Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuropharmacology ; 180: 108307, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32941853

RESUMEN

Opioid signaling controls the activity of the brain's reward system. It is involved in signaling the hedonic effects of rewards and has essential roles in reinforcement and motivational processes. Here, we focused on opioid signaling through mu and delta receptors on dopaminoceptive neurons and evaluated the role these receptors play in reward-driven behaviors. We generated a genetically modified mouse with selective double knockdown of mu and delta opioid receptors in neurons expressing dopamine receptor D1. Selective expression of the transgene was confirmed using immunostaining. Knockdown was validated by measuring the effects of selective opioid receptor agonists on neuronal membrane currents using whole-cell patch clamp recordings. We found that in the nucleus accumbens of control mice, the majority of dopamine receptor D1-expressing neurons were sensitive to a mu or delta opioid agonist. In mutant mice, the response to the delta receptor agonist was blocked, while the effects of the mu agonist were strongly attenuated. Behaviorally, the mice had no obvious impairments. The mutation did not affect the sensitivity to the rewarding effects of morphine injections or social contact and had no effect on preference for sweet taste. Knockdown had a moderate effect on motor activity in some of the tests performed, but this effect did not reach statistical significance. Thus, we found that knocking down mu and delta receptors on dopamine receptor D1-expressing cells does not appreciably affect some of the reward-driven behaviors previously attributed to opioid signaling.


Asunto(s)
Neuronas/metabolismo , Receptores de Dopamina D1/biosíntesis , Receptores Opioides delta/deficiencia , Receptores Opioides mu/deficiencia , Recompensa , Analgésicos Opioides/farmacología , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Morfina/farmacología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Receptores de Dopamina D1/genética , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
2.
Neuropharmacology ; 166: 107920, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31870855

RESUMEN

Dopaminergic neurons have the ability to release Dopamine from their axons as well as from their soma and dendrites. This somatodendritically-released Dopamine induces an autoinhibition of Dopaminergic neurons mediated by D2 autoreceptors, and the stimulation of neighbor GABAergic neurons mediated by D1 receptors (D1r). Here, our results suggest that the somatodendritic release of Dopamine in the substantia nigra (SN) may stimulate GABAergic neurons that project their axons into the hippocampus. Using semiquantitative multiplex RT-PCR we show that chronic blockade of the Dopaminergic neurotransmission with both AMPT and reserpine specifically decreases the expression levels of D1r, remarkably this may be the result of an antagonistic effect between AMPT and reserpine, as they induced the expression of a different set of genes when treated by separate. Furthermore, using anterograde and retrograde tracing techniques, we found that the GABAergic neurons that express D1r also project their axons in to the CA1 region of the hippocampus. Finally, we also found that the same treatment that decreases the expression levels of D1r in SN, also induces an impairment in the performance in an appetitive learning task that requires the coding of reward as well as navigational skills. Overall, our findings show the presence of a GABAergic interconnection between the SNr and the hippocampus mediated by D1r.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Receptores de Dopamina D1/biosíntesis , Reserpina/farmacología , Sustancia Negra/metabolismo , alfa-Metiltirosina/farmacología , Inhibidores de Captación Adrenérgica/farmacología , Animales , Antagonistas de los Receptores de Dopamina D2/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/biosíntesis , Neuronas Dopaminérgicas/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Expresión Génica , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos BALB C , Fenotipo , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/biosíntesis , Receptores de Dopamina D2/genética , Sustancia Negra/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
3.
Artif Cells Nanomed Biotechnol ; 48(1): 276-287, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31858826

RESUMEN

Background: The abnormal expression Dopamine D1 receptor (DRD1) gives rise to the dysfunction of dopaminergic neurotransmitter and may be associated with the occurrence of schizophrenia. MicroRNAs (miRNAs) can regulate the DRD1 expression by binding 3'UTR and be involved in the post-transcriptional regulation.Methods: We first constructed the pmirGLO-recombined vectors of series of DRD1 gene 3'UTR-truncated fragments and performed the luciferase receptor assay to screen the underlying 3'UTR sequence targeted by miRNAs. Then, we predicted the potential miRNAs binding the target sequence and confirmed their effects using luciferase receptor assay after transfection of the miRNA mimics/inhibitors. We also examined the effects of the miRNA on the endogenous DRD1 expression.Results: We found that the DRD1 3'UTR ranging from -12 to +1135 bp was essential for the post-transcriptional regulation of miRNAs. The deletion of -12 to +154 bp fragment significantly increased the luciferase expression but not the mRNA expression. The miRNA-15a, miRNA-15b and miRNA 16 affected DRD1 expression in HEK293, U87, SK-N-SH and SH-SY5Y cell lines.Conclusion: The miRNA-15a, miRNA-15b and miRNA-16 inhibit the human dopamine D1 receptor expression by targeting 3'UTR -12 to +154 bp.HighlightsDRD1 3'UTR ranging from -12 to +1135 bp was essential for the post-transcriptional regulation of miRNAs.The deletion of -12 to +154 bp fragment significantly increased the luciferase expression but not the mRNA expression.The miRNA-15a, miRNA-15b and miRNA 16 affected DRD1 expression in different cell lines, respectively.


Asunto(s)
Regiones no Traducidas 3' , Regulación de la Expresión Génica , MicroARNs/metabolismo , Receptores de Dopamina D1/biosíntesis , Línea Celular Tumoral , Células HEK293 , Humanos , MicroARNs/genética , Receptores de Dopamina D1/genética
4.
Physiol Behav ; 215: 112787, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31866232

RESUMEN

Phytoestrogens are plant-derived compounds that can modulate estrogen activity in the brain and periphery. Laboratory rodent diets are typically high in soy-based phytoestrogens and therefore may influence neurophysiological and behavioural measures that are sensitive to estrogen signaling. Here we assessed such measures in rats (males and females) fed Australian made diets that varied in their soy levels. We found that a low-soy diet promoted greater weight, and lower levels of plasma estradiol, particularly in male rats. It also produced sex-specific effects on estrogen receptor gene expression in the brain, increasing ESR2 expression in the hippocampus and prefrontal cortex in female rats, and decreasing dopamine D1 receptor gene expression in the striatum of both male and female rats. We also found a dietary effect on short-term place recognition memory, but this was independent of soy levels in the diet. These results demonstrate that the choice of rodent laboratory diet can influence physiology, neurobiology and behavior, particularly on measures related to estrogen signaling.


Asunto(s)
Dieta , Estrógenos/fisiología , Transducción de Señal/fisiología , Memoria Espacial/fisiología , Animales , Peso Corporal/efectos de los fármacos , Ciclo Estral , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Fitoestrógenos/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D1/genética , Caracteres Sexuales , Alimentos de Soja
5.
J Neurosci ; 39(48): 9546-9559, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31628176

RESUMEN

Sensory cortices process stimuli in manners essential for perception. Very little is known regarding interactions between olfactory cortices. The piriform "primary" olfactory cortex, especially its anterior division (aPCX), extends dense association fibers into the ventral striatum's olfactory tubercle (OT), yet whether this corticostriatal pathway is capable of shaping OT activity, including odor-evoked activity, is unknown. Further unresolved is the synaptic circuitry and the spatial localization of OT-innervating PCX neurons. Here we build upon standing literature to provide some answers to these questions through studies in mice of both sexes. First, we recorded the activity of OT neurons in awake mice while optically stimulating principal neurons in the aPCX and/or their association fibers in the OT while the mice were delivered odors. This uncovered evidence that PCX input indeed influences OT unit activity. We then used patch-clamp recordings and viral tracing to determine the connectivity of aPCX neurons upon OT neurons expressing dopamine receptor types D1 or D2, two prominent cell populations in the OT. These investigations uncovered that both populations of neurons receive monosynaptic inputs from aPCX glutamatergic neurons. Interestingly, this input originates largely from the ventrocaudal aPCX. These results shed light on some of the basic physiological properties of this pathway and the cell-types involved and provide a foundation for future studies to identify, among other things, whether this pathway has implications for perception.SIGNIFICANCE STATEMENT Sensory cortices interact to process stimuli in manners considered essential for perception. Very little is known regarding interactions between olfactory cortices. The present study sheds light on some of the basic physiological properties of a particular intercortical pathway in the olfactory system and provides a foundation for future studies to identify, among other things, whether this pathway has implications for perception.


Asunto(s)
Ácido Glutámico/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Tubérculo Olfatorio/metabolismo , Corteza Piriforme/metabolismo , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Odorantes , Neuronas Receptoras Olfatorias/efectos de los fármacos , Tubérculo Olfatorio/efectos de los fármacos , Corteza Piriforme/efectos de los fármacos , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/genética , Olfato/fisiología
6.
eNeuro ; 6(5)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31541002

RESUMEN

The GABAergic medium-size spiny neuron (MSN), the striatal output neuron, may be classified into striosome, also known as patch, and matrix, based on neurochemical differences between the two compartments. At this time, little is known regarding the regulation of the development of the two compartments. Nr4a1, primarily described as a nuclear receptor/immediate early gene involved in the homeostasis of the dopaminergic system, is a striosomal marker. Using Nr4a1-overexpressing and Nr4a1-null mice, we sought to determine whether Nr4a1 is necessary and/or sufficient for striosome development. We report that in vivo and in vitro, Nr4a1 and Oprm1 mRNA levels are correlated. In the absence of Nr4a, there is a decrease in the percentage of striatal surface area occupied by striosomes. Alterations in Nr4a1 expression leads to dysregulation of multiple mRNAs of members of the dopamine receptor D1 signal transduction system. Constitutive overexpression of Nr4a1 decreases both the induction of phosphorylation of ERK after a single cocaine exposure and locomotor sensitization following chronic cocaine exposure. Nr4a1 overexpression increases MSN excitability but reduces MSN long-term potentiation. In the resting state, type 5 adenylyl cyclase (AC5) activity is normal, but the ability of AC5 to be activated by Drd1 G-protein-coupled receptor inputs is decreased. Our results support a role for Nr4a1 in determination of striatal patch/matrix structure and in regulation of dopaminoceptive neuronal function.


Asunto(s)
Cuerpo Estriado/metabolismo , Neuronas/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/biosíntesis , Receptores de Dopamina D1/biosíntesis , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Cocaína/farmacología , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Humanos , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/efectos de los fármacos , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Transducción de Señal/efectos de los fármacos
7.
eNeuro ; 6(5)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31451604

RESUMEN

Genetically modified mice have become standard tools in neuroscience research. Our understanding of the basal ganglia in particular has been greatly assisted by BAC mutants with selective transgene expression in striatal neurons forming the direct or indirect pathways. However, for more sophisticated behavioral tasks and larger intracranial implants, rat models are preferred. Furthermore, BAC lines can show variable expression patterns depending upon genomic insertion site. We therefore used CRISPR/Cas9 to generate two novel knock-in rat lines specifically encoding Cre recombinase immediately after the dopamine D1 receptor (Drd1a) or adenosine 2a receptor (Adora2a) loci. Here, we validate these lines using in situ hybridization and viral vector mediated transfection to demonstrate selective, functional Cre expression in the striatal direct and indirect pathways, respectively. We used whole-genome sequencing to confirm the lack of off-target effects and established that both rat lines have normal locomotor activity and learning in simple instrumental and Pavlovian tasks. We expect these new D1-Cre and A2a-Cre rat lines will be widely used to study both normal brain functions and neurological and psychiatric pathophysiology.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Integrasas/genética , Receptor de Adenosina A2A/genética , Receptores de Dopamina D1/genética , Animales , Femenino , Técnicas de Sustitución del Gen/métodos , Integrasas/biosíntesis , Masculino , Ratas , Ratas Long-Evans , Ratas Transgénicas , Receptor de Adenosina A2A/biosíntesis , Receptores de Dopamina D1/biosíntesis
8.
J Neuroimmunol ; 332: 233-241, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30954278

RESUMEN

CD8+ T regulatory/suppressor cells (Treg) affect peripheral tolerance and may be involved in autoimmune diseases as well as in cancer. In view of our previous data showing the ability of DA to affect adaptive immune responses, we investigated the dopaminergic phenotype of human CD8+ Treg as well as the ability of DA to affect their generation and activity. Results show that CD8+ T cells express both D1-like and D2-like dopaminergic receptors (DR), tyrosine hydroxylase (TH), the rate-limiting enzyme in the synthesis of DA, and vesicular monoamine transporter (VMAT) 2 and contain high levels of intracellular DA. Preferential upregulation of DR mRNA levels in the CD8+CD28- T cell compartment occurs during generation of CD8+ Treg, which is reduced by DA and by the D1-like DR agonist SKF-38393. DA and SKF-38393 also reduce the suppressive activity of CD8+ Treg on human peripheral blood mononuclear cells. Treg are crucial for tumor escape from the host immune system, thus the ability of DA to inhibits Treg function supports dopaminergic pathways as a druggable targets to develop original and innovative antitumor strategies.


Asunto(s)
Dopamina/farmacología , Receptores de Dopamina D1/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Citometría de Flujo , Humanos , Neuroinmunomodulación/fisiología , ARN Mensajero/biosíntesis , Receptores de Dopamina D1/análisis , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D1/genética , Linfocitos T Reguladores/metabolismo , Tirosina 3-Monooxigenasa/biosíntesis , Tirosina 3-Monooxigenasa/genética , Proteínas de Transporte Vesicular de Monoaminas/biosíntesis , Proteínas de Transporte Vesicular de Monoaminas/genética
9.
Physiol Behav ; 201: 95-103, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30553896

RESUMEN

Overconsumption of sugars contributes to poor health outcomes. Sugars are often added to commercial foods and beverages in low concentrations and these hidden sugars are consumed unnoticed, continuously. These hidden sugars are suggested to increase the motivation for foodstuffs with higher sugar contents, due to their rewarding properties. This process has been attributed in part, to the activity of both dopaminergic and opioidergic systems in the nucleus accumbens. We asked the question whether prolonged continuous consumption of a low concentration sucrose solution was sufficient to trigger alterations in both dopaminergic and opioidergic systems in the nucleus accumbens of male Sprague-Dawley rats. Rats were given access to either, 1% sucrose and water ad libitum for 3 weeks, or water alone, we then assayed the nucleus accumbens for mRNA and protein expression levels of D1 and D2 dopamine receptors which mediate appetitive motivation and wanting behaviors and for µ-opioid receptors which mediate liking of rewarding stimuli. Our data revealed that rats express a strong preference for 1% sucrose, and showed increased µ-opioid receptor mRNA expression bilaterally in the nucleus accumbens; increased D1 receptor mRNA expression in the left nucleus accumbens; and increased D2 receptor mRNA expression and decreased D2 receptor protein expression in the right nucleus accumbens. We also noted clear individual differences in the volumes of sucrose ingested over this period, however these differences did not correlate with the changes in neurochemistry. Our data show that prolonged ad libitum access to low concentration sucrose alters brain circuits critical for coding reward which may contribute to an enhanced drive for sweet foods and beverages.


Asunto(s)
Núcleo Accumbens/química , Sacarosa/efectos adversos , Animales , Apetito/efectos de los fármacos , Glucemia/metabolismo , Masculino , Motivación/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis , Receptores Opioides/fisiología , Receptores Opioides mu/efectos de los fármacos , Recompensa
10.
Neuropsychopharmacology ; 44(4): 805-816, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30504927

RESUMEN

Vulnerability for cocaine abuse in humans is associated with low dopamine D2 receptor (D2R) availability in the striatum. The mechanisms driving this vulnerability are poorly understood. In this study, we found that downregulating D2R expression selectively in striatal indirect-pathway neurons triggers a multitude of changes in D1 receptor (D1R)-expressing direct-pathway neurons, which comprise the other main subpopulation of striatal projection neurons. These changes include a leftward shift in the dose-response to a D1-like agonist that indicates a behavioral D1R hypersensitivity, a shift from PKA to ERK intracellular signaling cascades upon D1R activation, and a reduction in the density of bridging collaterals from D1R-expressing neurons to pallidal areas. We hypothesize that the D1R hypersensitivity underlies abuse vulnerability by facilitating the behavioral responses to repeated cocaine, such as locomotor sensitization and drug self-administration. We found evidence that littermate control mice develop D1R hypersensitivity after they are sensitized to cocaine. Indeed, D1-like agonist and cocaine cross-sensitize in control littermates and this effect was potentiated in mice lacking striatal D2Rs from indirect-pathway neurons. To our surprise, mice with low striatal D2Rs acquired cocaine self-administration similarly to littermate controls and showed no significant change in motivation to take cocaine but lower seeking. These findings indicate that downregulation of striatal D2Rs triggers D1R hypersensitivity to facilitate cocaine locomotor sensitization, which by itself was not associated with greater cocaine taking or seeking under the conditions tested.


Asunto(s)
Sensibilización del Sistema Nervioso Central/fisiología , Cocaína/farmacología , Cuerpo Estriado/metabolismo , Locomoción/efectos de los fármacos , Receptores de Dopamina D1/fisiología , Receptores de Dopamina D2/fisiología , Animales , Benzazepinas/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Noqueados , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis , Autoadministración , Potenciales Sinápticos/fisiología
11.
Neuropharmacology ; 141: 249-259, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30172845

RESUMEN

Identifying biological markers predicting vulnerability to develop excessive alcohol consumption may lead to a real improvement of clinical care. With converging evidence suggesting that gut microbiome is capable of influencing brain and behavior, this study aimed at investigating whether changes in gut microbiome composition is associated with conditioned responses to alcohol. We trained Wistar rats to self-administer alcohol for a prolonged period before screening those exhibiting uncontrolled alcohol seeking and taking by modeling diagnostic criteria for AUD: inability to abstain during a signaled period of reward unavailability, increased motivation assessed in a progressive effortful task and persistent alcohol intake despite aversive foot shocks. Based on addiction criteria scores, rats were assigned to either Vulnerable or Resilient groups. Vulnerable rats not only displayed increased impulsive and compulsive behaviors, but also displayed increased relapse after abstinence and increased sensitivity to baclofen treatments compared to resilient animals. Then, rats underwent a 3-month wash out period before sacrifice. Dorsal striatum was collected to assess dopamine receptor mRNA expression, and 16S microbiome sequencing was performed on caecal contents. Multiple significant correlations were found between gut microbiome and impulsivity measures, as well as augmentations in striatal Dopamine 1 receptor (D1R) and reductions in D2R as vulnerability to AUD increased. Therefore, using a singular translational approach based on biobehavioral dispositions to excessive alcohol seeking without heavy intoxication, our observations suggests an association between gut microbiome composition and these specific "at risk" behavioral traits observed in our translationally relevant model.


Asunto(s)
Conducta Compulsiva/fisiopatología , Cuerpo Estriado/fisiología , Comportamiento de Búsqueda de Drogas/fisiología , Etanol/administración & dosificación , Microbioma Gastrointestinal/fisiología , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis , Animales , Baclofeno/farmacología , Ciego/microbiología , Cuerpo Estriado/metabolismo , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/farmacología , Extinción Psicológica/efectos de los fármacos , Masculino , Motivación/efectos de los fármacos , Ratas , Autoadministración
12.
Neuropsychopharmacology ; 43(12): 2383-2389, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29982266

RESUMEN

Transient upregulation of GluN2B-containing NMDA receptors (R) in the nucleus accumbens (NAc) is proposed as an intermediate to long-term AMPAR plasticity associated with persistent cocaine-related behaviors. However, cell type- and input-specific contributions of GluN2B underlying lasting actions of cocaine remain to be elucidated. We utilized GluN2B cell type-specific knockouts and optogenetics to deconstruct the role of GluN2B in cocaine-induced NAc synaptic and behavioral plasticity. While reward learning was unaffected, loss of GluN2B in D1 dopamine receptor-expressing cells (D1) led to prolonged retention of reward memory. In control mice, prefrontal cortex (PFC)-D1(+) NAc AMPAR function was unaffected by cocaine exposure, while midline thalamus (mThal)-D1(+) NAc AMPAR function was potentiated but diminished after withdrawal. In D1-GluN2B-/- mice, the potentiation of mThal-D1(+) NAc AMPAR function persisted following withdrawal, corresponding with continued expression of cocaine reward behavior. These data suggest NAc GluN2B-containing NMDARs serve a feedback role and may weaken reward-related memories.


Asunto(s)
Cocaína/administración & dosificación , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/biosíntesis , Receptores de N-Metil-D-Aspartato/deficiencia , Recompensa , Tálamo/metabolismo , Animales , Eliminación de Gen , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleo Accumbens/efectos de los fármacos , Receptores de Dopamina D1/genética , Receptores de N-Metil-D-Aspartato/genética , Tálamo/efectos de los fármacos
13.
Drug Alcohol Depend ; 188: 113-118, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29772497

RESUMEN

BACKGROUND: The association with opioid-abusing individuals or even the perception of opioid abuse by peers are risk factors for the initiation and escalation of abuse. Similarly, we demonstrated that morphine-treated animals housed with only morphine-treated animals (referred to as morphine only) acquire morphine conditioned place-preference (CPP) more readily than morphine-treated animals housed with drug-naïve animals (referred to as morphine cage-mates). However, the molecular mechanisms underlying these effects are still elusive. METHODS: Mice received repeated morphine or saline while housed as saline only, morphine only, or cage-mates. Then, they were examined for the expression levels of D1 dopamine receptor (D1DR), D2 dopamine receptor (D2DR), dopamine transporter (DAT), oxytocin, and Arginine-vasopressin (AVP) in the striatum using qPCR. Additionally, we examined the effects of the AVP-V1b receptor antagonist, SSR149415, on the acquisition of morphine conditioned place-preference (CPP). RESULTS: Increased striatal expression of D1DR and AVP was observed in morphine only animals, but not morphine cage-mates. No significant effects were observed on the striatal expression of D2DR, DAT, or oxytocin. Antagonizing the AVP-V1b receptors decreased the acquisition of morphine CPP in the morphine only mice, but did not alter the acquisition of morphine CPP in the morphine cage-mate mice. CONCLUSIONS: Housing with drug-naïve animals protects against the increase in striatal expression of D1DR and AVP elicited by morphine exposure. Moreover, our studies suggest that the protective effect of housing with drug-naïve animals on the acquisition of morphine reward might be, at least partially, mediated by AVP.


Asunto(s)
Arginina Vasopresina/biosíntesis , Vivienda para Animales , Morfina/administración & dosificación , Receptores de Dopamina D1/biosíntesis , Recompensa , Conducta Social , Animales , Arginina Vasopresina/antagonistas & inhibidores , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Dopamina D1/antagonistas & inhibidores
14.
Respir Res ; 19(1): 53, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29606146

RESUMEN

Background: Dopamine receptors comprise two subgroups, Gs protein-coupled "D1-like" receptors (D1, D5) and Gicoupled "D2-like" receptors (D2, D3, D4). In airways, both dopamine D1 and D2 receptors are expressed on airway smooth muscle and regulate airway smooth muscle force. However, functional expression of the dopamine D1 receptor has never been identified on airway epithelium. Activation of Gs-coupled receptors stimulate adenylyl cyclase leading to cyclic AMP (cAMP) production, which is known to induce mucus overproduction through the cAMP response element binding protein (CREB) in airway epithelial cells. We questioned whether the dopamine D1 receptor is expressed on airway epithelium, and whether it promotes CREB phosphorylation and MUC5AC expression. Methods: We evaluated the protein expression of the dopamine D1 receptor on native human airway epithelium and three sources of cultured human airway epithelial cells including primary cultured airway epithelial cells, the bronchial epithelial cell line (16HBE14o-), and the pulmonary mucoepidermoid carcinoma cell line (NCI-H292) using immunohistochemistry and immunoblotting. To characterize the stimulation of cAMP through the dopamine D1 receptor, 16HBE14o- cells and NCI-H292 cells were treated with dopamine or the dopamine D1 receptor agonists (SKF38393 or A68930) before cAMP measurements. The phosphorylation of CREB by A68930 in both 16HBE14o- and NCI-H292 cells was measured by immunoblot. The effect of dopamine or A68930 on the expression of MUC5AC mRNA and protein in NCI-H292 cells was evaluated by real-time PCR and immunofluorescence staining, respectively. Results: The dopamine D1 receptor protein was detected in native human airway epithelium and three sources of cultured human airway epithelial cells. Dopamine or the dopamine D1-like receptor agonists stimulated cAMP production in 16HBE14o- cells and NCI-H292 cells, which was reversed by the selective dopamine D1-like receptor antagonists (SCH23390 or SCH39166). A68930 significantly increased phosphorylation of CREB in both 16HBE14o- and NCI-H292 cells, which was attenuated by the inhibitors of PKA (H89) and MEK (U0126). Expression of MUC5AC mRNA and protein were also increased by either dopamine or A68930 in NCI-H292 cells. Conclusions: These results suggest that the activation of the dopamine D1 receptor on human airway epithelium could induce mucus overproduction, which could worsen airway obstructive symptoms.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Mucina 5AC/biosíntesis , Receptores de Dopamina D1/biosíntesis , Mucosa Respiratoria/metabolismo , Línea Celular , Células Cultivadas , Agonistas de Dopamina/farmacología , Expresión Génica , Humanos , Mucina 5AC/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/genética , Mucosa Respiratoria/efectos de los fármacos
15.
Nat Commun ; 9(1): 1576, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29679009

RESUMEN

Nucleus accumbens (NAc) is involved in behaviors that depend on heightened wakefulness, but its impact on arousal remains unclear. Here, we demonstrate that NAc dopamine D1 receptor (D1R)-expressing neurons are essential for behavioral arousal. Using in vivo fiber photometry in mice, we find arousal-dependent increases in population activity of NAc D1R neurons. Optogenetic activation of NAc D1R neurons induces immediate transitions from non-rapid eye movement sleep to wakefulness, and chemogenetic stimulation prolongs arousal, with decreased food intake. Patch-clamp, tracing, immunohistochemistry, and electron microscopy reveal that NAc D1R neurons project to the midbrain and lateral hypothalamus, and might disinhibit midbrain dopamine neurons and lateral hypothalamus orexin neurons. Photoactivation of terminals in the midbrain and lateral hypothalamus is sufficient to induce wakefulness. Silencing of NAc D1R neurons suppresses arousal, with increased nest-building behaviors. Collectively, our data indicate that NAc D1R neuron circuits are essential for the induction and maintenance of wakefulness.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Área Hipotalámica Lateral/fisiología , Mesencéfalo/fisiología , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo , Vigilia/fisiología , Animales , Ritmo Circadiano/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Placa-Clamp , Fotometría/métodos , Receptores de Dopamina D1/biosíntesis , Sueño/fisiología
16.
J Neurosci ; 37(49): 11894-11911, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29089442

RESUMEN

Exposure to cocaine-associated contextual cues contributes significantly to relapse. Extinction of these contextual associations, which involves a new form of learning, reduces cocaine-seeking behavior; however, the molecular mechanisms underlying this process remain largely unknown. We report that extinction, but not acquisition, of cocaine conditioned place preference (CPP) in male mice increased Cav1.2 L-type Ca2+ channel mRNA and protein in postsynaptic density (PSD) fractions of the hippocampus, a brain region involved in drug-context associations. Moreover, viral-mediated deletion of Cav1.2 in the dorsal hippocampus attenuated extinction of cocaine CPP. Molecular studies examining downstream Cav1.2 targets revealed that extinction recruited calcium/calmodulin (Ca2+/CaMK)-dependent protein kinase II (CaMKII) to the hippocampal PSD. This occurred in parallel with an increase in phosphorylation of the AMPA GluA1 receptor subunit at serine 831 (S831), a CaMKII site, along with an increase in total PSD GluA1. The necessity of S831 GluA1 was further demonstrated by the lack of extinction in S831A GluA1 phosphomutant mice. Of note hippocampal GluA1 levels remained unaltered at the PSD, but were reduced near the PSD and at perisynaptic sites of dendritic spines in extinction-resistant S831A mutant mice. Finally, conditional knock-out of Cav1.2 in dopamine D1 receptor (D1R)-expressing cells resulted in attenuation of cocaine CPP extinction and lack of extinction-dependent changes in hippocampal PSD CaMKII expression and S831 GluA1 phosphorylation. In summary, we demonstrate an essential role for the hippocampal Cav1.2/CaMKII/S831 GluA1 pathway in cocaine CPP extinction, with data supporting contribution of hippocampal D1R-expressing cells in this process. These findings demonstrate a novel role for Cav1.2 channels in extinction of contextual cocaine-associated memories.SIGNIFICANCE STATEMENT Continued drug-seeking behavior, a defining characteristic of cocaine addiction, can be precipitated by contextual cues, yet the molecular mechanisms required for extinction of these context-specific memories remain poorly understood. Here, we have uncovered a novel and selective role of the Cav1.2 L-type Ca2+ channel and its downstream signaling pathway in the hippocampus that mediate extinction of cocaine conditioned place preference (CPP). We additionally provide evidence that supports a role of Cav1.2 within dopamine D1 receptor-expressing cells of the hippocampus for extinction of cocaine CPP. Therefore, these findings reveal a previously unknown role of Cav1.2 channels within the hippocampus and in D1 receptor-expressing cells in extinction of cocaine-associated memories, providing a framework for further exploration of mechanisms underlying extinction of cocaine-seeking behavior.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Cocaína/administración & dosificación , Extinción Psicológica/fisiología , Hipocampo/metabolismo , Memoria/fisiología , Receptores de Dopamina D1/biosíntesis , Animales , Trastornos Relacionados con Cocaína/metabolismo , Extinción Psicológica/efectos de los fármacos , Expresión Génica , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Dopamina D1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Método Simple Ciego
17.
J Neurosci ; 37(49): 11930-11946, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29097596

RESUMEN

We have previously shown that casein kinase 2 (CK2) negatively regulates dopamine D1 and adenosine A2A receptor signaling in the striatum. Ablation of CK2 in D1 receptor-positive striatal neurons caused enhanced locomotion and exploration at baseline, whereas CK2 ablation in D2 receptor-positive neurons caused increased locomotion after treatment with A2A antagonist, caffeine. Because both, D1 and A2A receptors, play major roles in the cellular responses to l-DOPA in the striatum, these findings prompted us to examine the impact of CK2 ablation on the effects of l-DOPA treatment in the unilateral 6-OHDA lesioned mouse model of Parkinson's disease. We report here that knock-out of CK2 in striatonigral neurons reduces the severity of l-DOPA-induced dyskinesia (LID), a finding that correlates with lowered pERK but unchanged pPKA substrate levels in D1 medium spiny neurons as well as in cholinergic interneurons. In contrast, lack of CK2 in striatopallidal neurons enhances LID and ERK phosphorylation. Coadministration of caffeine with a low dose of l-DOPA reduces dyskinesia in animals with striatopallidal knock-out to wild-type levels, suggesting a dependence on adenosine receptor activity. We also detect reduced Golf levels in the striatonigral but not in the striatopallidal knock-out in response to l-DOPA treatment.Our work shows, in a rodent model of PD, that treatment-induced dyskinesia and striatal ERK activation are bidirectionally modulated by ablating CK2 in D1- or D2-positive projection neurons, in male and female mice. The results reveal that CK2 regulates signaling events critical to LID in each of the two main populations of striatal neurons.SIGNIFICANCE STATEMENT To date, l-DOPA is the most effective treatment for PD. Over time, however, its efficacy decreases, and side effects including l-DOPA-induced dyskinesia (LID) increase, affecting up to 78% of patients within 10 years of therapy (Hauser et al., 2007). It is understood that supersensitivity of the striatonigral pathway underlies LID, however, D2 agonists were also shown to induce LID (Bezard et al., 2001; Delfino et al., 2004). Our work implicates a novel player in the expression of LID, the kinase CK2: knock-out of CK2 in striatonigral and striatopallidal neurons has opposing effects on LID. The bidirectional modulation of dyskinesia reveals a central role for CK2 in striatal physiology and indicates that both pathways contribute to LID.


Asunto(s)
Quinasa de la Caseína II/fisiología , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/metabolismo , Discinesia Inducida por Medicamentos/metabolismo , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis , Animales , Quinasa de la Caseína II/deficiencia , Cuerpo Estriado/efectos de los fármacos , Agonistas de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Discinesia Inducida por Medicamentos/genética , Femenino , Expresión Génica , Levodopa/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/genética
18.
Toxicol Appl Pharmacol ; 328: 46-53, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28479505

RESUMEN

During early development in sea urchins, classical neurotransmitters, including acetylcholine (ACh), dopamine (DA), and serotonin (5-HT), play important roles in the regulation of morphogenesis and swimming behavior. However, the underlying mechanisms of how organophosphate pesticides cause developmental neurotoxicity by interfering with different neurotransmitter systems are unclear. In this study, we investigated the effects of 0.01, 0.10, and 1.00mg/L monocrotophos (MCP) pesticide on the activity of acetyltransferase (ChAT), acetylcholinesterase (AChE), monoamine oxidase, the concentration of DA, dopamine transporter, and the transcription activity of DA receptor D1 and tyrosine hydroxylase, during critical periods in cholinergic and dopaminergic nervous system development in sea urchin (Hemicentrotus pulcherrimus) embryos and larvae. At the blastula stages, MCP disrupted DA metabolism but not 5-HT metabolism, resulting in abnormal development. High ChAT and AChE activity were observed at the gastrulation-completed stage and the two-armed pluteus stage, respectively, MCP inhibited ChAT activity and AChE activity/distribution and resulted in developmental defects of the plutei. From the gastrula stage to the two-armed pluteus stage, we found ubiquitous disrupting effects of MCP on ACh, DA, and 5-HT metabolism, particularly at critical periods during the development of these neurotransmitter systems. Therefore, we propose that this disruption is one of the main mechanisms of MCP-related developmental neurotoxicity, which would contribute better understanding insight into the mechanism of MCP pesticide's toxic effects.


Asunto(s)
Dopamina/metabolismo , Hemicentrotus , Insecticidas/toxicidad , Monocrotofos/toxicidad , Síndromes de Neurotoxicidad/metabolismo , Neurotransmisores/metabolismo , Sistema Nervioso Parasimpático/efectos de los fármacos , Acetilcolinesterasa/metabolismo , Animales , Colina O-Acetiltransferasa/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Gastrulación , Hemicentrotus/crecimiento & desarrollo , Monoaminooxidasa/metabolismo , Síndromes de Neurotoxicidad/patología , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D1/genética , Serotonina/metabolismo , Natación , Tirosina 3-Monooxigenasa/biosíntesis , Tirosina 3-Monooxigenasa/genética
19.
Int J Neuropsychopharmacol ; 20(3): 257-268, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815417

RESUMEN

Background: Chronic abuse of heroin leads to long-lasting and complicated cognitive impairment. Dopamine receptors are critically involved in the impulsive drug-driven behavior and the altered attention, processing speed, and mental flexibility that are associated with higher relapse rates. However, the effects of the different dopamine receptors and their possible involvement in heroin-induced cognitive impairment remain unclear. Methods: The 5-choice serial reaction time task was used to investigate the profiles of heroin-induced cognitive impairment in mice. The expression levels of dopamine D1- and D2-like receptors in the prefrontal cortex, nucleus accumbens, and caudate-putamen were determined. The effects of dopamine receptors on heroin-induced impulsivity in the 5-choice serial reaction time task were examined by agonist/antagonist treatment on D1 or D3 receptor mutant mice. Results: Systemic heroin administration influences several variables in the 5-choice serial reaction time task, most notably premature responses, a measure of motor impulsivity. These behavioral impairments are associated with increased D1 receptor and decreased D3 receptor mRNA and protein levels in 3 observed brain areas. The heroin-evoked increase in premature responses is mimicked by a D1 agonist and prevented by a D1 antagonist or genetic ablation of the D1 receptor gene. In contrast, a D3 agonist decreases both basal and heroin-evoked premature responses, while genetic ablation of the D3 receptor gene results in increased basal and heroin-evoked premature responses. Conclusions: Heroin-induced impulsive behavior in the 5-choice serial reaction time task is oppositely modulated by D1 and D3 receptor activation. The D1 receptors in the cortical-mesolimbic region play an indispensable role in modulating such behaviors.


Asunto(s)
Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/fisiopatología , Conducta Impulsiva/efectos de los fármacos , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D1/fisiología , Receptores de Dopamina D3/biosíntesis , Receptores de Dopamina D3/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Animales , Anticuerpos/farmacología , Benzazepinas/farmacología , Benzopiranos/farmacología , Heroína , Masculino , Ratones , Mutación , Neostriado/metabolismo , Núcleo Accumbens , Oxazinas/farmacología , Corteza Prefrontal/metabolismo , Tiempo de Reacción/efectos de los fármacos , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D2/biosíntesis , Receptores de Dopamina D3/agonistas , Receptores de Dopamina D3/genética
20.
Eur Neuropsychopharmacol ; 26(11): 1794-1805, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27642078

RESUMEN

It was previously shown that mice with genetic deletion of the neurotrophic factor pleiotrophin (PTN-/-) show enhanced amphetamine neurotoxicity and impair extinction of amphetamine conditioned place preference (CPP), suggesting a modulatory role of PTN in amphetamine neurotoxicity and reward. We have now studied the effects of amphetamine (10mg/kg, 4 times, every 2h) in the striatum of mice with transgenic PTN overexpression (PTN-Tg) in the brain and in wild type (WT) mice. Amphetamine caused an enhanced loss of striatal dopaminergic terminals, together with a highly significant aggravation of amphetamine-induced increase in the number of GFAP-positive astrocytes, in the striatum of PTN-Tg mice compared to WT mice. Given the known contribution of D1 and D2 dopamine receptors to the neurotoxic effects of amphetamine, we also performed quantitative receptor autoradiography of both receptors in the brains of PTN-Tg and WT mice. D1 and D2 receptors binding in the striatum and other regions of interest was not altered by genotype or treatment. Finally, we found that amphetamine CPP was significantly reduced in PTN-Tg mice. The data demonstrate that PTN overexpression in the brain blocks the conditioning effects of amphetamine and enhances the characteristic striatal dopaminergic denervation caused by this drug. These results indicate for the first time deleterious effects of PTN in vivo by mechanisms that are probably independent of changes in the expression of D1 and D2 dopamine receptors. The data also suggest that PTN-induced neuroinflammation could be involved in the enhanced neurotoxic effects of amphetamine in the striatum of PTN-Tg mice.


Asunto(s)
Anfetamina/farmacología , Proteínas Portadoras/biosíntesis , Estimulantes del Sistema Nervioso Central/farmacología , Cuerpo Estriado/metabolismo , Citocinas/biosíntesis , Neuronas Dopaminérgicas/efectos de los fármacos , Inflamación/metabolismo , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Autorradiografía , Proteínas Portadoras/genética , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Citocinas/genética , Desnervación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...