed strict developmental expression and may play a role in striatal neurodevelopment. An additional 89 genes were identified as strong candidates for involvement in this neurodevelopmental process. We show that during the first two postnatal weeks in rat, an early gene expression network, still lacking key striatal-specific signaling pathways, is downregulated and replaced by a mature gene expression network, containing key striatal-specific genes including the dopamine D1 and D2 receptors, conferring to these neurons their functional identity. Therefore, before this developmental switch, striatal neurons lack many of their key phenotypic characteristics. This maturation process is followed by a striking rise in expression of myelination genes, indicating a striatal-specific myelination event. Such strictly controlled developmental program has the potential to be a point of susceptibility to disruption by external factors. Indeed, this period is known to be a susceptibility period in both humans and rats.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Vaina de Mielina/metabolismo , Neostriado/crecimiento & desarrollo , Animales , Neuronas GABAérgicas/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Modelos Animales , Neostriado/embriología , Neostriado/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Esquizofrenia/genética , Transmisión Sináptica/genética
12.
PLoS One ; 7(3): e33348, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22428025

RESUMEN

In basal ganglia a significant subset of GABAergic medium spiny neurons (MSNs) coexpress D1 and D2 receptors (D1R and D2R) along with the neuropeptides dynorphin (DYN) and enkephalin (ENK). These coexpressing neurons have been recently shown to have a region-specific distribution throughout the mesolimbic and basal ganglia circuits. While the functional relevance of these MSNs remains relatively unexplored, they have been shown to exhibit the unique property of expressing the dopamine D1-D2 receptor heteromer, a novel receptor complex with distinct pharmacology and cell signaling properties. Here we showed that MSNs coexpressing the D1R and D2R also exhibited a dual GABA/glutamate phenotype. Activation of the D1R-D2R heteromer in these neurons resulted in the simultaneous, but differential regulation of proteins involved in GABA and glutamate production or vesicular uptake in the nucleus accumbens (NAc), ventral tegmental area (VTA), caudate putamen and substantia nigra (SN). Additionally, activation of the D1R-D2R heteromer in NAc shell, but not NAc core, differentially altered protein expression in VTA and SN, regions rich in dopamine cell bodies. The identification of a MSN with dual inhibitory and excitatory intrinsic functions provides new insights into the neuroanatomy of the basal ganglia and demonstrates a novel source of glutamate in this circuit. Furthermore, the demonstration of a dopamine receptor complex with the potential to differentially regulate the expression of proteins directly involved in GABAergic inhibitory or glutamatergic excitatory activation in VTA and SN may potentially provide new insights into the regulation of dopamine neuron activity. This could have broad implications in understanding how dysregulation of neurotransmission within basal ganglia contributes to dopamine neuronal dysfunction.


Asunto(s)
Ganglios Basales/citología , Regulación de la Expresión Génica/fisiología , Complejos Multiproteicos/metabolismo , Neuronas/metabolismo , Fenotipo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Análisis de Varianza , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Fluorescencia , Eliminación de Gen , Glutamato Descarboxilasa/metabolismo , Ácido Glutámico/metabolismo , Immunoblotting , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/genética , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Ácido gamma-Aminobutírico/metabolismo
13.
J Biol Chem ; 285(47): 36625-34, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20864528

RESUMEN

The distribution and function of neurons coexpressing the dopamine D1 and D2 receptors in the basal ganglia and mesolimbic system are unknown. We found a subset of medium spiny neurons coexpressing D1 and D2 receptors in varying densities throughout the basal ganglia, with the highest incidence in nucleus accumbens and globus pallidus and the lowest incidence in caudate putamen. These receptors formed D1-D2 receptor heteromers that were localized to cell bodies and presynaptic terminals. In rats, selective activation of D1-D2 heteromers increased grooming behavior and attenuated AMPA receptor GluR1 phosphorylation by calcium/calmodulin kinase IIα in nucleus accumbens, implying a role in reward pathways. D1-D2 heteromer sensitivity and functional activity was up-regulated in rat striatum by chronic amphetamine treatment and in globus pallidus from schizophrenia patients, indicating that the dopamine D1-D2 heteromer may contribute to psychopathologies of drug abuse, schizophrenia, or other disorders involving elevated dopamine transmission.


Asunto(s)
Anfetamina/farmacología , Dinorfinas/metabolismo , Encefalinas/metabolismo , Neuronas/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Esquizofrenia/metabolismo , Animales , Ganglios Basales/efectos de los fármacos , Ganglios Basales/metabolismo , Conducta Animal , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Dopaminérgicos/farmacología , Globo Pálido/efectos de los fármacos , Globo Pálido/metabolismo , Humanos , Técnicas para Inmunoenzimas , Masculino , Neuronas/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/patología
14.
Psychopharmacology (Berl) ; 207(4): 599-618, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19830406

RESUMEN

RATIONALE AND OBJECTIVES: Amphetamine-induced sensitization is thought to involve dopamine D(1) receptors. Using mice lacking dopamine D(1) receptors (D (1) (-/-) ), we found that they exhibited blunted sensitization to low doses of amphetamine, while others using different treatment and testing regimens reported inconsistent results. We investigated whether experimental variables, alteration in gene expression or cholinergic input played a role in amphetamine-induced responses. METHODS: D (1) (-/-) and wild-type (D (1) (+/+) ) mice pretreated with amphetamine (1 mg/kg, 3-7 days) or various doses of nicotine (chronically but intermittently) were challenged with amphetamine (0.7 and/or 1 mg/kg) after short and long abstinence periods. Expression of brain-derived neurotrophic factor (BDNF) and phosphorylated c-AMP response element binding protein (p-CREB) genes were measured under basal conditions and after acute or repeated amphetamine treatments. RESULTS: D (1) (-/-) mice failed to exhibit amphetamine-induced sensitization following short-term treatments and long abstinence periods, but expressed sensitization following prolonged amphetamine treatment or a shorter abstinence period. Basal expression of p-CREB (but not BDNF) was higher in D (1) (-/-) than D (1) (+/+) mice and was reduced after amphetamine treatment. Prolonged nicotine pretreatment augmented locomotor responses to amphetamine in both genotypes and restored sensitization in D (1) (-/-) mice. CONCLUSIONS: D(1) receptors were necessary for induction, but may not be necessary for expression of amphetamine-induced sensitization at low doses. The manifestation of amphetamine sensitization depended on the duration of treatment and length of the withdrawal period. Cholinergic-nicotinic stimulation restored amphetamine-induced sensitization in D (1) (-/-) mice. Enhanced basal expression of p-CREB in D (1) (-/-) mice may represent an adaptive mechanism related to lack of D(1) receptors.


Asunto(s)
Anfetamina/farmacología , Dopaminérgicos/farmacología , Actividad Motora/efectos de los fármacos , Receptores de Dopamina D1/genética , Anfetamina/administración & dosificación , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dopaminérgicos/administración & dosificación , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nicotina/administración & dosificación , Nicotina/farmacología , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/farmacología , Fosforilación , Factores de Tiempo
15.
Proc Natl Acad Sci U S A ; 106(50): 21377-82, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19948956

RESUMEN

Although the perturbation of either the dopaminergic system or brain-derived neurotrophic factor (BDNF) levels has been linked to important neurological and neuropsychiatric disorders, there is no known signaling pathway linking these two major players. We found that the exclusive stimulation of the dopamine D1-D2 receptor heteromer, which we identified in striatal neurons and adult rat brain by using confocal FRET, led to the activation of a signaling cascade that links dopamine signaling to BDNF production and neuronal growth through a cascade of four steps: (i) mobilization of intracellular calcium through Gq, phospholipase C, and inositol trisphosphate, (ii) rapid activation of cytosolic and nuclear calcium/calmodulin-dependent kinase IIalpha, (iii) increased BDNF expression, and (iv) accelerated morphological maturation and differentiation of striatal neurons, marked by increased microtubule-associated protein 2 production. These effects, although robust in striatal neurons from D5(-/-) mice, were absent in neurons from D1(-/-) mice. We also demonstrated that this signaling cascade was activated in adult rat brain, although with regional specificity, being largely limited to the nucleus accumbens. This dopaminergic pathway regulating neuronal growth and maturation through BDNF may have considerable significance in disorders such as drug addiction, schizophrenia, and depression.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Señalización del Calcio , Neurogénesis , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Animales , Química Encefálica , Diferenciación Celular , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Núcleo Accumbens , Multimerización de Proteína
16.
Biochemistry ; 46(45): 12997-3009, 2007 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-17941650

RESUMEN

The cellular site of formation, Galpha-coupling preference, and agonist regulation of mu-delta opioid receptor (OR) heterooligomers were studied. Bioluminescence resonance energy transfer (BRET) showed that mu-deltaOR heterooligomers, composed of preformed mu and delta homooligomers, interacted constitutively in the endoplasmic reticulum (ER) with Galpha-proteins forming heteromeric signaling complexes before being targeted to the plasma membrane. Compared to muOR homooligomers, the mu-delta heterooligomers showed higher affinity and efficiency of interaction for Gz over Gi, indicating a switch in G-protein preference. Treatment with DAMGO or deltorphin II led to coregulated internalization of both receptors, whereas DPDPE and DSLET had no effect on mu-delta internalization. Staggered expression resulted in non-interacting mu and delta receptors, even though both receptors were colocalized at the cell surface. Agonists failed to induce BRET between staggered receptors, and resulted in internalization solely of the receptor targeted by agonist. Thus, mu-deltaOR heterooligomers form and preferentially associate with Gz to generate a signaling complex in the ER, and have a distinct agonist-internalization profile compared to either mu or delta homooligomers.


Asunto(s)
Membrana Celular/efectos de los fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalina D-Penicilamina (2,5)/farmacología , Proteínas de Unión al GTP/metabolismo , Humanos , Oligopéptidos/farmacología , Transporte de Proteínas , Ratas , Receptores Opioides delta/agonistas , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/efectos de los fármacos
17.
Mol Pharmacol ; 72(5): 1157-70, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17699686

RESUMEN

There is accumulating evidence that G protein-coupled receptor signaling is regulated by localization in lipid raft microdomains. In this report, we determined that the D1 dopamine receptor (D1R) is localized in caveolae, a subset of lipid rafts, by sucrose gradient fractionation and confocal microscopy. Through coimmunoprecipitation and bioluminescence resonance energy transfer assays, we demonstrated that this localization was mediated by an interaction between caveolin-1 and D1R in COS-7 cells and an isoform-selective interaction between D1R and caveolin-1alpha in rat brain. We determined that the D1R interaction with caveolin-1 required a putative caveolin binding motif identified in transmembrane domain 7. Agonist stimulation of D1R caused translocation of D1R into caveolin-1-enriched sucrose fractions, which was determined to be a result of D1R endocytosis through caveolae. This was found to be protein kinase A-independent and a kinetically slower process than clathrin-mediated endocytosis. Site-directed mutagenesis of the caveolin binding motif at amino acids Phe313 and Trp318 significantly attenuated caveolar endocytosis of D1R. We also found that these caveolin binding mutants had a diminished capacity to stimulate cAMP production, which was determined to be due to constitutive desensitization of these receptors. In contrast, we found that D1Rs had an enhanced ability to maximally generate cAMP in chemically induced caveolae-disrupted cells. Taken together, these data suggest that caveolae has an important role in regulating D1R turnover and signaling in brain.


Asunto(s)
Encéfalo/metabolismo , Caveolas/metabolismo , Caveolina 1/metabolismo , Endocitosis , Receptores de Dopamina D1/metabolismo , Secuencias de Aminoácidos , Animales , Células COS , Caveolas/química , Caveolina 1/análisis , Membrana Celular/química , Membrana Celular/metabolismo , Chlorocebus aethiops , Colesterol/metabolismo , Humanos , Mutación , Ratas , Receptores de Dopamina D1/análisis , Receptores de Dopamina D1/genética , Transducción de Señal
18.
Mol Pharmacol ; 70(1): 78-89, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16597839

RESUMEN

The role of oligomerization in D1 dopamine receptor trafficking to the cell surface was examined using conformationally distinct variants of this receptor. Substitution of the highly conserved aspartic acid (Asp103) in transmembrane domain 3 resulted in a constitutively active receptor, D103A, that did not bind agonists or antagonists but trafficked to the cell surface as oligomers. Coexpression of D103A with the wild-type D1 receptor in human embryonic kidney 293t cells resulted in inhibition of cell surface expression of the D1 receptor because of receptor oligomerization, causing intracellular retention of both proteins. Rescue of the intracellularly retained oligomer could be achieved only by membrane-permeable full and partial agonists, which resulted in cell surface expression of the D1 receptor, whereas cell-permeable antagonists and cell impermeable agonists had no effect. Cell surface fluorescence resonance energy transfer studies of cells coexpressing D103A and D1 revealed no signal before agonist treatment but a robust signal after agonist treatment, indicating that the intact D1/D103A oligomer reached the cell surface only after agonist treatment but not under basal conditions. This suggests that rescue of the retained D1/D103A oligomer to the cell surface was a result of an agonist-induced change in the conformation of D1, permitting cell surface trafficking of the D1/D103A receptor oligomeric complex from the endoplasmic reticulum.


Asunto(s)
Agonistas de Dopamina/farmacología , Receptores de Dopamina D1/metabolismo , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Sustitución de Aminoácidos/genética , Animales , Apomorfina/farmacología , Benzazepinas/metabolismo , Benzazepinas/farmacología , Unión Competitiva/efectos de los fármacos , Células COS , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Chlorocebus aethiops , AMP Cíclico/metabolismo , Dimerización , Dopamina/farmacología , Expresión Génica , Humanos , Microscopía Fluorescente , Mutación/genética , Transporte de Proteínas/efectos de los fármacos , Ensayo de Unión Radioligante , Receptores de Dopamina D1/química , Receptores de Dopamina D1/genética , Transfección
19.
J Biol Chem ; 280(46): 38478-88, 2005 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16159882

RESUMEN

Opioid receptor pharmacology in vivo has predicted a greater number of receptor subtypes than explained by the profiles of the three cloned opioid receptors, and the functional dependence of the receptors on each other shown in gene-deleted animal models remains unexplained. One mechanism for such findings is the generation of novel signaling complexes by receptor hetero-oligomerization, which we previously showed results in significantly different pharmacology for mu and delta receptor hetero-oligomers compared with the individual receptors. In the present study, we show that deltorphin-II is a fully functional agonist of the mu-delta heteromer, which induced desensitization and inhibited adenylyl cyclase through a pertussis toxin-insensitive G protein. Activation of the mu-delta receptor heteromer resulted in preferential activation of Galpha(z), illustrated by incorporation of GTPgamma(35)S, whereas activation of the individually expressed mu and delta receptors preferentially activated Galpha(i). The unique pharmacology of the mu-delta heteromer was dependent on the reciprocal involvement of the distal carboxyl tails of both receptors, so that truncation of the distal mu receptor carboxyl tail modified the delta-selective ligand-binding pocket, and truncation of the delta receptor distal carboxyl tail modified the mu-selective binding pocket. The distal carboxyl tails of both receptors also had a significant role in receptor interaction, as evidenced by the reduced ability to co-immunoprecipitate when the carboxyl tails were truncated. The interaction between mu and delta receptors occurred constitutively when the receptors were co-expressed, but did not occur when receptor expression was temporally separated, indicating that the hetero-oligomers were generated by a co-translational mechanism.


Asunto(s)
Proteínas de Unión al GTP/química , Receptores Opioides delta/química , Receptores Opioides mu/química , Adenilil Ciclasas/metabolismo , Analgésicos Opioides/farmacología , Animales , Células CHO , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Clonación Molecular , Cricetinae , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalina D-Penicilamina (2,5)/farmacología , Eliminación de Gen , Guanina/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Ligandos , Microscopía Fluorescente , Oligopéptidos/química , Toxina del Pertussis/farmacología , Unión Proteica , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Ratas , Transducción de Señal , Factores de Tiempo , Transfección
20.
J Pharmacol Exp Ther ; 307(2): 481-9, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12970389

RESUMEN

G protein-coupled receptors have a core consisting of seven transmembrane alpha-helices that is important in maintaining the structure of the receptor. We postulated that disruption of the transmembrane core may interfere with receptor function. In this study, the function of integral membrane proteins was disrupted in vivo using peptides mimicking their transmembrane domains. A peptide derived from transmembrane 7 of the D2 dopamine receptor injected unilaterally into caudate nucleus of rats challenged with apomorphine resulted in rotational behavior, indicating D2 receptor blockade. No rotational behavior was seen with a similar peptide based on the beta2 adrenergic receptor and the D2 transmembrane peptide did not affect the D1 dopamine receptor, indicating that the D2 receptor-derived peptide had a specific effect. The intravenous administration of a transmembrane peptide derived from the alpha1-adrenergic receptor resulted in lowered arterial blood pressure and injection of a beta1-adrenergic receptor peptide resulted in decreased heart rate. Injection of a V2 vasopressin receptor-derived transmembrane peptide resulted in increased urine output, suggesting antagonism of the effects of vasopressin. Finally, dopamine release in rat brain after cocaine administration was blocked by a transmembrane peptide based on the dopamine transporter. Circular dichroism spectroscopy of the peptides revealed alpha-helical structure similar to that of native transmembrane domains. Thus, transmembrane peptides can disrupt membrane proteins in vivo likely by competing with native transmembrane domains. The disruption of the hydrophobic core architecture of membrane proteins represents a novel mechanism of achieving functional inhibition that may be possible to exploit in developing novel therapeutics.


Asunto(s)
Glicoproteínas de Membrana , Proteínas de la Membrana/farmacología , Moduladores del Transporte de Membrana , Proteínas de Transporte de Membrana/antagonistas & inhibidores , Proteínas del Tejido Nervioso , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Péptidos/farmacología , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...