Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuropsychopharmacology ; 48(13): 1878-1888, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37355732

RESUMEN

The high rates of relapse associated with current medications used to treat opioid use disorder (OUD) necessitate research that expands our understanding of the neural mechanisms regulating opioid taking to identify molecular substrates that could be targeted by novel pharmacotherapies to treat OUD. Recent studies show that activation of calcitonin receptors (CTRs) is sufficient to reduce the rewarding effects of addictive drugs in rodents. However, the role of central CTR signaling in opioid-mediated behaviors has not been studied. Here, we used single nuclei RNA sequencing (snRNA-seq), fluorescent in situ hybridization (FISH), and immunohistochemistry (IHC) to characterize cell type-specific patterns of CTR expression in the nucleus accumbens (NAc), a brain region that plays a critical role in voluntary drug taking. Using these approaches, we identified CTRs expressed on D1R- and D2R-expressing medium spiny neurons (MSNs) in the medial shell subregion of the NAc. Interestingly, Calcr transcripts were expressed at higher levels in D2R- versus D1R-expressing MSNs. Cre-dependent viral-mediated miRNA knockdown of CTRs in transgenic male rats was then used to determine the functional significance of endogenous CTR signaling in opioid taking. We discovered that reduced CTR expression specifically in D1R-expressing MSNs potentiated/augmented opioid self-administration. In contrast, reduced CTR expression specifically in D2R-expressing MSNs attenuated opioid self-administration. These findings highlight a novel cell type-specific mechanism by which CTR signaling in the ventral striatum bidirectionally modulates voluntary opioid taking and support future studies aimed at targeting central CTR-expressing circuits to treat OUD.


Asunto(s)
Analgésicos Opioides , Núcleo Accumbens , Ratas , Animales , Masculino , Analgésicos Opioides/farmacología , Analgésicos Opioides/metabolismo , Receptores de Calcitonina/genética , Receptores de Calcitonina/metabolismo , Neuronas Espinosas Medianas , Hibridación Fluorescente in Situ , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D1/metabolismo
2.
Neuropharmacology ; 192: 108599, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33965397

RESUMEN

There has been a dramatic increase in illicit fentanyl use in the United States over the last decade. In 2018, more than 31,000 overdose deaths involved fentanyl or fentanyl analogs, highlighting an urgent need to identify effective treatments for fentanyl use disorder. An emerging literature shows that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate the reinforcing efficacy of drugs of abuse. However, the effects of GLP-1R agonists on fentanyl-mediated behaviors are unknown. The first goal of this study was to determine if the GLP-1R agonist exendin-4 reduced fentanyl self-administration and the reinstatement of fentanyl-seeking behavior, an animal model of relapse, in rats. We found that systemic exendin-4 attenuated fentanyl taking and seeking at doses that also produced malaise-like effects in rats. To overcome these adverse effects and enhance the clinical potential of GLP-1R agonists, we recently developed a novel dual agonist of GLP-1Rs and neuropeptide Y2 receptors (Y2Rs), GEP44, that does not produce nausea-like behavior in drug-naïve rats or emesis in drug-naïve shrews. The second goal of this study was to determine if GEP44 reduced fentanyl self-administration and reinstatement with fewer adverse effects compared to exendin-4 alone. In contrast to exendin-4, GEP44 attenuated opioid taking and seeking at a dose that did not suppress food intake or produce adverse malaise-like effects in fentanyl-experienced rats. Taken together, these findings indicate a novel role for GLP-1Rs and Y2Rs in fentanyl reinforcement and highlight a potential new therapeutic approach to treating opioid use disorders.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Conducta Adictiva/tratamiento farmacológico , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Fentanilo/administración & dosificación , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptores de Neuropéptido Y/agonistas , Animales , Conducta Adictiva/metabolismo , Conducta Adictiva/psicología , Relación Dosis-Respuesta a Droga , Comportamiento de Búsqueda de Drogas/fisiología , Exenatida/farmacología , Exenatida/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/metabolismo , Autoadministración
3.
Neuropsychopharmacology ; 45(3): 451-461, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31581176

RESUMEN

Despite the effectiveness of current medications to treat opioid use disorder, there is still a high rate of relapse following detoxification. Thus, there is critical need for innovative studies aimed at identifying novel neurobiological mechanisms that could be targeted to treat opioid use disorder. A growing body of preclinical evidence indicates that glucagon-like peptide-1 (GLP-1) receptor agonists reduce drug reinforcement. However, the efficacy of GLP-1 receptor agonists in attenuating opioid-mediated behaviors has not been thoroughly investigated. Using recently established models of opioid-taking and -seeking behaviors, we showed that systemic administration of the GLP-1 receptor agonist exendin-4 reduced oxycodone self-administration and the reinstatement of oxycodone-seeking behavior in rats. We also identified behaviorally selective doses of exendin-4 that reduced opioid-taking and -seeking behaviors and did not produce adverse feeding effects in oxycodone-experienced rats. To identify a central site of action, we showed that systemic exendin-4 penetrated the brain and bound putative GLP-1 receptors on dopamine D1 receptor- and dopamine D2 receptor-expressing medium spiny neurons in the nucleus accumbens shell. Consistent with our systemic studies, infusions of exendin-4 directly into the accumbens shell attenuated oxycodone self-administration and the reinstatement of oxycodone-seeking behavior without affecting ad libitum food intake. Finally, exendin-4 did not alter the analgesic effects of oxycodone, suggesting that activation of GLP-1 receptors attenuated opioid reinforcement without reducing the thermal antinociceptive effects of oxycodone. Taken together, these findings suggest that GLP-1 receptors could serve as potential molecular targets for pharmacotherapies aimed at reducing opioid use disorder.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos/administración & dosificación , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Oxicodona/administración & dosificación , Dimensión del Dolor/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Comportamiento de Búsqueda de Drogas/fisiología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Masculino , Dimensión del Dolor/métodos , Dimensión del Dolor/psicología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...