Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 378(2): 133-145, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34011529

RESUMEN

Opioid use disorder (OUD) is a major socioeconomic burden. An ideal OUD pharmacotherapy will mitigate the suffering associated with opioid-withdrawal, inhibit the effects of high efficacy opioids, and minimize opioid-cravings while being safe and accessible to a diverse patient population. Although current OUD pharmacotherapies inhibit the euphoric effects of opioids of abuse, the extent to which they safely alleviate withdrawal and opioid-cravings corresponds with their intrinsic µ opioid receptor (MOR) efficacy. In addition to inhibiting the euphoric effects of opioids of abuse, the medium efficacy MOR agonist buprenorphine alleviates withdrawal and opioid-cravings, but its intrinsic MOR efficacy is sufficient such that its utility is limited by abuse and safety liabilities. Although the MOR antagonist naltrexone minimizes euphoria and has no abuse liability, it exacerbates suffering associated with withdrawal and opioid cravings. Therefore, a therapeutic with intrinsic MOR activity between the partial agonist (buprenorphine) and the antagonist (naltrexone) would strike a balance between the benefits and liabilities of these two therapeutics. To address this need, we derived RM1490, an MOR agonist based on a nonmorphinan scaffold that exhibits approximately half the intrinsic MOR efficacy of buprenorphine. In a series of preclinical assays, we compared RM1490 with buprenorphine and naltrexone at doses that achieve therapeutic levels of central nervous system MOR occupancy. RM1490 exhibited a behavioral profile consistent with reduced reward, dependence, and precipitated withdrawal liabilities. RM1490 was also more effective than buprenorphine at reversing the respiratory depressant effects of fentanyl and did not suppress respiration when combined with diazepam. SIGNIFICANCE STATEMENT: In preclinical studies, RM1490 has a physiological and behavioral profile suitable for opioid use disorder maintenance therapy.


Asunto(s)
Trastornos Relacionados con Opioides , Buprenorfina , Naltrexona
2.
J Pharmacol Exp Ther ; 360(1): 117-128, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27811173

RESUMEN

Allosteric potentiators amplify the sensitivity of physiologic control circuits, a mode of action that could provide therapeutic advantages. This hypothesis was tested with the dopamine D1 receptor potentiator DETQ [2-(2,6-dichlorophenyl)-1-((1S,3R)-3-(hydroxymethyl)-5-(2-hydroxypropan-2-yl)-1-methyl-3,4-dihydroisoquinolin-2(1H)-yl)ethan-1-one]. In human embryonic kidney 293 (HEK293) cells expressing the human D1 receptor, DETQ induced a 21-fold leftward shift in the cAMP response to dopamine, with a Kb of 26 nM. The maximum response to DETQ alone was ∼12% of the maximum response to dopamine, suggesting weak allosteric agonist activity. DETQ was ∼30-fold less potent at rat and mouse D1 receptors and was inactive at the human D5 receptor. To enable studies in rodents, an hD1 knock-in mouse was generated. DETQ (3-20 mg/kg orally) caused a robust (∼10-fold) increase in locomotor activity (LMA) in habituated hD1 mice but was inactive in wild-type mice. The LMA response to DETQ was blocked by the D1 antagonist SCH39166 and was dependent on endogenous dopamine. LMA reached a plateau at higher doses (30-240 mg/kg) even though free brain levels of DETQ continued to increase over the entire dose range. In contrast, the D1 agonists SKF 82958, A-77636, and dihydrexidine showed bell-shaped dose-response curves with a profound reduction in LMA at higher doses; video-tracking confirmed that the reduction in LMA caused by SKF 82958 was due to competing stereotyped behaviors. When dosed daily for 4 days, DETQ continued to elicit an increase in LMA, whereas the D1 agonist A-77636 showed complete tachyphylaxis by day 2. These results confirm that allosteric potentiators may have advantages compared with direct-acting agonists.


Asunto(s)
Conducta Animal/efectos de los fármacos , Técnicas de Sustitución del Gen , Isoquinolinas/farmacología , Locomoción/efectos de los fármacos , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Taquifilaxis , Adamantano/análogos & derivados , Adamantano/farmacología , Regulación Alostérica/efectos de los fármacos , Animales , Benzopiranos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Isoquinolinas/efectos adversos , Masculino , Ratones , Transporte de Proteínas/efectos de los fármacos , Receptores de Dopamina D1/agonistas
3.
J Pharmacol Exp Ther ; 348(1): 192-201, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24163440

RESUMEN

Weight gain and diabetes have been reported during treatment with atypical antipsychotic drugs (AAPDs). Patients treated with the glucocorticoid receptor antagonist (GRA) and the progesterone receptor antagonist (PRA) mifepristone [estra-4,9-dien-3-one, 11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(1-propynyl)-(11ß,17ß)-(9CI)] experienced significant reduction in the weight gain observed when patients were treated with olanzapine or risperidone. To understand the pharmacology responsible for this finding, we discovered LLY-2707 [N-(5-(tert-butyl)-3-(2-fluoro-5-methylpyridin-4-yl)-2-methyl-1H-indol-7-yl)methanesulfonamide], a novel and selective GRA, and evaluated its utility in preclinical models of AAPD-associated weight gain and diabetes. In vitro, LLY-2707 was a highly selective and potent GRA. GR occupancy in vivo was assessed using ex vivo binding where LLY-2707 inhibited [(3)H]dexamethasone binding to the liver. Modest but statistically significant decreases in brain ex vivo binding were observed with high doses of CORT-108297 [(R)-4α-(ethoxymethyl)-1-(4-fluorophenyl)-6-((4-(trifluoromethyl)phenyl)sulfonyl)-4,4a,5,6,7,8-hexahydro-1H-pyrazolo[3,4-g]isoquinoline] and LLY-2707, but mifepristone inhibited at all doses. Central activity of the GRAs was confirmed by their ability to suppress amphetamine-induced increases in locomotor activity. The increases in the body weight of female rats treated with olanzapine (2 mg/kg PO) over 14 days were reduced in a dose-dependent manner by coadministration of LLY-2707. Similar decreases, although less robust, in body weight were seen with mifepristone and CORT-108297. In addition, sGRAs prevented the glucose excursion after intragastric olanzapine infusions consistent with a direct effect on the hyperglycemia observed during treatment with AAPDs. At doses effectively preventing weight gain, LLY-2707 did not substantially interfere with the dopamine D2 receptor occupancy by olanzapine. Therefore, GRA coadministration may provide a novel treatment modality to prevent the weight gain and diabetes observed during treatment with AAPDs.


Asunto(s)
Antipsicóticos/toxicidad , Indoles/farmacología , Receptores de Glucocorticoides/antagonistas & inhibidores , Sulfonamidas/farmacología , Aumento de Peso/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos , Animales , Compuestos Aza/química , Compuestos Aza/farmacología , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Femenino , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Indoles/química , Masculino , Ratones , Ratones Endogámicos C57BL , Mifepristona/química , Mifepristona/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/fisiología , Sulfonamidas/química , Aumento de Peso/fisiología , Pérdida de Peso/fisiología
4.
Front Psychiatry ; 14: 1170541, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37457777

RESUMEN

Low doses of µ-opioid receptor (MOR) agonists rapidly ameliorate symptoms in treatment-resistant obsessive-compulsive disorder (OCD) patients (10-50% of OCD patients). However, the utility of MOR agonists is limited by their safety liabilities. We developed a novel MOR partial agonist (EPD1540) that has an improved respiratory safety profile when compared to buprenorphine. Buprenorphine is a MOR partial agonist primarily used in the treatment of opiate-use disorder, which in investigator-led trials, has been shown to rapidly ameliorate symptoms in treatment-resistant OCD patients. In this study, we show that doses of EPD1504 and buprenorphine that occupy small fractions of MORs in the CNS (approximately 20%) are as effective as fluoxetine at ameliorating OCD-like behaviors in two different rat models (an operant probabilistic reversal task and marble burying). Importantly, effective doses of EPD1504 did not impair either locomotor activity, or respiration under normoxic or hypercapnic conditions. Additionally, EPD1504 had effects comparable to buprenorphine in the conditioned place preference assay. These results indicate that EPD1504 may provide a safer alternative to buprenorphine for the treatment of OCD patients.

5.
J Pharmacol Exp Ther ; 341(3): 672-80, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22402929

RESUMEN

Corticotropin-releasing factor (CRF) is the major hypothalamic neuropeptide responsible for stimulation of the hypothalamic-pituitary-adrenal axis (HPAA), resulting in the synthesis and release of glucocorticoids from the adrenal cortex. In a recent study, we reported the discovery of the CRF1 receptor antagonist, 3-(4-chloro-2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo[1,2-b]pyridazine (MTIP), which has efficacy in preclinical models of stress-induced alcohol consumption. Because CRF1 is important in HPAA activation, we evaluated the effects of MTIP administration on rodent HPAA function. Initial studies established the MTIP doses required for brain and pituitary CRF1 occupancy and those associated with the inhibition of intracerebroventricular CRF on the HPAA in mice. Then, rat basal plasma corticosterone (CORT) concentrations were measured hourly by radioimmunoassay for 24 h after three daily doses of MTIP or vehicle. In these studies, the early phase of the nocturnal CORT surge was reduced; however, the area under the CORT curve was identical for the 24-h period. In subsequent studies, increases in plasma CORT due to direct pharmacological manipulation of the HPAA axis or by stressors were evaluated after MTIP treatment in mice. MTIP attenuated CORT responses generated by immediate bolus administration of insulin or ethanol; however, MTIP did not affect activation of the HPAA by other stressors and pharmacological agents. Therefore, MTIP can modulate basal HPAA activity during the CORT surge and reduced activation after a select number of stressors but does not produce a lasting suppression of basal CORT. The ability of MTIP to modulate plasma CORT after hyperinsulinemia may provide a surrogate strategy for a target occupancy biomarker.


Asunto(s)
Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Piridazinas/farmacología , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Tiazoles/farmacología , Hormona Adrenocorticotrópica/sangre , Proteínas Anfibias/metabolismo , Animales , Área Bajo la Curva , Corticosterona/sangre , Infusiones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Hormonas Peptídicas/metabolismo , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley
6.
J Neurosci ; 28(4): 893-903, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18216197

RESUMEN

Resilience to mental and physical stress is a key determinant for the survival and functioning of mammals. Although the importance of stress resilience has been recognized, the underlying neural mediators have not yet been identified. Neuropeptide Y (NPY) is a peptide known for its anti-anxiety-like effects mediated via the amygdala. The results of our current study demonstrate, for the first time that repeated administration of NPY directly into the basolateral nucleus of the amygdala (BLA) produces selective stress-resilient behavioral responses to an acute restraint challenge as measured in the social interaction test, but has no effect on hypothalamic-adrenal-pituitary axis activity or stress-induced hyperthermia. More importantly, the resilient behaviors observed in the NPY-treated animals were present for up to 8 weeks. Antagonizing the activity of calcineurin, a protein phosphatase involved in neuronal remodeling and present in NPY receptor containing neurons within the BLA, blocked the development of long-term, but not the acute increases in social interaction responses induced by NPY administration. This suggests that the NPY-induced long-term behavioral resilience to restraint stress may occur via mechanisms involving neuronal plasticity. These studies suggest one putative physiologic mechanism underlying stress resilience and could identify novel targets for development of therapies that can augment the ability to cope with stress.


Asunto(s)
Fiebre/fisiopatología , Sistema Hipotálamo-Hipofisario/fisiología , Neuropéptido Y/administración & dosificación , Sistema Hipófiso-Suprarrenal/fisiología , Conducta Social , Estrés Psicológico/prevención & control , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Fiebre/tratamiento farmacológico , Fiebre/psicología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Masculino , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Ratas , Ratas Wistar , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología , Tiempo
7.
J Pharmacol Exp Ther ; 329(2): 429-38, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19182070

RESUMEN

The mammalian neuropeptide, melanin-concentrating hormone, interacts with two G protein-coupled receptors, melanin-concentrating hormone receptor (MCHR) 1 and MCHR2; however, only MCHR1 is expressed in rats and mice. In the present study, we evaluated MCHR1 antagonism in preclinical models believed to be predictive of antiobesity and antidepressant activity. Central activity of the selective MCHR1 antagonist, GW803430 [6-(4-chloro-phenyl)-3-[3-methoxy-4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-3H-thieno[3,2-d]pyrimidin-4-one], was evaluated using ex vivo binding with autoradiography. Effective doses of GW803430 (1 and 3 mg/kg p.o.) were correlated with antiobesity activity in a 14-day study of diet-induced obese rats. GW803430 was evaluated subsequently for antidepressant-like effects in mice and rats. Acute and subchronic administration reduced immobility time in the mouse forced-swim test at doses of 3 (acute) and 3 and 10 (chronic) mg/kg p.o., an effect that was absent in MCHR1(-/-) mice. Combined subeffective doses of GW803430 (0.3 and 1 mg/kg p.o.) and imipramine (5 mg/kg) produced a robust antidepressant-like response. The compound was also active in the tail suspension test at a dose of 10 mg/kg p.o. GW803430 (30 mg/kg p.o.) significantly reduced submissive behaviors at weeks 2 and 3, a model of submissive behavior that may predict antidepressant onset. GW803430 decreased marble burying in mice at doses of 3, 10, and 30 mg/kg p.o., an assay that detects anxiolytic-like effects. Thus, GW803430 produces robust antiobesity and antidepressant-like effects in rats and mice at doses that compete for central MCHR1 in vivo. As such, MCHR1 should be considered as a promising target for future drug discovery efforts.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Pirimidinonas/uso terapéutico , Receptores de Somatostatina/antagonistas & inhibidores , Tiofenos/uso terapéutico , Animales , Fármacos Antiobesidad/administración & dosificación , Fármacos Antiobesidad/farmacología , Antidepresivos/administración & dosificación , Antidepresivos/farmacología , Autorradiografía , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Depresión/metabolismo , Depresión/fisiopatología , Depresión/psicología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Masculino , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Obesidad/metabolismo , Obesidad/fisiopatología , Obesidad/psicología , Unión Proteica , Pirimidinonas/administración & dosificación , Pirimidinonas/farmacología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Receptores de Somatostatina/genética , Predominio Social , Natación , Tiofenos/administración & dosificación , Tiofenos/farmacología
8.
J Neurosci ; 27(10): 2718-26, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344409

RESUMEN

We describe a novel corticotropin-releasing factor receptor 1 (CRF1) antagonist with advantageous properties for clinical development, and its in vivo activity in preclinical alcoholism models. 3-(4-Chloro-2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo[1,2-b]pyridazine (MTIP) inhibited 125I-sauvagine binding to rat pituitary membranes and cloned human CRF1 with subnanomolar affinities, with no detectable activity at the CRF2 receptor or other common drug targets. After oral administration to rats, MTIP inhibited 125I-sauvagine binding to rat cerebellar membranes ex vivo with an ED50 of approximately 1.3 mg/kg and an oral bioavailability of 91.1%. Compared with R121919 (2,5-dimethyl-3-(6-dimethyl-4-methylpyridin-3-yl)-7-dipropylamino-pyrazolo[1,5-a]pyrimidine) and CP154526 (N-butyl-N-ethyl-4,9-dimethyl-7-(2,4,6-trimethylphenyl)-3,5,7-triazabicyclo[4.3.0]nona-2,4,8,10-tetraen-2-amine), MTIP had a markedly reduced volume of distribution and clearance. Neither open-field activity nor baseline exploration of an elevated plus-maze was affected by MTIP (1-10 mg/kg). In contrast, MTIP dose-dependently reversed anxiogenic effects of withdrawal from a 3 g/kg alcohol dose. Similarly, MTIP blocked excessive alcohol self-administration in Wistar rats with a history of dependence, and in a genetic model of high alcohol preference, the msP rat, at doses that had no effect in nondependent Wistar rats. Also, MTIP blocked reinstatement of stress-induced alcohol seeking both in postdependent and in genetically selected msP animals, again at doses that were ineffective in nondependent Wistar rats. Based on these findings, MTIP is a promising candidate for treatment of alcohol dependence.


Asunto(s)
Alcoholismo/tratamiento farmacológico , Encéfalo/metabolismo , Piridazinas/farmacocinética , Piridazinas/uso terapéutico , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Tiazoles/farmacocinética , Tiazoles/uso terapéutico , Administración Oral , Consumo de Bebidas Alcohólicas/psicología , Alcoholismo/genética , Alcoholismo/psicología , Proteínas Anfibias , Animales , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Etanol/administración & dosificación , Masculino , Hormonas Peptídicas , Péptidos/antagonistas & inhibidores , Péptidos/metabolismo , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Piridazinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Autoadministración , Estrés Fisiológico/psicología , Síndrome de Abstinencia a Sustancias/psicología , Tiazoles/administración & dosificación
9.
Neuropeptides ; 42(3): 367-75, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18329096

RESUMEN

Neuropeptide Y (NPY) is an important central regulator of food consumption and energy expenditure via the hypothalamus. NPY containing neurons have a broad central distribution and are often colocalized with norepinephrine (NE). However, NPY deficient mice do not exhibit any substantial changes in food consumption, body weight or body composition when compared to wild type mice. Since NE and serotonin (5HT) are also important regulators of appetite and metabolism, we evaluated these systems in NPY deficient mice. Brain sections from NPY deficient and wild type mice were labeled with either (3)H-nisoxetine for the NE transporter (NET) or (3)H-citalopram for the 5HT transporter (SERT). Tyrosine hydroxylase expression was evaluated by radioimmunohistochemistry. Brain monoamines and metabolites were evaluated using HPLC. NPY deficient mice exhibited a substantial decrease in NET binding in most brain regions examined. NET binding was less than 50% of control binding in the cerebral cortex and subregions of the thalamus with the greatest decrease seen in the hypothalamus. In contrast, more modest and regionally variable changes were observed in the SERT binding with decreases in regions such as the accessory olfactory nucleus, glomerular layer of the olfactory bulb and the CA1 region of the hippocampus. Measurement of NE and 5HT content as well as the primary metabolites revealed increased NE turnover and decreased 5HT content in the hypothalamus. Therefore, developmental compensation by the NE and 5HT systems may contribute to the absence of a body weight phenotype in NPY deficient mice.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Química Encefálica/genética , Neuropéptido Y/deficiencia , Animales , Autorradiografía , Cromatografía Líquida de Alta Presión , Citalopram , Fluoxetina/análogos & derivados , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Neuropéptido Y/genética , Norepinefrina/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina , Tirosina 3-Monooxigenasa/biosíntesis , Tirosina 3-Monooxigenasa/genética
10.
J Neurosci ; 26(36): 9205-15, 2006 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-16957077

RESUMEN

Intravenous sodium lactate infusions or the noradrenergic agent yohimbine reliably induce panic attacks in humans with panic disorder but not in healthy controls. However, the exact mechanism of lactate eliciting a panic attack is still unknown. In rats with chronic disruption of GABA-mediated inhibition in the dorsomedial hypothalamus (DMH), achieved by chronic microinfusion of the glutamic acid decarboxylase inhibitor L-allylglycine, sodium lactate infusions or yohimbine elicits panic-like responses (i.e., anxiety, tachycardia, hypertension, and tachypnea). In the present study, previous injections of the angiotensin-II (A-II) type 1 receptor antagonist losartan and the nonspecific A-II receptor antagonist saralasin into the DMH of "panic-prone" rats blocked the anxiety-like and physiological components of lactate-induced panic-like responses. In addition, direct injections of A-II into the DMH of these panic-prone rats also elicited panic-like responses that were blocked by pretreatment with saralasin. Microinjections of saralasin into the DMH did not block the panic-like responses elicited by intravenous infusions of the noradrenergic agent yohimbine or by direct injections of NMDA into the DMH. The presence of the A-II type 1 receptors in the region of the DMH was demonstrated using immunohistochemistry. Thus, these results implicate A-II pathways and the A-II receptors in the hypothalamus as putative substrates for sodium lactate-induced panic-like responses in vulnerable subjects.


Asunto(s)
Angiotensina II/metabolismo , Hipotálamo/fisiopatología , Ácido Láctico , Inhibición Neural/efectos de los fármacos , Neurotransmisores/metabolismo , Trastorno de Pánico/fisiopatología , Ácido gamma-Aminobutírico/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Masculino , Trastorno de Pánico/inducido químicamente , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Distribución Tisular
11.
Biochem Pharmacol ; 74(11): 1652-64, 2007 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-17897631

RESUMEN

Centrally administered neuropeptide Y (NPY) produces anxiolytic and orexigenic effects by interacting with Y1 and Y5 receptors that are colocalized in many brain regions. Therefore, we tested the hypothesis that co-expression of Y1 and Y5 receptors results in heterodimerization, altered pharmacological properties and altered desensitization. To accomplish this, the carboxyl-termini of Y1 and Y5 receptors were fused with Renilla luciferase and green fluorescent protein and the proximity of the tagged receptors assessed using bioluminescent resonance energy transfer. Under basal conditions, cotransfection of tagged Y1 receptor and Y5 produced a substantial dimerization signal that was unaffected by the endogenous, nonselective agonists, NPY and peptide YY (PYY). Selective Y5 agonists produced an increase in the dimerization signal while Y5 antagonists also produced a slight but significant increase. In the absence of agonists, selective antagonists decreased dimerization. In functional studies, Y5 agonists produced a greater inhibition of adenylyl cyclase activity in Y1/Y5 cells than cells expressing Y5 alone while NPY and PYY exhibited no difference. With PYY stimulation, the Y1 antagonist became inactive and the Y5 antagonist exhibited uncompetitive kinetics in the Y1/Y5 cell line. In confocal microscopy studies, Y1/Y5 co-expression resulted in increased Y5 signaling following PYY stimulation. Addition of both Y1 and Y5 receptor antagonists was required to significantly decrease PYY-induced internalization. Therefore, Y1/Y5 co-expression results in heterodimerization, altered agonist and antagonist responses and reduced internalization rate. These results may account for the complex pharmacology observed when assessing the responses to NPY and analogs in vivo.


Asunto(s)
Receptores de Neuropéptido Y/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Línea Celular , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colforsina/farmacología , Cricetinae , AMP Cíclico/metabolismo , Dimerización , Relación Dosis-Respuesta a Droga , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Luciferasas de Renilla/genética , Luciferasas de Renilla/metabolismo , Macaca mulatta , Mesocricetus , Microscopía Confocal , Neuropéptido Y/química , Neuropéptido Y/farmacología , Fragmentos de Péptidos/farmacología , Ensayo de Unión Radioligante , Receptores de Neuropéptido Y/química , Receptores de Neuropéptido Y/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Transfección
12.
Peptides ; 28(2): 241-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17208335

RESUMEN

The brain neuropeptide Neuropeptide Y (NPY) is an important modulator of a number of centrally mediated processes including feeding, anxiety-like behaviors, blood pressure and others. NPY produces its effects through at least four functional G-protein coupled receptors termed Y1, Y2, Y4 and Y5. In the brain, the Y1 and Y2 receptor subtypes are the predominant receptor population. To better understand the roles of NPY, genetically modified mice lacking NPY were produced but lacked the expected phenotypes. These mice have previously been reported to have a marked increase in Y2 receptor binding. In the present study, we found an upregulation of both Y1 and Y2 receptor binding and extended these findings to the female. These increases were as large as 10-fold or greater in many brain regions. To assess functional coupling of the receptors, we performed agonist-induced [(35)S]GTPgammaS autoradiography. In the mouse brain, the Y1/Y4/Y5 agonist Leu(31),Pro(34)-NPY increased [(35)S]GTPgammaS binding with a regional distribution consistent with that produced when labeling adjacent sections with [(125)I]-Leu(31),Pro(34)-PYY. In a few brain regions, minor increases were noted in the agonist-induced binding when comparing knock out mice to wild type. The Y2 agonist C2-NPY stimulated [(35)S]GTPgammaS binding in numerous brain areas with a regional distribution similar to the binding observed with [(125)I]-PYY3-36. Again, no major increases were noted in the functional activation of Y2 receptors between knock out and wild type mice. Therefore, the increased Y1 and Y2 binding observed in the NPY knock out mice does not represent an increase in NPY receptor mediated signaling and is likely due to an increase in spare (uncoupled) receptors.


Asunto(s)
Encéfalo/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Autorradiografía , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Receptores de Neuropéptido Y/genética
13.
J Neurosci ; 25(4): 914-22, 2005 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-15673672

RESUMEN

Melanin-concentrating hormone (MCH) neurons and MCH-1 receptors (MCH1r) densely populate mesolimbic dopaminergic brain regions such as the nucleus accumbens (NAc). The regulation of dopamine by MCH1r was suggested to be an important mechanism underlying the hyperactive phenotype of MCH1r knock-out (ko) mice. However, MCH1r modulation of monoamine neurotransmission has yet to be examined. We tested whether dopamine, norepinephrine, and serotonin function is dysregulated in MCH1r ko and wild-type (wt) mice. MCH1r ko mice exhibited robust hyperactivity in a novel or familiar environment and were super-sensitive to the locomotor activating effects of d-amphetamine and the D1 agonist 2,3,4,5-tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benazepine HCl. The D2 agonist, quinpirole, decreased locomotion similarly in both ko and wt mice. Tissue contents of dopamine within the NAc and caudate-putamen were not significantly different in ko compared with wt mice. Basal and amphetamine-evoked NAc dopamine, norepinephrine, and serotonin efflux, as measured using in vivo microdialysis, were not significantly different between genotypes. In contrast, D1-like and D2-like receptor binding were significantly higher within the olfactory tubercle, ventral tegmental area, and NAc core and shell of ko mice. Norepinephrine transporter (NET) binding was significantly elevated within the NAc shell and globus pallidus of ko mice, whereas serotonin transporter binding was decreased in the NAc shell. Thus, deletion of MCH1r results in an upregulation of mesolimbic dopamine receptors and NET, indicating that MCH1r may negatively modulate mesolimbic monoamine function. MCH1r may be an important therapeutic target for neuropsychiatric disorders involving dysregulation of limbic monoamine systems.


Asunto(s)
Dopamina/fisiología , Sistema Límbico/fisiología , Mesencéfalo/fisiología , Receptores de Somatostatina/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Animales , Dextroanfetamina/farmacología , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Masculino , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Vías Nerviosas/fisiología , Norepinefrina/metabolismo , Núcleo Accumbens/metabolismo , Quinpirol/farmacología , Receptores Dopaminérgicos/metabolismo , Receptores de Somatostatina/genética , Serotonina/metabolismo
14.
Neuropsychopharmacology ; 31(6): 1135-45, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16205780

RESUMEN

Repeated exposure to stressful conditions is linked to the etiology of affective disorders. The melanin-concentrating hormone-1 receptor (MCHR1) may be a novel mechanism that is involved in the modulation of stress responses and affective states. The role of MCHR1 in neuroendocrine, behavioral, and neurochemical stress, and anxiety-related responses was examined by monitoring the effects of melanin-concentrating hormone (MCH) and the selective MCHR1 antagonist, GW3430, in inbred C57Bl/6NTac and MCHR1-knockout (KO) and wild-type (WT) mice. Intracerebroventricular injection of MCH increased plasma corticosterone, and produced anxiety-related responses in the elevated plus maze. The selective MCHR1 antagonist, GW3430, blocked the neuroendocrine and behavioral effects of MCH and produced anxiolytic-like effects by itself in animal models of anxiety. Moreover, KO mice had an anxiolytic-like phenotype in behavioral models of anxiety, and GW3430 had anxiolytic-like effects in WT, but not KO mice. Lastly, stressor-evoked acetylcholine release within the prefrontal cortex of inbred and WT mice, but not KO mice, was blocked by GW3430. We show that MCH elicits anxiety-like responses and that the effects of a selective MCHR1 antagonist and the phenotype of KO mice are consistent with anxiolytic-like action. Distinct behavioral, physiological, and neurochemical stress, and anxiety-related responses were selectively modulated by the MCHR1, and these actions may involve corticolimbic regulation of stress responsivity and anxiety.


Asunto(s)
Conducta Animal/fisiología , Química Encefálica/fisiología , Sistemas Neurosecretores/metabolismo , Receptores de Somatostatina/fisiología , Estrés Fisiológico , Acetilcolina/metabolismo , Hormona Adrenocorticotrópica/sangre , Alprazolam/farmacología , Animales , Ansiolíticos/farmacología , Conducta Animal/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Corticosterona/sangre , Relación Dosis-Respuesta a Droga , Hormonas Hipotalámicas/administración & dosificación , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Melaninas/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sistemas Neurosecretores/efectos de los fármacos , Hormonas Hipofisarias/administración & dosificación , Distribución Aleatoria , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/deficiencia , Estrés Fisiológico/metabolismo , Estrés Fisiológico/fisiopatología , Estrés Fisiológico/psicología
15.
J Mol Neurosci ; 29(2): 109-14, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16954600

RESUMEN

The pharmacology and brain mRNA distribution of the neuropeptide Y (NPY) rat Y5 (rY5) receptor has led to the hypothesis that this receptor might mediate the hypothalamic feeding response to NPY in addition to many other physiologic functions. However, through the use of autoradiographic techniques, only very low levels of Y5-like immunoreactive (Y5-ir) binding are detected in the rat brain. To localize the Y5 protein in the rat brain, polyclonal antibodies were raised to the carboxyl terminus of the rY5 receptor. The resulting antisera were affinity purified and characterized by specific binding to HEK293 cells that had been stably transfected with the rY5 receptor. Utilizing immunohistochemical techniques, we found a discrete pattern of Y5-ir in the rat brain. In initial studies, very low levels of Y5-ir were detected, and TSA amplification was required to visualize the staining. Areas with the highest levels of expression include the piriform cortex, supraoptic nucleus, and hippocampus. Areas with moderate levels of expression include the lateral septum, amygdala, arcuate nucleus, paraventricular hypothalamic nucleus, locus coeruleus, and cerebellum. With several exceptions, this pattern of distribution is consistent with earlier reports of rY5 mRNA and receptor protein expression.


Asunto(s)
Encéfalo/metabolismo , Neuropéptido Y/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Encéfalo/anatomía & histología , Células CHO , Línea Celular , Cricetinae , Cricetulus , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
16.
Neuropeptides ; 40(2): 95-105, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16403422

RESUMEN

The peptide, nociceptin, was discovered as the endogenous ligand for the opioid-like receptor, ORL1. Since its discovery, this peptide has been shown to modulate the perception of pain, modulate feeding and produce behavioral effects in rodent models of mood disorders. Recently, the non-peptide agonist {(1S,3aS)-8-(2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4,5]decan-4-one} (Ro64-6198) of the ORL1 receptor has been reported in the literature. In the present study, we compared the distribution and potency of Ro64-6198 with nociceptin for their ability to stimulate [(35)S]-GTPgammaS binding to sections of rat brain. In initial studies, Ro64-6198 inhibited (125)I-nociceptin binding to the hORL1 receptors with a K(i) of 1.75 nM compared with 0.20 nM for nociceptin. To assess agonist potency in a whole cell assay, a cell line expressing the hORL1 receptor and G(alpha15) was created and used for calcium mobilization studies. In this assay system, Ro64-6198 increased intracellular calcium with an EC(50) of 52nM compared with 24 nM for nociceptin. Having verified the agonist properties of Ro64-6198, we then assessed the potency and distribution of ORL1 receptor activation in rat brain sections. In dose-response studies, Ro64-6198 increased [(35)S]-GTPgammaS binding to a variety of brain regions with EC(50) values ranging from 84.9 to 2,143 nM depending on the brain regions evaluated. These potencies were similar to that seen for nociceptin, but substantially lower than values established using [(125)I] nociceptin binding to the cloned human ORL1 receptor. In general, the brain distribution of agonist stimulated [(35)S]-GTPgammaS binding was similar when either Ro64-6198 or nociceptin were used. Using these techniques, we have demonstrated, for the first time that Ro64-6198 activates [(35)S]-GTPgammaS binding to rat brain sections and this compound stimulates a similar population of receptors as nociceptin.


Asunto(s)
Encéfalo/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Imidazoles/metabolismo , Péptidos Opioides/metabolismo , Compuestos de Espiro/metabolismo , Animales , Encéfalo/anatomía & histología , Línea Celular , Humanos , Imidazoles/química , Radioisótopos de Yodo/metabolismo , Masculino , Estructura Molecular , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Compuestos de Espiro/química , Radioisótopos de Azufre/metabolismo , Receptor de Nociceptina , Nociceptina
17.
Brain Res ; 1114(1): 24-30, 2006 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-16919607

RESUMEN

Psychogenic stress may be associated with the development of mood disorders and schizophrenia. The frontal cortex (FC) regulates stress responses, and its dysfunction contributes to certain neuropsychiatric disorders. We tested the effects of exposure to predator odor stress (POS), a psychogenic stressor, on the concurrent efflux of four major neurotransmitters within the FC in mice in comparison to immobilization stress (IMS), a physical/systemic stressor. POS and IMS significantly increased efflux of acetylcholine (ACh), serotonin (5-HT) and dopamine (DA), but not norephinephrine, within the FC. POS produced a somewhat longer-lasting efflux of 5-HT, as compared to IMS. The effects of POS and IMS on ACh, 5-HT and DA were blocked by chlordiazepoxide. Overall, we demonstrate a novel method to measure the effects of distinctly different stress modalities on FC neurotransmission and suggest that FC responsivity to stressors may be an important marker for evaluating anxiolytic drugs.


Asunto(s)
Acetilcolina/metabolismo , Ansiolíticos/uso terapéutico , Monoaminas Biogénicas/metabolismo , Clordiazepóxido/uso terapéutico , Lóbulo Frontal/efectos de los fármacos , Odorantes , Estrés Psicológico , Análisis de Varianza , Animales , Química Encefálica/efectos de los fármacos , Lóbulo Frontal/metabolismo , Ratones , Ratones Endogámicos C57BL , Restricción Física/métodos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/etiología , Estrés Psicológico/metabolismo
18.
Psychopharmacology (Berl) ; 233(12): 2355-63, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27044354

RESUMEN

RATIONALE: Melanin-concentrating hormone (MCH) is involved in the regulation of food intake and has recently been associated with alcohol-related behaviors. Blockade of MCH-1 receptors (MCH1-Rs) attenuates operant alcohol self-administration and decreases cue-induced reinstatement, but the mechanism through which the MCH1-R influences these behaviors remains unknown. MCH1-Rs are highly expressed in the nucleus accumbens shell (NAcSh) where they are co-expressed with dopamine (DA) receptors. MCH has been shown to potentiate responses to dopamine and to increase phosphorylation of DARPP-32, an intracellular marker of DA receptor activation, in the NAcSh. METHODS: In the present study, we investigated the role of the MCH1-R in alcohol reward using the conditioned place preference (CPP) paradigm. We then used immunohistochemistry (IHC) to assess activation of downstream signaling after administration of a rewarding dose of alcohol. RESULTS: We found that alcohol-induced CPP was markedly decreased in mice with a genetic deletion of the MCH1-R as well as after pharmacological treatment with an MCH1-R antagonist, GW803430. In contrast, an isocaloric dose of dextrose did not produce CPP. The increase in DARPP-32 phosphorylation seen in wildtype (WT) mice after acute alcohol administration in the NAcSh was markedly reduced in MCH1-R knock-out (KO) mice. CONCLUSION: Our results suggest that MCH1-Rs regulate the rewarding properties of alcohol through interactions with signaling cascades downstream of DA receptors in the NAcSh.


Asunto(s)
Condicionamiento Psicológico/efectos de los fármacos , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Etanol/administración & dosificación , Núcleo Accumbens/metabolismo , Receptores de Somatostatina/fisiología , Recompensa , Animales , Condicionamiento Psicológico/fisiología , Dopamina/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Núcleo Accumbens/efectos de los fármacos , Fosforilación/fisiología , Autoadministración
19.
J Neurosci ; 24(14): 3471-9, 2004 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-15071094

RESUMEN

The amygdala is involved in the associative processes for both appetitive and aversive emotions, and its function is modulated by stress hormones. The neuropeptide corticotrophin releasing factor (CRF) is released during stress and has been linked to many stress-related behavioral, autonomic, and endocrine responses. In the present study, nonanxiety-inducing doses of a potent CRF type 1 and 2 receptor agonist, urocortin (Ucn), was infused locally into the basolateral amygdala (BLA) of rats. After 5 daily injections of Ucn, the animals developed anxiety-like responses in behavioral tests. Intravenous administration of the anxiogenic agent sodium lactate elicited robust increases in blood pressure, respiratory rate, and heart rate. Furthermore, in the absence of any additional Ucn treatment, these behavioral and autonomic responses persisted for >30 d. Whole-cell patch-clamp recordings from BLA neurons of these hyper-reactive animals revealed a pronounced reduction in both spontaneous and stimulation-evoked IPSPs, leading to a hyperexcitability of the BLA network. This Ucn-induced plasticity appears to be dependent on NMDA receptor and subsequent calcium-calmodulin-dependent protein kinase II (CaMKII) activation, because it is blocked by pretreatment with NMDA receptor antagonists and by coadministration of CaMKII inhibitors. Our results show for the first time a stress peptide-induced behavioral syndrome that can be correlated with cellular mechanisms of neural plasticity, a novel mechanism that may explain the etiological role of stress in several chronic psychiatric and medical disorders.


Asunto(s)
Síntomas Afectivos/fisiopatología , Amígdala del Cerebelo/efectos de los fármacos , Hormona Liberadora de Corticotropina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Estrés Fisiológico/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Ansiedad/inducido químicamente , Ansiedad/fisiopatología , Presión Sanguínea/efectos de los fármacos , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Vías de Administración de Medicamentos , Esquema de Medicación , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/agonistas , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Lactato de Sodio/farmacología , Transmisión Sináptica/fisiología , Urocortinas
20.
Eur J Pharmacol ; 509(2-3): 145-53, 2005 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-15733549

RESUMEN

Corticotropin releasing factor (CRF) and Urocortin are important neurotransmitters in the regulation of physiological and behavioral responses to stress. Centrally administered CRF or Urocortin produces anxiety-like responses in numerous animal models of anxiety disorders. Previous studies in our lab have shown that Urocortin infused into the basolateral nucleus of the amygdala produces anxiety-like responses in the social interaction test. Subsequently, in the current study we prepared a specific CRF1 receptor antagonist (N-Cyclopropylmethyl-2,5-dimethyl-N-propyl-N'-(2,4,6-trichloro-phenyl)-pyrimidine-4,6-diamine, NBI3b1996) to examine in this paradigm. This CRF1 receptor antagonist inhibited the ex vivo binding of 125I-sauvagine to rat cerebellum with an ED50 of 6 mg/kg, i.p. NBI3b1996 produced a dose-dependent antagonism of Urocortin-induced anxiety-like behavior in Social Interaction test with an ED50 of 6 mg/kg, i.p. The compound had no effect on baseline social interaction. In addition, the CRF1 receptor antagonist prevented the stress-induced decrease in social interaction. These results provide further support for the CRF1 receptor in anxiety-like behavior and suggest this pathway is quiescent in unstressed animals.


Asunto(s)
Trastornos de Ansiedad/fisiopatología , Hormona Liberadora de Corticotropina/farmacología , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Conducta Social , Estrés Psicológico/fisiopatología , Proteínas Anfibias , Animales , Trastornos de Ansiedad/etiología , Autorradiografía , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Unión Competitiva/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/administración & dosificación , Relación Dosis-Respuesta a Droga , Radioisótopos de Yodo , Masculino , Hormonas Peptídicas , Péptidos/metabolismo , Pirimidinas/química , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Restricción Física , Urocortinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA