Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884757

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) is a 17-residue neuropeptide that binds the nociceptin opioid-like receptor (NOP). N/OFQ exhibits nucleotidic and aminoacidics sequence homology with the precursors of other opioid neuropeptides but it does not activate either MOP, KOP or DOP receptors. Furthermore, opioid neuropeptides do not activate the NOP receptor. Generally, activation of N/OFQ system exerts anti-opioids effects, for instance toward opioid-induced reward and analgesia. The NOP receptor is widely expressed throughout the brain, whereas N/OFQ localization is confined to brain nuclei that are involved in stress response such as amygdala, BNST and hypothalamus. Decades of studies have delineated the biological role of this system demonstrating its involvement in significant physiological processes such as pain, learning and memory, anxiety, depression, feeding, drug and alcohol dependence. This review discusses the role of this peptidergic system in the modulation of stress and stress-associated psychiatric disorders in particular drug addiction, mood, anxiety and food-related associated-disorders. Emerging preclinical evidence suggests that both NOP agonists and antagonists may represent a effective therapeutic approaches for substances use disorder. Moreover, the current literature suggests that NOP antagonists can be useful to treat depression and feeding-related diseases, such as obesity and binge eating behavior, whereas the activation of NOP receptor by agonists could be a promising tool for anxiety.


Asunto(s)
Péptidos Opioides/fisiología , Receptores Opioides/fisiología , Estrés Fisiológico/fisiología , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Trastornos de Alimentación y de la Ingestión de Alimentos/tratamiento farmacológico , Trastornos de Alimentación y de la Ingestión de Alimentos/fisiopatología , Humanos , Modelos Neurológicos , Trastornos del Humor/tratamiento farmacológico , Trastornos del Humor/fisiopatología , Péptidos Opioides/agonistas , Péptidos Opioides/antagonistas & inhibidores , Recompensa , Estrés Fisiológico/efectos de los fármacos , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/fisiopatología , Receptor de Nociceptina , Nociceptina
2.
Handb Exp Pharmacol ; 254: 187-212, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30968214

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) is a 17 amino acid peptide that was deorphanized in 1995 and has been widely studied since. The role of the N/OFQ system in drug abuse has attracted researchers' attention since its initial discovery. The first two scientific papers describing the effect of intracranial injection of N/OFQ appeared 20 years ago and reported efficacy of the peptide in attenuating alcohol intake, whereas heroin self-administration was insensitive. Since then more than 100 scientific articles investigating the role of the N/OFQ and N/OFQ receptor (NOP) system in drug abuse have been published. The present article provides an historical overview of the advances in the field with focus on three major elements. First, the most robust data supportive of the efficacy of NOP agonists in treating drug abuse come from studies in the field of alcohol research, followed by psychostimulant and opioid research. In contrast, activation of NOP appears to facilitate nicotine consumption. Second, emerging data challenge the assumption that activation of NOP is the most appropriate strategy to attenuate consumption of substances of abuse. This assumption is based first on the observation that animals carrying an overexpression of NOP system components are more prone to consume substances of abuse, whereas NOP knockout rats are less motivated to self-administer heroin, alcohol, and cocaine. Third, administration of NOP antagonists also reduces alcohol consumption. In addition, NOP blockade reduces nicotine self-administration. Hypothetical mechanisms explaining this apparent paradox are discussed. Finally, we focus on the possibility that co-activation of NOP and mu opioid (MOP) receptors is an alternative strategy, readily testable in the clinic, to reduce the consumption of psychostimulants, opiates, and, possibly, alcohol.


Asunto(s)
Analgésicos Opioides/farmacología , Péptidos Opioides/metabolismo , Receptores Opioides , Trastornos Relacionados con Sustancias , Analgésicos Opioides/química , Animales , Etanol/química , Péptidos Opioides/química , Ratas , Receptores Opioides/química , Autoadministración , Nociceptina
3.
Addict Biol ; 23(1): 6-15, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558790

RESUMEN

Hypothalamic orexin/hypocretin (Orx/Hcrt) neurons are thought to mediate both food-reinforced behaviors and behavior motivated by drugs of abuse. However, the relative role of the Orx/Hcrt system in behavior motivated by food versus drugs of abuse remains unclear. This investigation addressed this question by contrasting hypothalamic Orx/Hcrt neuronal activation associated with reinstatement of reward seeking induced by stimuli conditioned to cocaine (COC) versus highly palatable food reward, sweetened condensed milk (SCM). Orx/Hcrt neuronal activation in the lateral hypothalamus, dorsomedial hypothalamus and perifornical area, determined by dual c-fos/orx immunocytochemistry, was quantified in rat brains, following reinstatement of reward seeking induced by a discriminative stimulus (S+ ) conditioned to COC or SCM. The COC S+ and SCM S+ initially produced the same magnitude of reward seeking. However, over four subsequent tests, behavior induced by the SCM S+ decreased to extinction levels, whereas reinstatement induced by the COC S+ perseverated at undiminished levels. Following both the first and fourth tests, the percentage of Orx/Hcrt cells expressing Fos was significantly increased in all hypothalamic subregions in rats tested with the COC S+ but not rats tested with the SCM S+ . These findings point toward a role for the Orx/Hcrt system in perseverating, compulsive-like COC seeking but not behavior motivated by palatable food. Moreover, analysis of the Orx/Hcrt recruitment patterns suggests that failure of Orx/Hcrt neurons in the lateral hypothalamus to respond to inhibitory inputs from Orx/Hcrt neurons in the dorsomedial hypothalamus/perifornical area may contribute to the perseverating nature of COC seeking.


Asunto(s)
Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Alimentos , Hipotálamo/metabolismo , Neuronas/metabolismo , Orexinas/metabolismo , Animales , Conducta Animal , Condicionamiento Operante , Comportamiento de Búsqueda de Drogas , Hipotálamo/citología , Inmunohistoquímica , Leche , Motivación , Neuronas/citología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Recompensa , Autoadministración
4.
Addict Biol ; 22(4): 923-932, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26864474

RESUMEN

Associative learning is essential for establishing appropriate responses to cause-effect relationships and effective behavioral adjustments to environmental changes. However, learned associations also promote maladaptive behavior such as uncontrollable drug seeking in addicts exposed to drug-associated stimuli. Here, we sought to identify behavioral characteristics that distinguish reward seeking produced by environmental stimuli conditioned to highly potent but non-addictive conventional reinforcers from reward seeking induced by stimuli conditioned to addictive drugs. Rats were trained to associate discriminative (i.e. contextual) stimuli (S+ ) with availability of cocaine, ethanol, palatable sweet solutions or water during dehydration. Following extinction, response-reinstating effects of re-exposure to these stimuli were established in terms of magnitude and perseveration. Initially, the S+ produced strong reinstatement irrespective of association with conventional or drug reward. However, with repeated testing, S+ -induced reward seeking decreased to extinction levels when motivated by the sweet solutions but perseverated when motivated by cocaine or ethanol. In rats placed on water restriction to induce a motivational constraint, the S+ supported perseverating reinstatement identical to that produced by an S+ conditioned to cocaine. The findings suggest that behavior guided by associations between environmental stimuli and drugs of abuse is characterized by perseverating, apparently highly extinction-resistant reward seeking, whereas behavior controlled by stimuli associated with conventional reward extinguishes rapidly in the absence of primary reinforcement. Reward seeking elicited by stimuli associated with natural reward can, however, become perseverative during physiological deprivation states. Possibly, perseverating drug seeking engages mechanisms overlapping with those that have evolved to promote alleviation of physiological deprivation to secure survival.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cocaína/administración & dosificación , Condicionamiento Clásico/efectos de los fármacos , Ansia/efectos de los fármacos , Etanol/administración & dosificación , Refuerzo en Psicología , Animales , Señales (Psicología) , Azúcares de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Masculino , Motivación/efectos de los fármacos , Ratas , Ratas Wistar , Recompensa , Autoadministración , Agua/administración & dosificación
5.
Addict Biol ; 22(1): 70-77, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26096647

RESUMEN

The paraventricular nucleus of the thalamus (PVT) is not traditionally considered part of the brain addiction neurocircuitry but has received growing attention with regard to a role in the modulation of drug-seeking behavior. This study sought to establish the pattern of neural activation induced by a response-reinstating discriminative stimulus (SD ) conditioned to either cocaine (COC) or a conventional reinforcer using a palatable food substance, sweetened condensed milk (SCM). Male Wistar rats were trained to associate one SD (S+ ; COC or SCM availability) and a distinctly different SD (S- ; non-reward; i.e. the availability of saline or the absence of SCM). Following extinction of COC- and SCM-reinforced responding, rats were presented with the respective S+ or S- alone and tested for the reinstatement of reward seeking. The COC S+ and SCM S+ elicited identical reinstatement, whereas the non-reward S- was behaviorally ineffective. PVT sections were obtained following completion of the reinstatement tests and labeled for Fos. The number of Fos+ neurons was compared among rats that were presented with the COC S+ , SCM S+ or S- . Rats that were presented with the COC S+ exhibited a significant increase in Fos expression compared with rats that were presented with the S- . Moreover, Fos expression was significantly correlated with the number of reinstatement responses that were induced by the COC S+ . In contrast, the SCM S+ and S- produced identical increases in Fos expression, without behaviorally relevant correlations. The findings implicate the PVT as an important site that is selectively recruited during COC-seeking behavior.


Asunto(s)
Alimentación Animal , Conducta Animal/efectos de los fármacos , Cocaína/farmacología , Condicionamiento Operante/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Animales , Señales (Psicología) , Inhibidores de Captación de Dopamina/farmacología , Masculino , Modelos Animales , Ratas , Ratas Wistar , Autoadministración , Tálamo/efectos de los fármacos
6.
J Pharmacol Exp Ther ; 359(2): 273-279, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27540003

RESUMEN

Orexin/hypocretin (Orx/Hcrt) projections from the lateral hypothalamus to the paraventricular nucleus of the thalamus (PVT) are implicated in drug addiction. Specifically, the posterior section of the PVT (pPVT) innervates brain structures that modulate motivated behavior. This study investigated the role of pPVT-Orx/Hcrt transmission in cocaine-seeking behavior. Because the effects of Orx/Hcrt are mediated by two Orx/Hcrt receptors (Hcrt-r1 and Hcrt-r2), we examined the extent to which Hcrt-r1 and Hcrt-r2 are involved in Orx/Hcrt-induced cocaine seeking. Male Wistar rats were made cocaine dependent by self-administering cocaine 6 hours/day (long access) for 21 days. After self-administration training, the rats underwent daily extinction training, during which cocaine was withheld. After extinction, the rats were injected into the pPVT with Orx-A/Hcrt-1 (0-2 µg) alone or, using a single dose of 0.5 µg, in combination with an Hcrt-r1 antagonist (SB334867; 0-15 µg) or an Hcrt-r2 antagonist (TCSOX229; 0-15 µg). Orx-A/Hcrt-1 alone reinstated (primed) cocaine seeking. Unexpectedly, coadministration of Orx-A/Hcrt-1 with SB334867 did not have any effects on Orx-A/Hcrt-1-induced reinstatement, whereas when coadministered with Orx-A/Hcrt-1, TCSOX229 prevented cocaine-seeking behavior. These results indicate that Hcrt-r2 in the pPVT mediates the reinstating effect of Orx-A/Hcrt-1 in animals with a history of cocaine dependence and further identify Hcrt-r2 as a possible molecular target that can guide future therapeutic approaches for the prevention of drug-seeking behavior.


Asunto(s)
Cocaína/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Receptores de Orexina/metabolismo , Orexinas/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Animales , Benzoxazoles/farmacología , Interacciones Farmacológicas , Extinción Psicológica/efectos de los fármacos , Isoquinolinas/farmacología , Masculino , Naftiridinas , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Piridinas/farmacología , Ratas , Ratas Wistar , Urea/análogos & derivados , Urea/farmacología
7.
Alcohol Clin Exp Res ; 40(5): 945-54, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27084498

RESUMEN

BACKGROUND: The nociceptin/orphanin-FQ (or opioid receptor-like [ORL1]) receptor (NOP) is localized in the mesolimbic reward pathway and has been suggested to play a role in feeding, mood, stress, and addiction. Since its deorphanization in 1995, there has been a clear dichotomy in the literature regarding whether an agonist or antagonist would provide therapeutic benefit. Specifically, the literature reports indicate that NOP receptor antagonists produce efficacy in animal models of hyperphagia and antidepressant-like activity, whereas NOP agonists produce anxiolytic-like effects and dampen reward/addiction behaviors including ethanol consumption. METHODS: We characterize here the potent, orally bioavailable NOP antagonist, LY2940094, in rodent models of ethanol consumption, including ethanol self-administration, progressive ratio operant self-administration, stress-induced reinstatement of ethanol seeking, and in vivo microdialysis in the nucleus accumbens. RESULTS: LY2940094 dose dependently reduced homecage ethanol self-administration in Indiana alcohol-preferring (P) and Marchigian Sardinian alcohol-preferring (msP) rats, without affecting food/water intake or locomotor activity. Reduced ethanol intake in P rats did not show significant tolerance over 4 days of subchronic dosing. LY2940094 attenuated progressive ratio operant responding and break points for ethanol in P rats. Moreover, stress-induced reinstatement of ethanol seeking in msP rats was completely blocked by LY2940094. Furthermore, LY2940094 blocked ethanol-stimulated dopamine release in response to ethanol challenge (1.1 g/kg, intraperitoneally). CONCLUSIONS: Our findings demonstrate for the first time that blockade of NOP receptors attenuates ethanol self-administration and ethanol-motivated behaviors, stress-induced ethanol seeking, and ethanol-induced stimulation of brain reward pathways in lines of rats that exhibit excessive ethanol consumption. Results suggest that LY2940094 may have potential therapeutic utility in treating alcohol addiction.


Asunto(s)
Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/antagonistas & inhibidores , Piranos/farmacología , Receptores Opioides/efectos de los fármacos , Compuestos de Espiro/farmacología , Administración Oral , Animales , Condicionamiento Operante/efectos de los fármacos , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Etanol/administración & dosificación , Femenino , Masculino , Microdiálisis , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Piranos/administración & dosificación , Ratas , Ratas Endogámicas , Autoadministración , Compuestos de Espiro/administración & dosificación , Receptor de Nociceptina
8.
Addict Biol ; 20(4): 643-51, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24930632

RESUMEN

Dysregulation of the nociceptin (N/OFQ) system has been implicated in alcohol abuse and alcoholism, and growing evidence suggests that targeting this system may be beneficial for treating alcoholism. To further explore the treatment target potential of the N/OFQ system, the novel non-peptide, small-molecule N/OFQ (NOP) agonist MT-7716, (R)-2-{3-[1-(Acenaphthen-1-yl)piperidin-4-yl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl}-N-methylacetamide hydrochloride hydrate, was examined for its effects on ethanol self-administration and stress-induced reinstatement of alcohol seeking in non-dependent and post-dependent rats. Male Wistar rats were trained to self-administer ethanol and then made ethanol dependent via repeated intragastric ethanol intubation. The effects of MT-7716 (0.3 and 1 mg/kg; PO) on alcohol self-administration were determined 2 weeks following dependence induction, when baseline self-administration was restored. Effects of MT-7716 on stress-induced reinstatement were tested in separate cohorts of rats, 1 and 3 weeks post-withdrawal. MT-7716 reduced alcohol self-administration and stress-induced reinstatement of alcohol seeking in post-dependent rats, but was ineffective in non-dependent animals. Moreover, the prevention of stress-induced reinstatement by MT-7716 was more pronounced at 3 weeks post-dependence. The results further confirm treatment target potential for the NOP receptor and identify non-peptide NOP agonists as promising potential treatment drugs for alcohol abuse and relapse prevention. The findings also support dysregulation of the N/OFQ system as a factor in alcohol seeking and reinforcement.


Asunto(s)
Acenaftenos/farmacología , Alcoholismo/prevención & control , Bencimidazoles/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/antagonistas & inhibidores , Receptores Opioides , Refuerzo en Psicología , Alcoholismo/sangre , Análisis de Varianza , Animales , Condicionamiento Operante , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/metabolismo , Masculino , Nocicepción/efectos de los fármacos , Ratas Wistar , Receptores Opioides/efectos de los fármacos , Autoadministración , Estrés Psicológico/psicología , Receptor de Nociceptina , Nociceptina
9.
Addict Biol ; 19(2): 233-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22830647

RESUMEN

Orexins/hypocretins (Orx/Hcrt) are hypothalamic peptides that regulate a wide range of physiological processes and have been shown to be recruited by drugs of abuse. This study was designed to test the effect of the specific Orx/Hcrt receptor-1 (Hcrt-r1) antagonist, N-(2-methyl-6-benzoxazolyl)-N'-1,5-naphthyridin-4-yl urea (SB334867), on reinstatement elicited by ethanol (EtOH)-associated stimuli versus stimuli associated with a conventional reinforcer [i.e. SuperSac, consisting of 3% glucose and 0.125% saccharin (w/v)]. SB334867 (1-10 mg/kg, i.p.) dose-dependently reduced reinstatement induced by the EtOH- but not SuperSac-associated stimuli. These findings support a differential role of Hcrt-r1 in mediating EtOH seeking versus natural reward seeking.


Asunto(s)
Benzoxazoles/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuropéptidos/fisiología , Antagonistas de los Receptores de Orexina , Urea/análogos & derivados , Análisis de Varianza , Animales , Relación Dosis-Respuesta a Droga , Naftiridinas , Orexinas , Ratas , Refuerzo en Psicología , Recompensa , Prevención Secundaria , Autoadministración , Edulcorantes/administración & dosificación , Urea/farmacología
10.
Neuropharmacology ; 257: 110048, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38901642

RESUMEN

Maintenance therapy with buprenorphine and methadone is the gold standard pharmacological treatment for opioid use disorder (OUD). Despite these compounds demonstrating substantial efficacy, a significant number of patients do not show optimal therapeutic responses. The abuse liability of these medications is also a concern. Here we used rats to explore the therapeutic potential of the new long-acting pan-opioid agonist Cebranopadol in OUD. We tested the effect of cebranopadol on heroin self-administration and yohimbine-induced reinstatement of heroin seeking. In addition, we evaluated the abuse liability potential of cebranopadol in comparison to that of heroin under fixed ratio 1 (FR1) and progressive ratio (PR) operant self-administration contingencies. Oral administration of cebranopadol (0, 25, 50 µg/kg) significantly attenuated drug self-administration independent of heroin dose (1, 7, 20, 60µg/inf). Cebranopadol also reduced the break point for heroin (20 µg/inf). Finally, pretreatment with cebranopadol significantly attenuated yohimbine-induced reinstatement of drug seeking. In abuse liability experiments under FR1 contingency, rats maintained responding for heroin (1, 7, 20, 60µg/inf) to a larger extent than cebranopadol (0.03, 0.1, 0.3, 1.0, 6.0µg/inf). Under PR contingency, heroin maintained responding at high levels at all except the lowest dose, while the break point (BP) for cebranopadol did not differ from that of saline. Together, these data indicate that cebranopadol is highly efficacious in attenuating opioid self-administration and stress-induced reinstatement, while having limited abuse liability properties. Overall, the data suggest clinical potential of this compound for OUD treatment.

11.
Neuropharmacology ; 248: 109866, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38364970

RESUMEN

The Nociceptin/Orphanin FQ (N/OFQ) peptide and its receptor NOP are highly expressed within several regions of the mesolimbic system, including the ventral tegmental area (VTA). Evidence indicates that the N/OFQ-NOP receptor system is involved in reward processing and historically it has been proposed that activation of NOP receptors attenuates the motivation for substances of abuse. However, recent findings demonstrated that drug self-administration and relapse to drug-seeking are also attenuated after administration of NOP receptor antagonists. Here, to shed light on the mechanisms through which NOP receptor blockers modulate these processes, we utilized ex vivo patch-clamp recordings to investigate the effect of the selective NOP receptor antagonist LY2817412 on VTA dopaminergic (DA) function in male rats. Results showed that, similar to the endogenous NOP receptor agonist N/OFQ, LY2817412 reduced the spontaneous basal firing discharge of VTA DA neurons. Consistently, we found that NOP receptors are expressed both in VTA DA and GABA cells and that LY2817412 slice perfusion increased GABA release onto VTA DA cells. Finally, in the attempt to dissect the role of postsynaptic and presynaptic NOP receptors, we tested the effect of N/OFQ and LY2817412 in the presence of GABA receptors blockers. Results showed that the effect of LY2817412 was abolished following pretreatment with GABABR, but not GABAAR, blockers. Conversely, inhibition of DA neuronal activity by N/OFQ was unaffected by blockade of GABA receptors. Altogether, these results suggest that both NOP receptor agonists and antagonists can decrease VTA DA neuronal activity, but through distinct mechanisms of action. The effect of NOP receptor antagonists occurs through a GABABR-mediated mechanism while NOP receptor agonists seem to act via a direct effect on VTA DA neurons.


Asunto(s)
Dopamina , Receptores Opioides , Ratas , Masculino , Animales , Receptores Opioides/metabolismo , Área Tegmental Ventral/metabolismo , Receptor de Nociceptina , Receptores de GABA-B , Nociceptina , Neuronas Dopaminérgicas/metabolismo , Ácido gamma-Aminobutírico , Péptidos Opioides/farmacología
12.
Addict Biol ; 18(3): 467-79, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22804785

RESUMEN

Anxiety is a key consequence of ethanol withdrawal and important risk factor for relapse. The neuropeptide nociceptin/orphanin FQ (N/OFQ) or agonists at this peptide's receptor (NOP) exert anxiolytic-like and antistress actions. N/OFQ dysfunction has been linked to both a high-anxiety behavioral phenotype and excessive ethanol intake. Recent studies suggest a possible link between genetic polymorphisms of the NOP transcript and alcoholism. Thus, in the present study, the effects of intracerebroventricularly administered N/OFQ were tested for modification of anxiety-like behaviors, using the shock-probe defensive burying and elevated plus-maze tests, in ethanol-dependent versus non-dependent rats, 1 and 3 weeks following termination of ethanol exposure. Additionally, prepro-N/OFQ (ppN/OFQ) and NOP receptor gene expression was measured in the central nucleus of the amygdala, in the bed nucleus of the stria terminalis and in the lateral hypothalamus at the same timepoints in separate subjects. One week post-ethanol, N/OFQ dose-dependently attenuated elevated anxiety-like behavior in ethanol-dependent rats and produced anxiolytic-like effects in non-dependent controls in both behavioral tests. However, 3 weeks post-ethanol, N/OFQ altered behavior consistent with anxiogenic-like actions in ethanol-dependent rats but continued to exert anxiolytic-like actions in non-dependent controls. These findings were paralleled by ethanol history-dependent changes of ppN/OFQ and NOP gene expression that showed a distinctive time course in the examined brain structures. The results demonstrate that ethanol dependence and withdrawal are associated with neuroadaptive changes in the N/OFQ-NOP system, suggesting a role of this neuropeptidergic pathway as a therapeutic target for the treatment of alcohol abuse.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Péptidos Opioides/farmacología , Receptores Opioides/genética , Animales , Ansiedad/genética , Expresión Génica/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratas , Ratas Wistar , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Factores de Tiempo , Receptor de Nociceptina , Nociceptina
13.
Proc Natl Acad Sci U S A ; 107(45): 19567-72, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20974945

RESUMEN

Drug addiction is a chronic relapsing disorder characterized by compulsive drug seeking and use. Environmental conditioning factors are among the major determinants of relapse in abstinent cocaine users. Here we describe a role of the neuropeptide S (NPS) system in regulating relapse. In rats with a history of cocaine self-administration, presentation of stimuli predictive of drug availability reinstates drug seeking, triggering relapse. Intracerebroventricular (ICV) injection of NPS increased conditioned reinstatement of cocaine seeking, whereas peripheral administration of the NPS receptor antagonist SHA 68 reduced it. Manipulation of the NPS receptor system did not modify cocaine self-administration. We also found that ICV NPS administration activates c-Fos expression in hypocretin-1/orexin-A (Hcrt-1/Ox-A) immunoreactive neurons in the lateral hypothalamus (LH) and in the perifornical area (PeF). Of note, intra-LH and intra-PeF administration of NPS increased conditioned reinstatement of cocaine responding, an effect that was selectively blocked with the Hcrt-1/Ox-A receptor selective antagonist SB334867. Finally, results showed that intra-LH injection of the NPS antagonist [D-Cys(tBu) (5)]NPS blocked cue-induced cocaine seeking, indicating a role for this system in the pathophysiology of drug relapse.


Asunto(s)
Trastornos Relacionados con Cocaína/etiología , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Neuropéptidos/fisiología , Animales , Cocaína/administración & dosificación , Señales (Psicología) , Vías de Administración de Medicamentos , Hipotálamo/citología , Neuronas , Neuropéptidos/administración & dosificación , Neuropéptidos/antagonistas & inhibidores , Neurotransmisores , Orexinas , Ratas , Ratas Long-Evans , Recurrencia
14.
bioRxiv ; 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37546836

RESUMEN

The gold standard pharmacological treatment for opioid use disorder (OUD) consists of maintenance therapy with long-acting opioid agonists such as buprenorphine and methadone. Despite these compounds having demonstrated substantial efficacy, a significant number of patients do not show optimal therapeutic responses. Moreover, the abuse liability of these medications remains a major concern. Cebranopadol, is a new, long-acting pan-opioid agonist that also activates the nociception/orphanin FQ NOP receptor. Here we used rats to explore the therapeutic potential of this agent in OUD. First, in operant intravenous self-administration experiments we compared the potential abuse liability of cebranopadol with the prototypical opioid heroin. Under a fixed ratio 1 (FR1) contingency, rats maintained responding for heroin (1, 7, 20, 60 µg/inf) to a larger extent than cebranopadol (0.03, 0.1, 0.3, 1.0, 6.0 µg/inf). When the contingency was switched to a progressive ratio (PR) reinforcement schedule, heroin maintained responding at high levels at all except the lowest dose. Conversely, in the cebranopadol groups responding decreased drastically and the break point (BP) did not differ from saline controls. Next, we demonstrated that oral administration of cebranopadol (0, 25, 50 µg/kg) significantly attenuated drug self-administration independent of heroin dose (1, 7, 20, 60 µg/inf). Cebranopadol also reduced the break point for heroin (20 µg/inf). Furthermore, in a heroin self-administration training extinction/reinstatement paradigm, pretreatment with cebranopadol significantly attenuated yohimbine stress-induced reinstatement of drug seeking. Together, these data indicate that cebranopadol has limited abuse liability compared to heroin and is highly efficacious in attenuating opioid self-administration and stress-induced reinstatement, suggesting clinical potential of this compound for OUD treatment.

15.
Addict Biol ; 17(3): 557-64, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21790902

RESUMEN

Metabotropic glutamate receptors (mGluRs) have been implicated in the regulation of anxiety, stress responses and the neurobehavioral effects of psychostimulants. The present study was designed to examine whether antagonizing mGluR5 or activating mGluR2/3 prevents stress-induced reinstatement of cocaine seeking. Male Wistar rats were trained to self-administer cocaine and then subjected to daily extinction training for 2 weeks. Subsequent exposure to 15 minutes of intermittent footshock elicited robust reinstatement of responding at the previously active lever. Both the selective mGluR5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]piperidine (MTEP) (0-3 mg/kg, intraperitoneally) and the selective mGluR2/3 agonist (-)-2-oxa-4-aminobicylco[3.1.0]hexane-4,6-dicarboxylic acid (LY379268) (0-3 mg/kg, subcutaneously) prevented cocaine seeking induced by footshock stress following the same dose-response function. The data show that although mGluR2/3 and mGluR5 are differentially located on synaptic compartments, both LY379268 and MTEP produced the same behavioral effects in reducing stress-induced reinstatement. These results are important because they demonstrate that a reduction in glutamate-mediated neural excitability (albeit via different mechanisms of action) reverses footshock-induced reinstatement and suggest that pharmacological manipulations of mGluR2/3 and mGluR5 can prevent the effects of stress, a major precipitating factor for relapse. These findings further confirm that mGluR2/3 or mGluR5 are promising targets for relapse prevention.


Asunto(s)
Aminoácidos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Trastornos Relacionados con Cocaína/psicología , Piperidinas/farmacología , Receptores de Glutamato Metabotrópico/fisiología , Estrés Psicológico/psicología , Tiazoles/farmacología , Análisis de Varianza , Animales , Condicionamiento Operante/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Extinción Psicológica/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Receptores de Glutamato Metabotrópico/efectos de los fármacos
16.
Addict Biol ; 17(2): 300-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21762287

RESUMEN

Cocaine-induced neuroadaptation of stress-related circuitry and increased access to cocaine each putatively contribute to the transition from cocaine use to cocaine dependence. The present study tested the hypothesis that rats receiving extended versus brief daily access to cocaine would exhibit regional differences in levels of the stress-regulatory neuropeptide corticotropin-releasing factor (CRF). A secondary goal was to explore how CRF levels change in relation to the time since cocaine self-administration. Male Wistar rats acquired operant self-administration of cocaine and were assigned to receive daily long access (6 hours/day, LgA, n=20) or short access (1 hour/day, ShA, n=18) to intravenous cocaine self-administration (fixed ratio 1, ~0.50 mg/kg/infusion). After at least 3 weeks, tissue CRF immunoreactivity was measured at one of three timepoints: pre-session, post-session or 3 hours post-session. LgA, but not ShA, rats showed increased total session and first-hour cocaine intake. CRF immunoreactivity increased within the dorsal raphe (DR) and basolateral, but not central, nucleus of the amygdala (BLA, CeA) of ShA rats from pre-session to 3 hours post-session. In LgA rats, CRF immunoreactivity increased from pre-session to 3 hours post-session within the CeA and DR but tended to decrease in the BLA. LgA rats showed higher CRF levels than ShA rats in the DR and, pre-session, in the BLA. Thus, voluntary cocaine intake engages stress-regulatory CRF systems of the DR and amygdala. Increased availability of cocaine promotes greater tissue CRF levels in these extrahypothalamic brain regions, changes associated here with a model of cocaine dependence.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Trastornos Relacionados con Cocaína/metabolismo , Cocaína/farmacología , Hormona Liberadora de Corticotropina/metabolismo , Animales , Cocaína/administración & dosificación , Condicionamiento Operante/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Autoadministración , Estrés Psicológico/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo
17.
Br J Pharmacol ; 179(17): 4330-4343, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35411600

RESUMEN

BACKGROUND AND PURPOSE: Learned associations between environmental stimuli and drugs of abuse represent a major factor in the chronically relapsing nature of drug addiction. In drug dependent subjects these associations must be presumed to include associations linked to reversal of adverse withdrawal states by drug use-"withdrawal-associated learning" (WDL). However, their significance in drug seeking has received little experimental scrutiny. EXPERIMENTAL APPROACH: Using alcohol as a drug of abuse, the behavioural consequences of WDL were investigated in animal models of relapse and compulsive drug seeking by comparing the effects of WD L-associated stimuli versus stimuli associated with alcohol without WDL experience in nondependent and post-dependent rats. Brain sites activated by exposure to the respective stimuli were identified by c-fos immunohistochemistry. KEY RESULTS: (1) WDL-associated stimuli elicited significant alcohol seeking. In rats with WDL experience, stimuli associated with alcohol in the nondependent state no longer elicited robust alcohol seeking. (2) Responding elicited by WDL-associated stimuli, but not stimuli conditioned to alcohol in the nondependent state, was resistant to footshock punishment and increased response effort requirements for presentation of WDL-related stimuli. (3) Stimuli conditioned to alcohol in rats with a dependence but not WDL history did not sustain punished responding or tolerance of increased effort. (4) The central nucleus of the amygdala was identified as a site selectively responsive to WDL stimulus exposure. CONCLUSION AND IMPLICATIONS: Environmental stimuli associated with reversal of adverse withdrawal states by alcohol elicit compulsive-like alcohol seeking and establish WDL as a major, not well-recognized factor, in relapse vulnerability.


Asunto(s)
Síndrome de Abstinencia a Sustancias , Trastornos Relacionados con Sustancias , Animales , Conducta Compulsiva , Condicionamiento Operante , Comportamiento de Búsqueda de Drogas , Etanol/farmacología , Humanos , Ratas , Recurrencia , Autoadministración
18.
Br J Pharmacol ; 179(11): 2647-2658, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34854073

RESUMEN

BACKGROUND AND PURPOSE: The nociceptin/orphanin FQ (N/OFQ)-nociceptin opioid-like peptide (NOP) receptor system is widely distributed in the brain and pharmacological activation of this system revealed therapeutic potential in animal models of substance use disorder. Studies also showed that genetic deletion or pharmacological blockade of NOP receptors confer resistance to the development of alcohol abuse. Here, we have used a genetic and pharmacological approach to evaluate the therapeutic potential of NOP antagonism in smoking cessation. EXPERIMENTAL APPROACH: Constitutive NOP receptor knockout rats (NOP-/- ) and their wild-type counterparts (NOP+/+ ) were tested over a range of behaviours to characterize their motivation for nicotine. We next explored the effects of systemic administration of the NOP receptor antagonist LY2817412 (1.0 & 3.0 mg·kg-1 ) on nicotine self-administration. NOP receptor blockade was further evaluated at the brain circuitry level, by microinjecting LY2817412 (3.0 & 6.0 µg·µl-1 ) into the ventral tegmental area (VTA), nucleus accumbens (NAc) and central amygdala (CeA). KEY RESULTS: Genetic NOP receptor deletion resulted in decreased nicotine intake, decreased motivation to self-administer and attenuation of cue-induced nicotine reinstatement. LY2817412 reduced nicotine intake in NOP+/+ but not in NOP-/- rats, confirming that its effect is mediated by inhibition of NOP transmission. Finally, injection of LY2817412 into the VTA but not into the NAc or CeA decreased nicotine self-administration. CONCLUSIONS AND IMPLICATIONS: These findings indicate that inhibition of NOP transmission attenuates the motivation for nicotine through mechanisms involving the VTA and suggest that NOP receptor antagonism may represent a potential treatment for smoking cessation.


Asunto(s)
Nicotina , Área Tegmental Ventral , Animales , Nicotina/farmacología , Péptidos Opioides/metabolismo , Ratas , Receptores Opioides , Área Tegmental Ventral/metabolismo , Receptor de Nociceptina , Nociceptina
19.
Biol Psychiatry Glob Open Sci ; 2(1): 70-78, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35252951

RESUMEN

BACKGROUND: Cannabidiol (CBD) has received attention for the treatment of substance use disorders. In preclinical models of relapse, CBD attenuates drug seeking across several drugs of abuse, including cocaine. However, in these models CBD has not been consistently effective. This inconsistency in CBD effects may be related to presently insufficient information on the full spectrum of CBD dose effects on drug-related behaviors. METHODS: We address this issue by establishing a full dose-response profile of CBD's actions using expression of cocaine-induced conditioned place preference as a model for drug-motivated behavior in male rats and by concurrently identifying dose-dependent effects of CBD on underlying neuronal activation and distinct neuronal phenotypes showing dose-dependent activation changes. Additionally, we established CBD levels in plasma and brain samples. RESULTS: CBD produced linear increases in CBD brain/plasma concentrations but suppressed conditioned place preference in a distinct U-shaped manner. In parallel with its behavioral effects, CBD produced U-shaped suppressant effects on neuronal activation in the prelimbic but not infralimbic cortex or nucleus accumbens core and shell. RNAscope in situ hybridization identified suppression of glutamatergic and GABAergic (gamma-aminobutyric acidergic) signaling in the prelimbic cortex as a possible cellular mechanism for the attenuation of cocaine-induced conditioned place preference by CBD. CONCLUSIONS: The findings extend previous evidence on the potential of CBD in preventing drug-motivated behavior. However, CBD's dose-response profile may have important dosing implications for future clinical applications and may contribute to the understanding of discrepant CBD effects on drug seeking reported in the literature.

20.
Br J Pharmacol ; 179(11): 2589-2609, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35023154

RESUMEN

BACKGROUND AND PURPOSE: 'Food addiction' is the subject of intense public and research interest. However, this nosology based on neurobehavioural similarities among obese individuals, patients with eating disorders and those with substance use disorders (drug addiction) remains controversial. We thus sought to determine which aspects of disordered eating are causally linked to preclinical models of drug addiction. We hypothesized that extensive drug histories, known to cause addiction-like brain changes and drug motivation in rats, would also cause addiction-like food motivation. EXPERIMENTAL APPROACH: Rats underwent extensive cocaine, alcohol, caffeine or obesogenic diet histories and were subsequently tested for punishment-resistant food self-administration or 'compulsive appetite', as a measure of addiction-like food motivation. KEY RESULTS: Extensive cocaine and alcohol (but not caffeine) histories caused compulsive appetite that persisted long after the last drug exposure. Extensive obesogenic diet histories also caused compulsive appetite, although neither cocaine nor alcohol histories caused excess calorie intake and bodyweight during abstinence. Hence, compulsive appetite and obesity appear to be dissociable, with the former sharing common mechanisms with preclinical drug addiction models. CONCLUSION AND IMPLICATIONS: Compulsive appetite, as seen in subsets of obese individuals and patients with binge-eating disorder and bulimia nervosa (eating disorders that do not necessarily result in obesity), appears to epitomize 'food addiction'. Because different drug and obesogenic diet histories caused compulsive appetite, overlapping dysregulations in the reward circuits, which control drug and food motivation independently of energy homeostasis, may offer common therapeutic targets for treating addictive behaviours across drug addiction, eating disorders and obesity.


Asunto(s)
Conducta Adictiva , Cocaína , Adicción a la Comida , Trastornos Relacionados con Sustancias , Animales , Apetito , Conducta Alimentaria , Alimentos , Adicción a la Comida/complicaciones , Humanos , Obesidad/etiología , Preparaciones Farmacéuticas , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA