Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Addict Biol ; 28(12): e13338, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38017638

RESUMEN

Cues associated with smoking can induce relapse, which is likely driven by cue-induced neurobiological and physiological mechanisms. For instance, greater relapse vulnerability is associated with increases in cue-induced insula activation and heightened cortisol concentrations. Determining if there is a link between such cue-induced responses is critical given the need for biomarkers that can be easily measured in clinical settings and used to drive targeted treatment. Further, comprehensively characterising biological reactions to cues promises to aid in the development of therapies that address this specific relapse risk factor. To determine whether brain and cortisol responses to smoking cues are linked, this study recruited 27 nicotine-dependent tobacco-smoking individuals and acquired whole-brain functional activation during a cue reactivity task; salivary cortisol was measured before and after scanning. The results showed that increases in blood-oxygen-level-dependent activation in the right anterior insula and right dorsolateral prefrontal cortex (DLPFC) when viewing smoking versus neutral cues were positively correlated with a post-scan rise in salivary cortisol concentrations. These brain regions have been previously implicated in substance use disorders for their role in salience, interoception and executive processes. These findings show that those who have a rise in cortisol following smoking cue exposure also have a related rise in cue-induced brain reactivity, in brain regions previously linked with heightened relapse vulnerability. This is clinically relevant as measuring cue-induced cortisol responses is a more accessible proxy for assessing the engagement of cue-induced neurobiological processes associated with the maintenance of nicotine dependence.


Asunto(s)
Señales (Psicología) , Hidrocortisona , Fumar , Humanos , Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Nicotina , Recurrencia
2.
Behav Pharmacol ; 31(6): 565-573, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32209809

RESUMEN

The α4ß2* nicotinic acetylcholine receptor (nAChR) subtypes are targeted for the development of smoking cessation aids, and the use of drug discrimination in mice provides a robust screening tool for the identification of drugs acting through nAChRs. Here, we established that the α4ß2* nAChR agonist epibatidine can function as a discriminative stimulus in mice. Male C57BL/6J mice discriminated epibatidine (0.0032 mg/kg, subcutaneously) and were tested with agonists varying in selectivity and efficacy for α4ß2* nAChRs. The discriminative stimulus effects of epibatidine were characterized with the nonselective, noncompetitive nicotinic antagonist mecamylamine, with the selective ß2-substype-containing nAChR antagonist dihydro-ß-erythroidine hydrobromide (DHßE), and the α7 antagonist methyllycaconitine (MLA). Nicotine (0.32-1.0 mg/kg, subcutaneously), the partial nAChR agonist cytisine (1.0-5.6 mg/kg, subcutaneously), and the α7 nAChR agonist N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-4-chlorobenzamide (10-56 mg/kg, intraperitoneally) produced no more than 33% epibatidine-appropriate responding. The partial α4ß2* nAChR agonists varenicline and 2'-fluoro-3'-(4-nitro-phenyl)deschloroepibatidine produced 61 and 69% epibatidine-appropriate responding, respectively. DHßE and mecamylamine, but not MLA, significantly antagonized the discriminative stimulus effects of epibatidine. These results show that epibatidine may be trained as a discriminative stimulus in mice and has utility in elucidating the in-vivo pharmacology of α4ß2* nAChR ligands.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Aprendizaje Discriminativo/efectos de los fármacos , Piridinas/farmacología , Aconitina/análogos & derivados , Aconitina/farmacología , Animales , Dihidro-beta-Eritroidina/farmacología , Masculino , Mecamilamina/farmacología , Ratones , Ratones Endogámicos C57BL
3.
Brain Behav ; 10(4): e01581, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32092237

RESUMEN

OBJECTIVES: There is a long-standing interest in developing nicotinic acetylcholine receptor (nAChR) antagonists for concomitant use with nAChR agonists (e.g., nicotine replacement) as complementary smoking cessation aids. Previous studies demonstrate that daily nicotine treatment confers tolerance to some effects of nicotine, as well as cross-tolerance to other nAChR agonists. The current study assessed the extent to which antagonism of nicotine varies as a function of daily nicotine treatment. METHODS: Schedule-controlled responding and hypothermia were selected for study because they have been previously used to examine the pharmacology of nicotine, and both are sensitive to the development nicotine tolerance. The rate-decreasing and hypothermic effects of nicotine, as well as antagonism of those effects, were examined in C57BL/6J mice before, during treatment with, and after discontinuation of three daily injections of 1.78 mg/kg nicotine. The nonselective nAChR antagonist mecamylamine and the ß2 nAChR antagonist dihydro-ß-erythroidine (DHßE) were studied in combination with nicotine. RESULTS: The ED50 values of nicotine to produce rate-decreasing and hypothermic effects were, respectively, 0.44 and 0.82 mg/kg prior, 1.6 and 3.2 mg/kg during, and 0.74 and 1.1 mg/kg after discontinuation of daily nicotine treatment. Prior to daily nicotine treatment, mecamylamine decreased response rate and rectal temperature. However, during daily nicotine, mecamylamine (up to 5.6 mg/kg) only decreased rectal temperature. DHßE (up to 5.6 mg/kg) when studied prior to daily nicotine decreased rectal temperature, but that decrease was abolished during chronic nicotine treatment. Mecamylamine and DHßE antagonized the rate-decreasing and hypothermic effects of nicotine before and after daily nicotine; however, during daily nicotine, mecamylamine and DHßE antagonized only the hypothermic effects of nicotine. CONCLUSIONS: The differential antagonism of rate-decreasing and hypothermic effects implicates differential involvement of nAChR subtypes. The decreased capacity of mecamylamine and DHßE to antagonize nicotine during chronic nicotine treatment may indicate that their effectiveness as smoking cessations might vary as a function of nicotine tolerance and dependence.


Asunto(s)
Dihidro-beta-Eritroidina/farmacología , Tolerancia a Medicamentos , Mecamilamina/farmacología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Cese del Hábito de Fumar
4.
Exp Clin Psychopharmacol ; 28(5): 517-526, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31789555

RESUMEN

Chronic health problems associated with long-term nicotine use are the leading cause of preventable death in the United States. The use of tobacco products is 3-4 times greater among individuals with cocaine use disorder than that observed in the general population. This may reflect the propensity of nicotine to augment the reinforcing effects of cocaine. However, the mechanism of action of nicotine differs from that of cocaine, which presents a significant challenge for the development of pharmacotherapeutic interventions for the management of nicotine + cocaine polydrug abuse. Bupropion, an FDA-approved smoking cessation aid, has pharmacological actions at both monoamine transporters and nicotinic receptors, suggesting that it may be effective at decreasing nicotine + cocaine coabuse. Here, rhesus monkeys (n = 4) responded for food pellets and, separately, intravenous injections of nicotine, cocaine, or nicotine + cocaine mixtures under a second-order FR2(VR16:S) schedule of reinforcement during 7- to 10-day continuous treatment with saline or bupropion (1.0 and 1.8 mg/kg/hr). Results show that bupropion treatment dose-dependently decreased self-administration of nicotine combined with a low dose of cocaine (0.0032 mg/kg/inj); however, when the dose of cocaine in the mixture was higher (i.e., 0.01 mg/kg/inj), bupropion attenuated self-administration in only a subset of subjects. The effective dosage of bupropion increased responding for cocaine alone, nicotine alone, and for saline injections and significantly increased measures of daily activity. The apparent stimulant-like effects of bupropion at the dosage required to decrease cocaine + nicotine self-administration does not support its clinical use for the management of nicotine + cocaine polydrug abuse. (PsycInfo Database Record (c) 2020 APA, all rights reserved).


Asunto(s)
Bupropión/administración & dosificación , Cocaína/administración & dosificación , Macaca mulatta/fisiología , Nicotina/administración & dosificación , Agentes para el Cese del Hábito de Fumar/administración & dosificación , Animales , Estimulantes del Sistema Nervioso Central/uso terapéutico , Relación Dosis-Respuesta a Droga , Masculino , Refuerzo en Psicología , Autoadministración
5.
Behav Pharmacol ; 30(5): 412-421, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30398980

RESUMEN

Varenicline is a smoking cessation pharmacotherapy with a presumed mechanism of action of partial efficacy at the α4ß2 nicotinic acetylcholine receptor (nAChR); however, the extent to which daily varenicline use leads to changes in nAChR sensitivity is unclear. This study examined the consequences of daily varenicline treatment on disruptions in operant responding (i.e. rate-decreasing effects) and hypothermia induced by administration of nicotine, epibatidine, cytisine, and cocaine in C57BL/6J mice. Furthermore, mecamylamine was used to assess the involvement of nAChRs in the effects of varenicline. Mice were trained under a fixed ratio 20 of milk reinforcement, and rectal temperatures were measured after 30 min following drug-administration. Varenicline, nicotine, epibatidine, and cytisine produced dose-dependent decreases in response rate and rectal temperature. Chronic varenicline (30 mg/kg) engendered tolerance to varenicline, but more cross-tolerance to nicotine, for both disruptions in operant responding and hypothermia. Cross-tolerance only developed to the hypothermic effects of epibatidine, and no cross-tolerance developed to any effects of cytisine and cocaine. In varenicline-tolerant mice, mecamylamine did not antagonize the effects of varenicline. The varying magnitudes of tolerance and cross-tolerance among effects and drugs are indicative of a nonuniform nAChR pharmacology in vivo.


Asunto(s)
Tolerancia a Medicamentos/fisiología , Receptores Nicotínicos/efectos de los fármacos , Vareniclina/farmacología , Alcaloides/farmacología , Animales , Azocinas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Cocaína/farmacología , Condicionamiento Operante/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Piridinas/farmacología , Quinolizinas/farmacología , Receptores Nicotínicos/fisiología , Refuerzo en Psicología , Cese del Hábito de Fumar/métodos , Vareniclina/metabolismo
6.
Psychopharmacology (Berl) ; 234(5): 781-792, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28028600

RESUMEN

RATIONALE: The extent to which non-α4ß2 versus α4ß2* nAChRs contribute to the behavioral effects of varenicline and other nAChR agonists is unclear. OBJECTIVES: The purpose of this study was to characterize the discriminative stimulus effects of varenicline and nicotine using various nAChR agonists and antagonists to elucidate possible non-α4ß2 nAChR mechanisms. METHODS: Separate groups of male C57BL/6J mice were trained to discriminate varenicline (3.2 mg/kg) or nicotine (1 mg/kg). Test drugs included mecamylamine; the nAChR agonists epibatidine, nicotine, cytisine, varenicline, and RTI-102; the ß2-containing nAChR antagonist dihydro-ß-erythroidine (DHßE); the α7 nAChR agonist PNU-282987; the α7 antagonist methyllycaconitine (MLA); the α3ß4 antagonist 18-methoxycoronaridine (18-MC); and the non-nAChR drugs midazolam and cocaine. RESULTS: In nicotine-trained mice, maximum nicotine-appropriate responding was 95% nicotine, 94% epibatidine, 63% varenicline, 58% cytisine, and less than 50% for RTI-102, PNU-282987, midazolam, and cocaine. In varenicline-trained mice, maximum varenicline-appropriate responding was 90% varenicline, 86% epibatidine, 74% cytisine, 80% RTI-102, 50% cocaine, and 50% or less for nicotine, PNU-282987, and midazolam. Drugs were studied to doses that abolished operant responding. Mecamylamine antagonized the discriminative stimulus effects, but not the rate-decreasing effects, of nicotine and varenicline. DHßE antagonized the discriminative stimulus and rate-decreasing effects of nicotine but not varenicline in either the nicotine or varenicline discrimination assays. The discriminative stimulus, but not the rate-decreasing, effects of epibatidine were antagonized by DHßE regardless of the training drug. CONCLUSIONS: These results suggest that α4ß2* nAChRs differentially mediate the discriminative stimulus effects of nicotine and varenicline, and suggest that varenicline has substantial non-α4ß2 nAChR activity.


Asunto(s)
Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/efectos de los fármacos , Vareniclina/farmacología , Aconitina/análogos & derivados , Aconitina/farmacología , Alcaloides/farmacología , Animales , Azocinas/farmacología , Benzamidas/farmacología , Compuestos Bicíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Cocaína/farmacología , Dihidro-beta-Eritroidina/farmacología , Discriminación en Psicología , Inhibidores de Captación de Dopamina/farmacología , Hipnóticos y Sedantes/farmacología , Ibogaína/análogos & derivados , Ibogaína/farmacología , Masculino , Mecamilamina/farmacología , Ratones , Ratones Endogámicos C57BL , Midazolam/farmacología , Proteínas del Tejido Nervioso/efectos de los fármacos , Piridinas/farmacología , Quinolizinas/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/efectos de los fármacos
7.
Eur J Pharmacol ; 786: 169-178, 2016 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-27238974

RESUMEN

Some drugs that are positive allosteric nAChR modulators in vitro, desformylflustrabromine (dFBr), PNU-120596 and LY 2087101, have not been fully characterized in vivo. These drugs were examined for their capacity to share or modify the hypothermic and discriminative stimulus effects of nicotine (1mg/kg s.c.) in male C57Bl/6J mice. Nicotine, dFBr, and PNU-120596 produced significant hypothermia, whereas LY 2087101 (up to 100mg/kg) did not. Nicotine dose-dependently increased nicotine-appropriate responding and decreased response rate; the respective ED50 values were 0.56mg/kg and 0.91mg/kg. The modulators produced no more than 38% nicotine-appropriate responding up to doses that disrupted operant responding. Rank order potency was the same for hypothermia and rate-decreasing effects: nicotine>dFBr>PNU-120596=LY 2087101. Mecamylamine and the α4ß2 nAChR antagonist dihydro-ß-erythroidine, but not the α7 antagonist methyllycaconitine, antagonized the hypothermic effects of nicotine. In contrast, mecamylamine did not antagonize the hypothermic effects of the modulators. The combined discriminative stimulus effects of DFBr and nicotine were synergistic, whereas the combined hypothermic effects of nicotine with either dFBr or PNU-120596 were infra-additive. PNU-120596 did not modify the nicotine discriminative stimulus, and LY 2087101 did not significantly modify either effect of nicotine. Positive modulation of nicotine at nAChRs by PNU-120596 and LY 2087101 in vitro does not appear to confer enhancement of the nAChR-mediated hypothermic or discriminative stimulus effects of nicotine. However, dFBr appears to be a positive allosteric modulator of some behavioral effects of nicotine at doses of dFBr smaller than the doses producing unwanted effects (e.g. hypothermia) through non-nAChR mechanisms.


Asunto(s)
Discriminación en Psicología/efectos de los fármacos , Hipotermia/inducido químicamente , Nicotina/farmacología , Receptores Nicotínicos/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Behav Pharmacol ; 27(2-3 Spec Issue): 240-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26910582

RESUMEN

The tobacco-dependence pharmacotherapies varenicline and cytisine act as partial α4ß2 nAChR agonists. However, the extent to which α4ß2 nicotinic acetylcholine receptors (nAChRs) mediate their in-vivo effects remains unclear. Nicotine, varenicline, cytisine, and epibatidine were studied in male C57BL/6J mice for their effects on rates of fixed ratio responding and rectal temperature alone and in combination with the nonselective nAChR antagonist mecamylamine and the α4ß2 nAChR antagonist dihydro-ß-erythroidine. The effects of nicotine, varenicline, cytisine, epibatidine, and cocaine were assessed before and during chronic nicotine treatment. The rate-decreasing and hypothermic effects of nicotine, varenicline, cytisine, and epibatidine were antagonized by mecamylamine (1 mg/kg), but only the effects of nicotine and epibatidine were antagonized by dihydro-ß-erythroidine (3.2 mg/kg). Chronic nicotine produced 4.7 and 5.1-fold rightward shifts in the nicotine dose-effect functions to decrease response rate and rectal temperature, respectively. Nicotine treatment decreased the potency of epibatidine to decrease response rate and rectal temperature 2.2 and 2.9-fold, respectively, and shifted the varenicline dose-effect functions 2.0 and 1.7-fold rightward, respectively. Cross-tolerance did not develop from nicotine to cytisine. These results suggest that the in-vivo pharmacology of tobacco cessation aids cannot be attributed to a single nAChR subtype; instead, multiple receptor subtypes differentially mediate their effects.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Hipertermia Inducida/métodos , Mecamilamina/uso terapéutico , Agonistas Nicotínicos/uso terapéutico , Antagonistas Nicotínicos/uso terapéutico , Tabaquismo/terapia , Animales , Cocaína , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Masculino , Mecamilamina/farmacología , Ratones , Ratones Endogámicos C57BL , Esquema de Refuerzo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA