Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(5)2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38474266

RESUMEN

One of the hallmarks of Parkinson's disease (PD) is the alteration in the expression and function of NMDA receptor (NMDAR) and cannabinoid receptor 1 (CB1R). The presence of CB1R-NMDAR complexes has been described in neuronal primary cultures. The activation of CB1R in CB1R-NMDAR complexes was suggested to counteract the detrimental NMDAR overactivation in an AD mice model. Thus, we aimed to explore the role of this receptor complex in PD. By using Bioluminescence Resonance Energy Transfer (BRET) assay, it was demonstrated that α-synuclein induces a reorganization of the CB1R-NMDAR complex in transfected HEK-293T cells. Moreover, α-synuclein treatment induced a decrease in the cAMP and MAP kinase (MAPK) signaling of both CB1R and NMDAR not only in transfected cells but also in neuronal primary cultures. Finally, the interaction between CB1R and NMDAR was studied by Proximity Ligation Assay (PLA) in neuronal primary cultures, where it was observed that the expression of CB1R-NMDAR complexes was decreased upon α-synuclein treatment. These results point to a role of CB1R-NMDAR complexes as a new therapeutic target in Parkinson's disease.


Asunto(s)
Enfermedad de Parkinson , Animales , Ratones , alfa-Sinucleína/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal
2.
Exp Neurol ; 362: 114319, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36632949

RESUMEN

It is of particular interest the potential of cannabinoid and angiotensin receptors as targets in the therapy of Parkinson's disease (PD). While endocannabinoids are neuromodulators that act through the CB1 and CB2 cannabinoid receptors, the renin angiotensin-system is relevant for regulation of the correct functioning of several brain circuits. Resonance energy transfer assays in a heterologous system showed that the CB1 receptor (CB1R) can directly interact with the angiotensin AT2 receptor (AT2R). Coactivation of the two receptors results in increased Gi-signaling. The AT2-CB1 receptor heteromer imprint consists of a blockade of AT2R-mediated signaling by rimonabant, a CB1R antagonist. Interestingly, the heteromer imprint, discovered in the heterologous system, was also found in primary striatal neurons thus demonstrating the expression of the heteromer in these cells. In situ proximity ligation assays confirmed the occurrence of AT2-CB1 receptor heteromers in striatal neurons. In addition, increased expression of the AT2-CB1 receptor heteromeric complexes was detected in the striatum of a rodent PD model consisting of rats hemilesioned using 6-hydroxydopamine. Expression of the heteromer was upregulated in the striatum of lesioned animals and, also, of lesioned animals that upon levodopa treatment became dyskinetic. In contrast, there was no upregulation in the striatum of lesioned rats that did not become dyskinetic upon chronic levodopa treatment. The results suggest that therapeutic developments focused on the CB1R should consider that this receptor can interact with the AT2R, which in the CNS is involved in mechanisms related to addictive behaviors and to neurodegenerative and neuroinflammatory diseases.


Asunto(s)
Cannabinoides , Enfermedad de Parkinson , Ratas , Animales , Receptores de Cannabinoides/metabolismo , Levodopa , Oxidopamina , Cuerpo Estriado/metabolismo , Enfermedad de Parkinson/metabolismo , Receptores de Angiotensina , Angiotensinas/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo
3.
Pharmacol Res ; 170: 105745, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34182128

RESUMEN

Polymorphic alleles of the human dopamine D4 receptor gene (DRD4) have been consistently associated with individual differences in personality traits and neuropsychiatric disorders, particularly between the gene encoding dopamine D4.7 receptor variant and attention deficit hyperactivity disorder (ADHD). The α2A adrenoceptor gene has also been associated with ADHD. In fact, drugs targeting the α2A adrenoceptor (α2AR), such as guanfacine, are commonly used in ADHD treatment. In view of the involvement of dopamine D4 receptor (D4R) and α2AR in ADHD and impulsivity, their concurrent localization in cortical pyramidal neurons and the demonstrated ability of D4R to form functional heteromers with other G protein-coupled receptors, in this study we evaluate whether the α2AR forms functional heteromers with D4R and weather these heteromers show different properties depending on the D4R variant involved. Using cortical brain slices from hD4.7R knock-in and wild-type mice, here, we demonstrate that α2AR and D4R heteromerize and constitute a significant functional population of cortical α2AR and D4R. Moreover, in cortical slices from wild-type mice and in cells transfected with α2AR and D4.4R, we detect a negative crosstalk within the heteromer. This negative crosstalk is lost in cortex from hD4.7R knock-in mice and in cells expressing the D4.7R polymorphic variant. We also show a lack of efficacy of D4R ligands to promote G protein activation and signaling only within the α2AR-D4.7R heteromer. Taken together, our results suggest that α2AR-D4R heteromers play a pivotal role in catecholaminergic signaling in the brain cortex and are likely targets for ADHD pharmacotherapy.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Corteza Cerebral/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Receptores de Dopamina D4/metabolismo , Animales , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno por Déficit de Atención con Hiperactividad/psicología , Corteza Cerebral/efectos de los fármacos , Agonistas de Dopamina/farmacología , Femenino , Células HEK293 , Humanos , Conducta Impulsiva , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Polimorfismo Genético , Unión Proteica , Receptores Adrenérgicos alfa 2/genética , Receptores de Dopamina D4/agonistas , Receptores de Dopamina D4/genética , Oveja Doméstica , Transducción de Señal
4.
Neurobiol Dis ; 143: 105017, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32679312

RESUMEN

BACKGROUND: Dopamine receptors interact with other receptors to form heterooligomers. One such complex, the D1-D2 heteromer, demonstrated in cultured striatal neurons and rat striatum has been linked to drug addiction, Parkinson's disease, schizophrenia, depression and anhedonia. METHODS: D1-D2 heteromer expression was evaluated using in situ proximity ligation assay, in parallel with cellular colocalization of D1 and D2 mRNA using in situ hybridization in 19 different key rat brain regions. Expression in higher species and changes in rat striatum after repeated cocaine administration were evaluated. RESULTS: Differences in D1-D2 heteromer expression in striatal subregions are documented in higher species with nonhuman primate and human demonstrating higher density of heteromer-expressing neurons compared to rodents. All species had higher density of D1-D2 neurons in nucleus accumbens compared to dorsal striatum. Multiple other brain regions are identified where D1-D2 heteromer is expressed, prominently in cerebral cortical subregions including piriform, medial prefrontal, orbitofrontal and others; subcortical regions such as claustrum, amygdala and lateral habenula. Three categories of regions are identified: D1-D2 heteromer expressed despite little to no observed D1/D2 mRNA colocalization, likely representing heteromer on neuronal projections from other brain regions; D1-D2 heteromer originating locally with the density of neurons expressing heteromer matching neurons with colocalized D1/D2 mRNA; regions with both a local origin and targeted inputs projecting from other regions. Repeated cocaine administration significantly increased density of neurons expressing D1-D2 heteromer and D1/D2 mRNA colocalization in rat striatum, with changes in both direct and indirect pathway neurons. CONCLUSION: The dopamine D1-D2 heteromer is expressed in key brain cortical and subcortical regions of all species examined. Species differences in striatum revealed greater abundance in human>nonhuman-primate>rat>mouse, suggesting an evolutionary biologic role for the D1-D2 heteromer in higher CNS function. Its upregulation in rat striatum following cocaine points to regulatory significance with possible relevance for clinical disorders such as drug addiction. The dopamine D1-D2 receptor heteromer may represent a potential target for neuropsychiatric and neurodegenerative disorders, given its distribution in highly relevant brain regions.


Asunto(s)
Cocaína/farmacología , Cuerpo Estriado/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Neuronas/metabolismo , Receptores Dopaminérgicos/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Trastornos Relacionados con Cocaína/metabolismo , Cuerpo Estriado/efectos de los fármacos , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Regulación hacia Arriba
5.
Expert Opin Drug Discov ; 14(12): 1297-1312, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31507210

RESUMEN

Introduction: G protein-coupled receptors (GPCRs) are a superfamily of membrane proteins highly expressed in the brain that are involved in almost all functions of the CNS. During the last twenty years, a large number of GPCRs have been reported to form homodimers, heterodimers and higher order oligomers. Areas covered: This review summarizes the functional and pharmacological characteristics of the dopamine D1 receptor (D1R) interactome constituted by heteromers with GPCRs or non-GPCRs. The review also focuses on heteromer-selective ligands reported for GPCRs, including those for the receptor-based interactome of D1R. Expert opinion: Since the D1R plays a key role in basal ganglia motor control, where all the mentioned D1R heteromers are present, the study of allosteric interactions within the D1R interactome may be of high therapeutic interest for treating motor dysfunction. Moreover, several of these heteromers have also been detected in the prefrontal cortex and hippocampus, where they are involved in learning, memory and attention dysfunction. We propose that drugs targeting specific D1R heteromers in the CNS will be more effective and safer, resulting in a reduction of side effects compared with traditional drugs targeting monomeric receptors. Heteromer-selective ligands will have a big impact on drug development with many pharmacological and clinical implications.


Asunto(s)
Desarrollo de Medicamentos/métodos , Receptores de Dopamina D1/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Diseño de Fármacos , Humanos , Ligandos , Receptores de Dopamina D1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
6.
Adv Pharmacol ; 84: 37-78, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31229177

RESUMEN

Dopaminergic and purinergic signaling play a pivotal role in neurological diseases associated with motor symptoms, including Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis, Huntington disease, Restless Legs Syndrome (RLS), spinal cord injury (SCI), and ataxias. Extracellular dopamine and adenosine exert their functions interacting with specific dopamine (DR) or adenosine (AR) receptors, respectively, expressed on the surface of target cells. These receptors are members of the family A of G protein-coupled receptors (GPCRs), which is the largest protein superfamily in mammalian genomes. GPCRs are target of about 40% of all current marketed drugs, highlighting their importance in clinical medicine. The striatum receives the densest dopamine innervations and contains the highest density of dopamine receptors. The modulatory role of adenosine on dopaminergic transmission depends largely on the existence of antagonistic interactions mediated by specific subtypes of DRs and ARs, the so-called A2AR-D2R and A1R-D1R interactions. Due to the dopamine/adenosine antagonism in the CNS, it was proposed that ARs and DRs could form heteromers in the neuronal cell surface. Therefore, adenosine can affect dopaminergic signaling through receptor-receptor interactions and by modulations in their shared intracellular pathways in the striatum and spinal cord. In this work we describe the allosteric modulations between GPCR protomers, focusing in those of adenosine and dopamine within the A1R-D1R heteromeric complex, which is involved in RLS. We also propose that the knowledge about the intricate allosteric interactions within the A1R-D1R heterotetramer, may facilitate the treatment of motor alterations, not only when the dopamine pathway is hyperactivated (RLS, chorea, etc.) but also when motor function is decreased (SCI, aging, PD, etc.).


Asunto(s)
Multimerización de Proteína , Receptor de Adenosina A1/química , Receptores Dopaminérgicos/química , Síndrome de las Piernas Inquietas/tratamiento farmacológico , Adenosina/metabolismo , Animales , Dopamina/metabolismo , Humanos , Receptor de Adenosina A1/metabolismo , Receptores Dopaminérgicos/metabolismo
7.
Front Pharmacol ; 10: 339, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024307

RESUMEN

The endocannabinoid system (ECS) has been placed in the anti-cancer spotlight in the last decade. The immense data load published on its dual role in both tumorigenesis and inhibition of tumor growth and metastatic spread has transformed the cannabinoid receptors CB1 (CB1R) and CB2 (CB2R), and other members of the endocannabinoid-like system, into attractive new targets for the treatment of various cancer subtypes. Although the clinical use of cannabinoids has been extensively documented in the palliative setting, clinical trials on their application as anti-cancer drugs are still ongoing. As drug repurposing is significantly faster and more economical than de novo introduction of a new drug into the clinic, there is hope that the existing pharmacokinetic and safety data on the ECS ligands will contribute to their successful translation into oncological healthcare. CB1R and CB2R are members of a large family of membrane proteins called G protein-coupled receptors (GPCR). GPCRs can form homodimers, heterodimers and higher order oligomers with other GPCRs or non-GPCRs. Currently, several CB1R and CB2R-containing heteromers have been reported and, in cancer cells, CB2R form heteromers with the G protein-coupled chemokine receptor CXCR4, the G protein-coupled receptor 55 (GPR55) and the tyrosine kinase receptor (TKR) human V-Erb-B2 Avian Erythroblastic Leukemia Viral Oncogene Homolog 2 (HER2). These protein complexes possess unique pharmacological and signaling properties, and their modulation might affect the antitumoral activity of the ECS. This review will explore the potential of the endocannabinoid network in the anti-cancer setting as well as the clinical and ethical pitfalls behind it, and will develop on the value of cannabinoid receptor heteromers as potential new targets for anti-cancer therapies and as prognostic biomarkers.

8.
Mol Neurobiol ; 56(2): 1196-1210, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29876881

RESUMEN

Despite ancient knowledge on cocaine appetite-suppressant action, the molecular basis of such fact remains unknown. Addiction/eating disorders (e.g., binge eating, anorexia, bulimia) share a central control involving reward circuits. However, we here show that the sigma-1 receptor (σ1R) mediates cocaine anorectic effects by interacting in neurons with growth/hormone/secretagogue (ghrelin) receptors. Cocaine increases colocalization of σ1R and GHS-R1a at the cell surface. Moreover, in transfected HEK-293T and neuroblastoma SH-SY5Y cells, and in primary neuronal cultures, pretreatment with cocaine or a σ1R agonist inhibited ghrelin-mediated signaling, in a similar manner as the GHS-R1a antagonist YIL-781. Results were similar in G protein-dependent (cAMP accumulation and calcium release) and in partly dependent or independent (ERK1/2 phosphorylation and label-free) assays. We provide solid evidence for direct interaction between receptors and the functional consequences, as well as a reliable structural model of the macromolecular σ1R-GHS-R1a complex, which arises as a key piece in the puzzle of the events linking cocaine consumption and appetitive/consummatory behaviors.


Asunto(s)
Cocaína/farmacología , Cuerpo Estriado/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Ghrelina/metabolismo , Neuronas/efectos de los fármacos , Ácido Oleanólico/análogos & derivados , Receptores sigma/metabolismo , Saponinas/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Células HEK293 , Humanos , Masculino , Modelos Moleculares , Neuronas/citología , Neuronas/metabolismo , Ácido Oleanólico/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Receptor Sigma-1
9.
J Neurosci ; 37(5): 1176-1186, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28007761

RESUMEN

The neuropeptide galanin has been shown to interact with the opioid system. More specifically, galanin counteracts the behavioral effects of the systemic administration of µ-opioid receptor (MOR) agonists. Yet the mechanism responsible for this galanin-opioid interaction has remained elusive. Using biophysical techniques in mammalian transfected cells, we found evidence for selective heteromerization of MOR and the galanin receptor subtype Gal1 (Gal1R). Also in transfected cells, a synthetic peptide selectively disrupted MOR-Gal1R heteromerization as well as specific interactions between MOR and Gal1R ligands: a negative cross talk, by which galanin counteracted MAPK activation induced by the endogenous MOR agonist endomorphin-1, and a cross-antagonism, by which a MOR antagonist counteracted MAPK activation induced by galanin. These specific interactions, which represented biochemical properties of the MOR-Gal1R heteromer, could then be identified in situ in slices of rat ventral tegmental area (VTA) with MAPK activation and two additional cell signaling pathways, AKT and CREB phosphorylation. Furthermore, in vivo microdialysis experiments showed that the disruptive peptide selectively counteracted the ability of galanin to block the dendritic dopamine release in the rat VTA induced by local infusion of endomorphin-1, demonstrating a key role of MOR-Gal1R heteromers localized in the VTA in the direct control of dopamine cell function and their ability to mediate antagonistic interactions between MOR and Gal1R ligands. The results also indicate that MOR-Gal1R heteromers should be viewed as targets for the treatment of opioid use disorders. SIGNIFICANCE STATEMENT: The µ-opioid receptor (MOR) localized in the ventral tegmental area (VTA) plays a key role in the reinforcing and addictive properties of opioids. With parallel in vitro experiments in mammalian transfected cells and in situ and in vivo experiments in rat VTA, we demonstrate that a significant population of these MORs form functional heteromers with the galanin receptor subtype Gal1 (Gal1R), which modulate the activity of the VTA dopaminergic neurons. The MOR-Gal1R heteromer can explain previous results showing antagonistic galanin-opioid interactions and offers a new therapeutic target for the treatment of opioid use disorder.


Asunto(s)
Receptores de Galanina/metabolismo , Receptores Opioides mu/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Neuronas Dopaminérgicas/efectos de los fármacos , Galanina/farmacología , Células HEK293 , Humanos , Ligandos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Oncogénica v-akt/fisiología , Fosforilación , Ratas , Receptor Cross-Talk , Receptor de Galanina Tipo 1/genética , Receptor de Galanina Tipo 1/metabolismo , Receptor de Galanina Tipo 2/genética , Receptor de Galanina Tipo 2/metabolismo , Receptores de Galanina/genética , Receptores Opioides mu/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Transfección
10.
Int J Mol Sci ; 17(5)2016 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-27331810

RESUMEN

Recent evidence suggests that C-X-C chemokine receptor type 4 (CXCR4) heteromerizes with α1A/B-adrenoceptors (AR) and atypical chemokine receptor 3 (ACKR3) and that CXCR4:α1A/B-AR heteromers are important for α1-AR function in vascular smooth muscle cells (VSMC). Structural determinants for CXCR4 heteromerization and functional consequences of CXCR4:α1A/B-AR heteromerization in intact arteries, however, remain unknown. Utilizing proximity ligation assays (PLA) to visualize receptor interactions in VSMC, we show that peptide analogs of transmembrane-domain (TM) 2 and TM4 of CXCR4 selectively reduce PLA signals for CXCR4:α1A-AR and CXCR4:ACKR3 interactions, respectively. While both peptides inhibit CXCL12-induced chemotaxis, only the TM2 peptide inhibits phenylephrine-induced Ca(2+)-fluxes, contraction of VSMC and reduces efficacy of phenylephrine to constrict isolated arteries. In a Cre-loxP mouse model to delete CXCR4 in VSMC, we observed 60% knockdown of CXCR4. PLA signals for CXCR4:α1A/B-AR and CXCR4:ACKR3 interactions in VSMC, however, remained constant. Our observations point towards TM2/4 of CXCR4 as possible contact sites for heteromerization and suggest that TM-derived peptide analogs permit selective targeting of CXCR4 heteromers. A molecular dynamics simulation of a receptor complex in which the CXCR4 homodimer interacts with α1A-AR via TM2 and with ACKR3 via TM4 is presented. Our findings further imply that CXCR4:α1A-AR heteromers are important for intrinsic α1-AR function in intact arteries and provide initial and unexpected insights into the regulation of CXCR4 heteromerization in VSMC.


Asunto(s)
Músculo Liso Vascular/metabolismo , Multimerización de Proteína , Receptores Adrenérgicos alfa 1/metabolismo , Receptores CXCR4/metabolismo , Animales , Sitios de Unión , Calcio/metabolismo , Línea Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Simulación de Dinámica Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/química , Receptores CXCR4/genética
11.
Psychopharmacology (Berl) ; 233(10): 1963-79, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26786412

RESUMEN

BACKGROUND: The psychostimulant properties of caffeine are reviewed and compared with those of prototypical psychostimulants able to cause substance use disorders (SUD). Caffeine produces psychomotor-activating, reinforcing, and arousing effects, which depend on its ability to disinhibit the brake that endogenous adenosine imposes on the ascending dopamine and arousal systems. OBJECTIVES: A model that considers the striatal adenosine A2A-dopamine D2 receptor heteromer as a key modulator of dopamine-dependent striatal functions (reward-oriented behavior and learning of stimulus-reward and reward-response associations) is introduced, which should explain most of the psychomotor and reinforcing effects of caffeine. HIGHLIGHTS: The model can explain the caffeine-induced rotational behavior in rats with unilateral striatal dopamine denervation and the ability of caffeine to reverse the adipsic-aphagic syndrome in dopamine-deficient rodents. The model can also explain the weaker reinforcing effects and low abuse liability of caffeine, compared with prototypical psychostimulants. Finally, the model can explain the actual major societal dangers of caffeine: the ability of caffeine to potentiate the addictive and toxic effects of drugs of abuse, with the particularly alarming associations of caffeine (as adulterant) with cocaine, amphetamine derivatives, synthetic cathinones, and energy drinks with alcohol, and the higher sensitivity of children and adolescents to the psychostimulant effects of caffeine and its potential to increase vulnerability to SUD. CONCLUSIONS: The striatal A2A-D2 receptor heteromer constitutes an unequivocal main pharmacological target of caffeine and provides the main mechanisms by which caffeine potentiates the acute and long-term effects of prototypical psychostimulants.


Asunto(s)
Cafeína/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Trastornos Relacionados con Sustancias/etiología , Animales , Humanos , Ratas
12.
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
13.
Prog Brain Res ; 211: 183-200, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24968781

RESUMEN

Dopamine neurotransmission is traditionally accepted as occurring through the five dopamine receptors that transduce its signal. Recent evidence has demonstrated that the range of physiologically relevant dopamine signaling complexes is greatly expanded by the ability of dopamine receptors to interact with other dopamine receptors and with receptors of other endogenous signaling ligands. These novel heteromeric complexes have functional properties distinct from the component receptors or are able to modulate the canonical signaling and function of the cognate receptors. These dopamine receptor heteromers provide new insight into physiological mechanisms and pathophysiological processes involving dopamine.


Asunto(s)
Encéfalo/metabolismo , Dopamina/metabolismo , Receptores Dopaminérgicos/metabolismo , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Animales , Humanos
14.
Expert Rev Neurother ; 14(7): 719-21, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24894440

RESUMEN

Allosteric mechanisms in receptor heteromers markedly increase the repertoire of receptor recognition and signaling. Of high importance is the altered function in the receptor heteromer versus the receptor homomer. Such a change in receptor function is mainly brought about by agonist induced allosteric receptor-receptor interactions and leads to functional and structural plasticity. Receptor-receptor interactions integrating synaptic and volume transmission signals participate in a significant way in modulating bidirectional synaptic plasticity and thus Hebbian plasticity. One molecular mechanism that can contribute to a change of synaptic weight may be represented by multiple interactions between plasma membrane receptors forming higher order heteroreceptor complexes via oligomerization at the pre- and post-junctional level. Such long-lived heteroreceptor complexes may play a significant role in learning and memory.


Asunto(s)
Encéfalo/metabolismo , Plasticidad Neuronal/fisiología , Receptores de Superficie Celular/metabolismo , Animales , Humanos , Receptores de Superficie Celular/genética , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología
15.
Neuropharmacology ; 79: 90-100, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24230991

RESUMEN

The molecular basis of priming for L-DOPA-induced dyskinesias in Parkinson's disease (PD), which depends on the indirect pathway of motor control, is not known. In rodents, the indirect pathway contains striatopallidal GABAergic neurons that express heterotrimers composed of A(2A) adenosine, CB(1) cannabinoid and D(2) dopamine receptors that regulate dopaminergic neurotransmission. The present study was designed to investigate the expression of these heteromers in the striatum of a primate model of Parkinson's disease and to determine whether their expression and pharmacological properties are altered upon L-DOPA treatment. By using the recently developed in situ proximity ligation assay and by identification of a biochemical fingerprint, we discovered a regional distribution of A(2A)/CB(1) /D(2) receptor heteromers that predicts differential D(2)-mediated neurotransmission in the caudate-putamen of Macaca fascicularis. Whereas heteromers were abundant in the caudate nucleus of both naïve and MPTP-treated monkeys, L-DOPA treatment blunted the biochemical fingerprint and led to weak heteromer expression. These findings constitute the first evidence of altered receptor heteromer expression in pathological conditions and suggest that drugs targeting A(2A)-CB(1) -D(2) receptor heteromers may be successful to either normalize basal ganglia output or prevent L-DOPA-induced side effects.


Asunto(s)
Antiparkinsonianos/farmacología , Núcleo Caudado/efectos de los fármacos , Levodopa/farmacología , Receptor de Adenosina A2A/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptores de Dopamina D2/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Agonistas del Receptor de Adenosina A2/farmacología , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Núcleo Caudado/metabolismo , Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Antagonistas de los Receptores de Dopamina D2 , Macaca fascicularis , Masculino , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/metabolismo , Putamen/efectos de los fármacos , Putamen/metabolismo , Receptor Cannabinoide CB1/agonistas
16.
Front Neuroanat ; 5: 31, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21747759

RESUMEN

Dopaminergic signaling within the basal ganglia has classically been thought to occur within two distinct neuronal pathways; the direct striatonigral pathway which contains the dopamine D1 receptor and the neuropeptides dynorphin (DYN) and substance P, and the indirect striatopallidal pathway which expresses the dopamine D2 receptor and enkephalin (ENK). A number of studies have also shown, however, that D1 and D2 receptors can co-exist within the same medium spiny neuron and emerging evidence indicates that these D1/D2-coexpressing neurons, which also express DYN and ENK, may comprise a third neuronal pathway, with representation in both the striatonigral and striatopallidal projections of the basal ganglia. Furthermore, within these coexpressing neurons it has been shown that the dopamine D1 and D2 receptor can form a novel and pharmacologically distinct receptor complex, the dopamine D1-D2 receptor heteromer, with unique signaling properties. This is indicative of a functionally unique role for these neurons in brain. The aim of this review is to discuss the evidence in support of a novel third pathway coexpressing the D1 and D2 receptor, to discuss the potential relevance of this pathway to basal ganglia signaling, and to address its potential value, and that of the dopamine D1-D2 receptor heteromer, in the search for new therapeutic strategies for disorders involving dopamine neurotransmission.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA