Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nutr Neurosci ; : 1-14, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38488783

RESUMEN

Objectives: Binge eating disorder (BED) is the most prevalent eating disorder associated with multiple adverse health effects, especially mental health issues, including substance use disorders and mood and anxiety disorders. Given these high comorbidities, the objective of our study was to examine whether bingeing behavior would lead to altered perception of reinforcing properties of EtOH and changes in well-being. Methods: We used a sucrose bingeing model based on an intermittent access paradigm with a two-bottle choice, without fasting, in male and female mice. We examined the effect of 2-week sucrose paradigm on ethanol-reinforcing properties using a conditioned place preference test (CPP). Well-being, anxiety- and depressive-like behavioral tests were performed to assess emotional state following 2 and 8-week sucrose bingeing paradigm. Results: Mice with intermittent access to sucrose developed a binge-like behavior assessed by higher sucrose intake and escalation rate during the 1st hour of access, in comparison with mice with a continuous sucrose access. We show for the first time that sucrose bingeing in mice modifies positive reinforcing effect of EtOH in a CPP paradigm without marked alteration of emotional state. Interestingly, prolonging sucrose access for 8 weeks revealed an exacerbated bingeing behavior in female mice, and some signs of emotional state alterations in female with continuous access. Discussion: In sum, our findings broaden the understanding of behavioral alterations associated with bingeing, highlighting the need to investigate addictive-like properties of palatable food both in male and female mice.

2.
Cereb Cortex ; 32(10): 2140-2155, 2022 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-34628498

RESUMEN

Neuron-derived 17ß-estradiol (E2) alters synaptic transmission and plasticity in brain regions with endocrine and non-endocrine functions. Investigations into a modulatory role of E2 in synaptic activity and plasticity have mainly focused on the rodent hippocampal formation. In songbirds, E2 is synthesized by auditory forebrain neurons and promotes auditory signal processing and memory for salient acoustic stimuli; however, the modulatory effects of E2 on memory-related synaptic plasticity mechanisms have not been directly examined in the auditory forebrain. We investigated the effects of bidirectional E2 manipulations on synaptic transmission and long-term potentiation (LTP) in the rat primary auditory cortex (A1). Immunohistochemistry revealed widespread neuronal expression of the E2 biosynthetic enzyme aromatase in multiple regions of the rat sensory and association neocortex, including A1. In A1, E2 application reduced the threshold for in vivo LTP induction at layer IV synapses, whereas pharmacological suppression of E2 production by aromatase inhibition abolished LTP induction at layer II/III synapses. In acute A1 slices, glutamate and γ-aminobutyric acid (GABA) receptor-mediated currents were sensitive to E2 manipulations in a layer-specific manner. These findings demonstrate that locally synthesized E2 modulates synaptic transmission and plasticity in A1 and suggest potential mechanisms by which E2 contributes to auditory signal processing and memory.


Asunto(s)
Aromatasa , Corteza Auditiva , Animales , Aromatasa/metabolismo , Aromatasa/farmacología , Corteza Auditiva/metabolismo , Estradiol/farmacología , Potenciación a Largo Plazo/fisiología , Masculino , Plasticidad Neuronal/fisiología , Prosencéfalo/metabolismo , Ratas , Sinapsis/fisiología , Transmisión Sináptica
3.
Cogn Affect Behav Neurosci ; 22(4): 736-753, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35396630

RESUMEN

Anhedonia is characteristically preceded by chronic stress, likely involving downstream effects of glucocorticoid alterations on dopamine (DA) function. To elucidate the neural underpinnings of this interaction, we examined whether acute pharmacological modulation of DA alters reward learning after chronic mild stress (CMS). Forty-eight male Wistar rats were exposed to a 21-day CMS regime (n = 48 no stress controls) before completing the probabilistic reward task (PRT), a well-validated cross-species test of reward learning. We first examined whether stress-induced reward dysfunction could be restored by systemic injections of low-dose amisulpride (AMI), which increases DA transmission via D2-like autoreceptor blockade. Then, we investigated region-specific effects through bilateral infusions of quinpirole (QUIN), a D2-like receptor agonist, into either the nucleus accumbens core (NAcc) or medial prefrontal cortex (mPFC). Blunted reward learning in CMS animals was reversed by acute AMI administration, but this treatment did not alter reward learning in the no stress group. Elevated adrenal gland weight, a proxy for stress reactivity, predicted lower reward learning in the untreated CMS group. This effect was extinguished following AMI treatment. These findings might be attributed to significantly higher D2 receptor density in the NAcc of high stress reactive animals. To this end, NAcc QUIN infusions potentiated reward learning relative to mPFC QUIN infusions in CMS rats, but there was no effect in no stress control rats. Collectively, these findings suggest that DA modulation reverses stress-induced reward dysfunction, even among the most stress-reactive animals. The effect might depend on D2-like receptor activation in the mesolimbic system.


Asunto(s)
Agonistas de Dopamina , Dopamina , Animales , Dopamina/fisiología , Agonistas de Dopamina/farmacología , Masculino , Núcleo Accumbens/fisiología , Quinpirol/farmacología , Ratas , Ratas Wistar , Recompensa
4.
J Neurosci Res ; 100(1): 297-308, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-31721270

RESUMEN

The rewarding effect of opiates is mediated through dissociable neural systems in drug naïve and drug-dependent states. Neuroadaptations associated with chronic drug use are similar to those produced by chronic pain, suggesting that opiate reward could also involve distinct mechanisms in chronic pain and pain-naïve states. We tested this hypothesis by examining the effect of dopamine (DA) antagonism on morphine reward in a rat model of neuropathic pain.Neuropathic pain was induced in male Sprague-Dawley rats through chronic constriction (CCI) of the sciatic nerve; reward was assessed in the conditioned place preference (CPP) paradigm in separate groups at early (4-8 days post-surgery) and late (11-15 days post-surgery) phases of neuropathic pain. Minimal effective doses of morphine that produced a CPP in early and late phases of neuropathic pain were 6 mg/kg and 2 mg/kg respectively. The DA D1 receptor antagonist, SCH23390, blocked a morphine CPP in sham, but not CCI, rats at a higher dose (0.5 mg/kg), but had no effect at a lower dose (0.1 mg/kg). The DA D2 receptor antagonist, eticlopride (0.1 and 0.5 mg/kg), had no effect on a morphine CPP in sham or CCI rats, either in early or late phases of neuropathic pain. In the CPP paradigm, morphine reward involves DA D1 mechanisms in pain-naïve but not chronic pain states. This could reflect increased sensitivity to drug effects in pain versus no pain conditions and/or differential mediation of opiate reward in these two states.


Asunto(s)
Dolor Crónico , Morfina , Animales , Dolor Crónico/tratamiento farmacológico , Masculino , Morfina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1 , Recompensa
5.
J Neurosci Res ; 100(1): 129-148, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-32623788

RESUMEN

Delta opioid receptor (DOR) agonists alleviate nociceptive behaviors in various chronic pain models, including neuropathic pain, while having minimal effect on sensory thresholds in the absence of injury. The mechanisms underlying nerve injury-induced enhancement of DOR function are unclear. We used a peripheral nerve injury (PNI) model of neuropathic pain to assess changes in the function and localization of DORs in mice and rats. Intrathecal administration of DOR agonists reversed mechanical allodynia and thermal hyperalgesia. The dose-dependent thermal antinociceptive effects of DOR agonists were shifted to the left in PNI rats. Administration of DOR agonists produced a conditioned place preference in PNI, but not in sham, animals, whereas the DOR antagonist naltrindole produced a place aversion in PNI, but not in sham, mice, suggesting the engagement of endogenous DOR activity in suppressing pain associated with the injury. GTPγS autoradiography revealed an increase in DOR function in the dorsal spinal cord, ipsilateral to PNI. Immunogold electron microscopy and in vivo fluorescent agonist assays were used to assess changes in the ultrastructural localization of DORs in the spinal dorsal horn. In shams, DORs were primarily localized within intracellular compartments. PNI significantly increased the cell surface expression of DORs within lamina IV-V dendritic profiles. Using neonatal capsaicin treatment, we identified that DOR agonist-induced thermal antinociception was mediated via receptors expressed on primary afferent sensory neurons but did not alter mechanical thresholds. These data reveal that the regulation of DORs following PNI and suggest the importance of endogenous activation of DORs in regulating chronic pain states.


Asunto(s)
Neuralgia , Receptores Opioides delta , Analgésicos Opioides/efectos adversos , Animales , Modelos Animales de Enfermedad , Hiperalgesia/inducido químicamente , Ratones , Neuralgia/metabolismo , Ratas
6.
Eur J Nutr ; 60(8): 4621-4633, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34165614

RESUMEN

OBJECTIVES: Increased availability of high-calorie palatable food in most countries has resulted in overconsumption of these foods, suggesting that excessive eating is driven by pleasure, rather than metabolic need. The behavior contributes to the rise in eating disorders, obesity, and associated pathologies like diabetes, cardiac disease, and cancers. The mesocorticolimbic dopamine and homeostatic circuits are interconnected and play a central role in palatable food intake. The endocannabinoid system is expressed in these circuits and represents a potent regulator of feeding, but the impact of an obesogenic diet on its expression is not fully known. METHODS: Food intake and body weight were recorded in male Wistar rats over a 6-week free-choice regimen of high fat and sugar; transcriptional regulations of the endocannabinoid system were examined post-mortem in brain reward regions (prefrontal cortex, nucleus accumbens, ventral tegmental area, and arcuate nucleus). K-means cluster analysis was used to classify animals based on individual sensitivity to obesity and palatable food intake. Endocannabinoid levels were quantified in the prefrontal cortex and nucleus accumbens. Gene expression in dopamine and homeostatic systems, including ghrelin and leptin receptors, and classical homeostatic peptides, were also investigated. RESULTS: The free-choice high-fat -and sugar diet induced hyperphagia and obesity in rats. Cluster analysis revealed that the propensity to develop obesity and excessive palatable food intake was differently associated with dopamine and endocannabinoid system gene expression in reward and homeostatic brain regions. CB2 receptor mRNA was increased in the nucleus accumbens of high sugar consumers, whereas CB1 receptor mRNA was decreased in obesity prone rats. CONCLUSIONS: Transcriptional data are consistent with observations of altered dopamine function in rodents that have access to an obesogenic diet and point to cannabinoid receptors as GPCR targets involved in neuroplasticity mechanisms associated with maladaptive intake of palatable food.


Asunto(s)
Dieta , Endocannabinoides , Animales , Encéfalo , Análisis por Conglomerados , Ingestión de Alimentos , Masculino , Obesidad/etiología , Ratas , Ratas Wistar , Recompensa
7.
Appetite ; 164: 105258, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33864862

RESUMEN

Binge eating, the defining feature of binge eating disorder (BED), is associated with a number of adverse health outcomes as well as a reduced quality of life. Animals, like humans, selectively binge on highly palatable food suggesting that the behaviour is driven by hedonic, rather than metabolic, signals. Given the links to both reward processing and food intake, this study examined the contribution of the endocannabinoid system (ECS) to binge-like eating in rats. Separate groups were given intermittent (12 h) or continuous (24 h) access to 10% sucrose and food over 28 days, with only the 12 h access group displaying excessive sucrose intake within a discrete period of time (i.e., binge eating). Importantly, this group also exhibited alterations in ECS transcripts and endocannabinoid levels in brain reward regions, including an increase in cannabinoid receptor 1 (CB1R) mRNA in the nucleus accumbens as well as changes in endocannabinoid levels in the prefrontal cortex and hippocampus. We then tested whether different doses (1 and 3 mg/kg) of a CB1R antagonist, Rimonabant, modify binge-like intake or the development of a conditioned place preference (CPP) to sucrose. CB1R blockade reduced binge-like intake of sucrose and blocked a sucrose CPP, but only in rats that had undergone 28 days of sucrose consumption. These findings indicate that sucrose bingeing alters the ECS in reward-related areas, modifications that exacerbate the effect of CB1R blockade on sucrose reward. Overall, our results broaden the understanding of neural alterations associated with bingeing eating and demonstrate an important role for CB1R mechanisms in reward processing. In addition, these findings have implications for understanding substance abuse, which is also characterized by excessive and maladaptive intake, pointing towards addictive-like properties of palatable food.


Asunto(s)
Trastorno por Atracón , Animales , Ingestión de Alimentos , Endocannabinoides , Conducta Alimentaria , Calidad de Vida , Ratas , Sacarosa
8.
J Neurosci ; 39(21): 4162-4178, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-30862664

RESUMEN

Pain is a multidimensional experience and negative affect, or how much the pain is "bothersome", significantly impacts the sufferers' quality of life. It is well established that the κ opioid system contributes to depressive and dysphoric states, but whether this system contributes to the negative affect precipitated by the occurrence of chronic pain remains tenuous. Using a model of persistent pain, we show by quantitative real-time-PCR, florescence in situ hybridization, Western blotting and GTPgS autoradiography an upregulation of expression and the function of κ opioid receptors (KORs) and its endogenous ligand dynorphin in the mesolimbic circuitry in animals with chronic pain compared with surgical controls. Using in vivo microdialysis and microinjection of drugs into the mesolimbic dopamine system, we demonstrate that inhibiting KORs reinstates evoked dopamine release and reward-related behaviors in chronic pain animals. Chronic pain enhanced KOR agonist-induced place aversion in a sex-dependent manner. Using various place preference paradigms, we show that activation of KORs drives pain aversive states in male but not female mice. However, KOR antagonist treatment was effective in alleviating anxiogenic and depressive affective-like behaviors in both sexes. Finally, ablation of KORs from dopamine neurons using AAV-TH-cre in KORloxP mice prevented pain-induced aversive states as measured by place aversion assays. Our results strongly support the use of KOR antagonists as therapeutic adjuvants to alleviate the emotional, tonic-aversive component of chronic pain, which is argued to be the most significant component of the pain experience that impacts patients' quality of life.SIGNIFICANCE STATEMENT We show that KORs are sufficient to drive the tonic-aversive component of chronic pain; the emotional component of pain that is argued to significantly impact a patient's quality of life. The impact of our study is broadly relevant to affective disorders associated with disruption of reward circuitry and thus likely contributes to many of the devastating sequelae of chronic pain, including the poor response to treatment of many patients, debilitating affective disorders (other disorders including anxiety and depression that demonstrate high comorbidity with chronic pain) and substance abuse. Indeed, coexisting psychopathology increases pain intensity, pain-related disability and effectiveness of treatments (Jamison and Edwards, 2013).


Asunto(s)
Dolor Crónico/metabolismo , Dolor Crónico/psicología , Emociones/fisiología , Percepción del Dolor/fisiología , Receptores Opioides kappa/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Long-Evans
9.
Behav Pharmacol ; 31(2&3): 249-255, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31503073

RESUMEN

Binge eating in humans is driven by hedonic properties of food, suggesting that brain reward systems may contribute to this behaviour. We examined the role of mu opioid receptors (MOP) in binge eating by examining sweet solution intake in mice with genetic deletion of the MOP. Wildtype and MOP knockout mice had 4 hours access to food in the home cage combined with limited (4 hours) access to sucrose (17.1% w/v) or saccharin (0.09% w/v), or continuous (24 hours) access to sucrose. Only limited access groups exhibited binge intake, measured as increased solution consumption during the first hour. Knockout mice consumed less solution and food during the first hour as well as less food each day compared with wildtype mice. Limited access groups consumed more food and gained more weight than continuous access groups, and the effect was magnified in saccharin-consuming mice. Indeed, the increased food consumption in animals given limited access to saccharin was so excessive that caloric intake of this group was significantly higher than either of the sucrose groups (limited or continuous access). Within this group, females consumed more food per bodyweight than males, highlighting important sex differences in feeding behaviours under restricted access schedules.


Asunto(s)
Bulimia/fisiopatología , Conducta Alimentaria/fisiología , Receptores Opioides mu/metabolismo , Animales , Trastorno por Atracón , Peso Corporal , Bulimia/metabolismo , Modelos Animales de Enfermedad , Ingestión de Alimentos/psicología , Ingestión de Energía/fisiología , Femenino , Preferencias Alimentarias/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Opioides mu/fisiología , Recompensa , Sacarosa/metabolismo
10.
Behav Pharmacol ; 29(2 and 3-Spec Issue): 241-254, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29481424

RESUMEN

Long-term use of opioid analgesics is limited by tolerance development and undesirable adverse effects. Paradoxically, spinal administration of ultra-low-dose (ULD) G-protein-coupled receptor antagonists attenuates analgesic tolerance. Here, we determined whether systemic ULD α2-adrenergic receptor (AR) antagonists attenuate the development of morphine tolerance, whether these effects extend to the cannabinoid (CB1) receptor system, and if behavioral effects are reflected in changes in opioid-induced spinal gliosis. Male rats were treated daily with morphine (5 mg/kg) alone or in combination with ULD α2-AR (atipamezole or efaroxan; 17 ng/kg) or CB1 (rimonabant; 5 ng/kg) antagonists; control groups received ULD injections only. Thermal tail flick latencies were assessed across 7 days, before and 30 min after the injection. On day 8, spinal cords were isolated, and changes in spinal gliosis were assessed through fluorescent immunohistochemistry. Both ULD α2-AR antagonists attenuated morphine tolerance, whereas the ULD CB1 antagonist did not. In contrast, both ULD atipamezole and ULD rimonabant attenuated morphine-induced microglial reactivity and astrogliosis in deep and superficial spinal dorsal horn. So, although paradoxical effects of ULD antagonists are common to several G-protein-coupled receptor systems, these may not involve similar mechanisms. Spinal glia alone may not be the main mechanism through which tolerance is modulated.


Asunto(s)
Tolerancia a Medicamentos/fisiología , Morfina/metabolismo , Neuroglía/efectos de los fármacos , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Animales , Benzofuranos/farmacología , Antagonistas de Receptores de Cannabinoides/farmacología , Cannabinoides , Relación Dosis-Respuesta a Droga , Gliosis/inducido químicamente , Imidazoles/farmacología , Inyecciones Espinales/métodos , Masculino , Morfina/farmacología , Norepinefrina , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1 , Rimonabant/farmacología , Columna Vertebral/efectos de los fármacos
11.
J Neurosci ; 35(22): 8442-50, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041913

RESUMEN

Chronic pain attenuates midbrain dopamine (DA) transmission, as evidenced by a decrease in opioid-evoked DA release in the ventral striatum, suggesting that the occurrence of chronic pain impairs reward-related behaviors. However, mechanisms by which pain modifies DA transmission remain elusive. Using in vivo microdialysis and microinjection of drugs into the mesolimbic DA system, we demonstrate in mice and rats that microglial activation in the VTA compromises not only opioid-evoked release of DA, but also other DA-stimulating drugs, such as cocaine. Our data show that loss of stimulated extracellular DA is due to impaired chloride homeostasis in midbrain GABAergic interneurons. Treatment with minocycline or interfering with BDNF signaling restored chloride transport within these neurons and recovered DA-dependent reward behavior. Our findings demonstrate that a peripheral nerve injury causes activated microglia within reward circuitry that result in disruption of dopaminergic signaling and reward behavior. These results have broad implications that are not restricted to the problem of pain, but are also relevant to affective disorders associated with disruption of reward circuitry. Because chronic pain causes glial activation in areas of the CNS important for mood and affect, our findings may translate to other disorders, including anxiety and depression, that demonstrate high comorbidity with chronic pain.


Asunto(s)
Dolor Crónico/patología , Sistema Límbico/patología , Microglía/patología , Red Nerviosa/patología , Recompensa , Animales , Área Bajo la Curva , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/etiología , Cocaína/uso terapéutico , Condicionamiento Clásico/efectos de los fármacos , Modelos Animales de Enfermedad , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Minociclina/uso terapéutico , Morfina/uso terapéutico , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Neuropatía Ciática/complicaciones , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiología
12.
Behav Pharmacol ; 27(7): 585-95, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27509312

RESUMEN

Both impulsivity and stress are risk factors for substance abuse, but it is not clear how these two processes interact to alter susceptibility for the disorder. The aim of this project was to examine the pharmacology of a stress-impulsivity interaction in rats. To do so, we tested the effects of yohimbine on impulsive action and then assessed whether behavioural changes could be reduced by antagonists at different receptor subtypes. Male Long-Evans rats were injected with various doses of yohimbine (0-5.0 mg/kg) before testing in the response-inhibition task. In subsequent experiments, yohimbine (2.5 mg/kg) was injected following pretreatment with the following receptor antagonists: corticotropin-releasing factor receptor 1, antalarmin (0-20 mg/kg); glucocorticoid, mifepristone (0-30 mg/kg); noradrenergic (NA) α1, prazosin (0-2 mg/kg); NA α2, guanfacine (0-0.5 mg/kg); NA ß2, propranolol (0.5-2.0 mg/kg); dopamine D1/5, SCH 39166 (0-0.0625 mg/kg); µ opioid, naloxone (0-2 mg/kg); or 5-HT2A, M100907 (0.005-0.05 mg/kg). In all experiments, impulsive action was measured as increased premature responding. Yohimbine dose dependently increased impulsive action, but the effect was not reversed by antagonist pretreatment. None of the drugs altered any other behavioural measure. We conclude that stress-impulsivity interactions are likely mediated by a synergy of multiple neurotransmitter systems.


Asunto(s)
Conducta Animal/efectos de los fármacos , Conducta Impulsiva/efectos de los fármacos , Estrés Psicológico/metabolismo , Yohimbina/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Neurotransmisores/metabolismo , Ratas , Ratas Long-Evans , Tiempo de Reacción/efectos de los fármacos , Yohimbina/administración & dosificación
13.
Behav Pharmacol ; 24(3): 207-13, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23591124

RESUMEN

In addition to sensory disturbances, neuropathic pain is associated with an ongoing and persistent negative affective state. This condition may be reflected as altered sensitivity to rewarding stimuli. We examined this hypothesis by testing whether the rewarding properties of morphine are altered in a rat model of neuropathic pain. Neuropathic pain was induced by chronic constriction of the common sciatic nerve. Drug reward was assessed using an unbiased, three-compartment conditioned place preference (CPP) paradigm. The rats underwent two habituation sessions beginning 6 days after surgery. Over the next 8 days, they were injected with drug or vehicle and were confined to one CPP compartment for 30 min. On the following test day, the rats had access to all three compartments for 30 min. Consistent with the literature, systemic administration of morphine dose-dependently increased the CPP in pain-naive animals. In rats with neuropathic pain, however, the dose-dependent effects of morphine were in a bell-shaped curve, with a low dose of morphine (2 mg/kg) producing a greater CPP than a higher dose of morphine (8 mg/kg). In a separate group of animals, acute administration of morphine reversed mechanical allodynia in animals with neuropathic pain at the same doses that produced a CPP. The increased potency of systemic morphine to produce a CPP in animals with neuropathic pain suggests that the motivation for opioid-induced reward is different in the two states.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Condicionamiento Operante/efectos de los fármacos , Morfina/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuralgia/psicología , Recompensa , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Masculino , Neuralgia/fisiopatología , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Long-Evans , Factores de Tiempo
14.
Alcohol Clin Exp Res ; 36(4): 594-603, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22004189

RESUMEN

BACKGROUND: Most adults in Western society consume alcohol regularly without negative consequences. For a small subpopulation, however, drinking can quickly progress to excessive and chronic intake. Given the dangers associated with alcohol abuse, it is critical to identify traits that may place an individual at risk for developing these behaviors. To that end, we used a rat model to determine whether anxiety-related behaviors, novelty seeking, or cognitive flexibility predict excessive alcohol drinking under both limited and continuous access conditions. METHODS: Adult male rats were assessed in a series of behavioral tasks (elevated plus maze [EPM], locomotor activity, and discrimination/reversal learning in a Y-maze) followed by 6 weeks of daily, 1-hour access to alcohol in a free-choice, 2-bottle paradigm (10% alcohol vs. tap water). Next, subjects were given the opportunity to consume alcohol for 72 hours in drinking chambers that permit separate measures of each drinking bout. Half of the animals experienced a 2-week deprivation period between the limited and continuous access sessions. RESULTS: Time spent on the open arms of the EPM, but not novelty seeking or discrimination/reversal learning, predicted alcohol consumption during limited, 1-h/d access sessions to alcohol. Anxiety-related behavior also predicted the escalation of intake when animals were given 72 hours of continuous access to alcohol. Bout size, but not frequency, was responsible for the increased consumption by high-anxiety subjects during this period. Finally, intake during limited access sessions predicted intake during continuous access, but only in subjects with low intake during limited access. CONCLUSIONS: These findings confirm that preexisting anxiety-related behavior predicts alcohol intake under several schedules of alcohol access. Moreover, when access is unlimited, the high-anxiety-related group exhibited an increase in bout size, but not frequency, of drinking. In addition, we show that modest intake when alcohol is restricted may or may not progress to excessive intake when the drug is freely available.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Ansiedad/psicología , Conducta Animal , Cognición/fisiología , Conducta Exploratoria/fisiología , Animales , Aprendizaje Discriminativo/efectos de los fármacos , Predicción , Modelos Lineales , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Ratas , Ratas Long-Evans , Aprendizaje Inverso/efectos de los fármacos
15.
Behav Pharmacol ; 23(1): 105-12, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22157142

RESUMEN

The objective of this study was to test the hypothesis that prenatal exposure to ethanol, through maternal consumption of an aqueous ethanol solution, induces neurobehavioral deficits and increases ethanol preference in offspring. Pregnant Dunkin-Hartley-strain guinea pigs were given 24-h access to an aqueous ethanol solution (5%, v/v) sweetened with sucralose (1 g/l), or water sweetened with sucralose (1 g/l), throughout gestation. Spontaneous locomotor activity was measured in the offspring on postnatal day (PD) 10. The offspring underwent either ethanol preference testing using a two-bottle-choice paradigm beginning on PD 40 or Morris water maze testing using a hidden moving platform design beginning on PD 60. Maternal consumption of a 5% (v/v) ethanol solution (average daily dose of 2.3±0.1 g of ethanol/kg maternal body weight; range: 1.8-2.8 g/kg) decreased offspring birth weight, increased spontaneous locomotor activity, and increased preference for an aqueous ethanol solution. In the Morris water maze test, sucralose-exposed offspring decreased escape latency on the second day of testing, whereas the ethanol-exposed offspring showed no improvement. These data demonstrate that moderate maternal consumption of ethanol produces hyperactivity, enhances ethanol preference, and impairs learning and memory in guinea pig offspring.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Etanol/toxicidad , Feto/efectos de los fármacos , Animales , Femenino , Cobayas , Intercambio Materno-Fetal , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Embarazo
16.
Mol Neurobiol ; 59(3): 1896-1911, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35032317

RESUMEN

Cocaine addiction is a complex pathology inducing long-term neuroplastic changes that, in turn, contribute to maladaptive behaviors. This behavioral dysregulation is associated with transcriptional reprogramming in brain reward circuitry, although the mechanisms underlying this modulation remain poorly understood. The endogenous cannabinoid system may play a role in this process in that cannabinoid mechanisms modulate drug reward and contribute to cocaine-induced neural adaptations. In this study, we investigated whether cocaine self-administration induces long-term adaptations, including transcriptional modifications and associated epigenetic processes. We first examined endocannabinoid gene expression in reward-related brain regions of the rat following self-administered (0.33 mg/kg intravenous, FR1, 10 days) cocaine injections. Interestingly, we found increased Cnr1 expression in several structures, including prefrontal cortex, nucleus accumbens, dorsal striatum, hippocampus, habenula, amygdala, lateral hypothalamus, ventral tegmental area, and rostromedial tegmental nucleus, with most pronounced effects in the hippocampus. Endocannabinoid levels, measured by mass spectrometry, were also altered in this structure. Chromatin immunoprecipitation followed by qPCR in the hippocampus revealed that two activating histone marks, H3K4Me3 and H3K27Ac, were enriched at specific endocannabinoid genes following cocaine intake. Targeting CB1 receptors using chromosome conformation capture, we highlighted spatial chromatin re-organization in the hippocampus, as well as in the nucleus accumbens, suggesting that destabilization of the chromatin may contribute to neuronal responses to cocaine. Overall, our results highlight a key role for the hippocampus in cocaine-induced plasticity and broaden the understanding of neuronal alterations associated with endocannabinoid signaling. The latter suggests that epigenetic modifications contribute to maladaptive behaviors associated with chronic drug use.


Asunto(s)
Cannabinoides , Cocaína , Animales , Cannabinoides/farmacología , Cocaína/farmacología , Hipocampo/metabolismo , Masculino , Núcleo Accumbens/metabolismo , Ratas , Receptores de Cannabinoides/metabolismo , Autoadministración
17.
J Neurosci ; 30(34): 11493-500, 2010 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-20739571

RESUMEN

Impulse control suppresses actions that are inappropriate in one context, but may be beneficial in others. The medial prefrontal cortex (mPFC) mediates this process by providing a top-down signal to inhibit competing responses, although the mechanism by which the mPFC acquires this ability is unknown. To that end, we examined synaptic changes in the mPFC associated with learning to inhibit an incorrect response. Rats were trained in a simple response inhibition task to withhold responding until a signal was presented. We then measured synaptic plasticity of excitatory synapses in the mPFC, using whole-cell patch-clamp recordings, in brain slices prepared from trained rats. Response inhibition training significantly increased the relative contribution of AMPA receptors to the overall EPSC in prelimbic, but not infralimbic, neurons of the mPFC. This potentiation of synaptic transmission closely paralleled the acquisition and extinction of response inhibition. Using a retrograde fluorescent tracer, we observed that these plastic changes were selective for efferents projecting to the ventral striatum, but not the dorsal striatum or amygdala. Therefore, we suggest that response inhibition is encoded by a selective strengthening of a subset of corticostriatal projections, uncovering a synaptic mechanism of impulse control. This information could be exploited in therapeutic interventions for disorders of impulse control, such as addiction, attention deficit-hyperactivity disorder, and schizophrenia.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Inhibición Neural/fisiología , Corteza Prefrontal/fisiología , Receptores AMPA/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Sinapsis/fisiología , Animales , Condicionamiento Operante/fisiología , Masculino , Ratas , Ratas Long-Evans
18.
Horm Behav ; 59(4): 465-72, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21272586

RESUMEN

This experiment used both biological and self-report measures to examine how alcohol modifies stress responses, and to test whether the interaction between these two factors alters risk-taking in healthy young adults. Participants were divided into stress or no-stress conditions and then further divided into one of three beverage groups. The alcohol group consumed a binge-drinking level of alcohol; the placebo group consumed soda, but believed they were consuming alcohol; the sober group was aware that they were not consuming alcohol. Following beverage consumption, the stress group was subjected to the Trier Social Stress Test (TSST) while the no-stress group completed crossword puzzles; all participants subsequently completed a computerized risk-taking task. Exposure to the TSST significantly increased salivary levels of the hormone cortisol and the enzyme alpha-amylase, as well as subjective self-ratings of anxiety and tension. In the stress condition, both placebo and intoxicated groups reported less tension and anxiety, and exhibited a smaller increase in cortisol, following the TSST than did the sober group. Thus, the expectation of receiving alcohol altered subjective and physiological responses to the stressor. Neither alcohol nor stress increased risk taking, however the sober group demonstrated lower risk-taking on the computer task on the second session. These findings clearly demonstrate that the expectation of alcohol (placebo) alters subsequent physiological responses to stress.


Asunto(s)
Etanol/administración & dosificación , Hidrocortisona/metabolismo , Asunción de Riesgos , Conducta Social , Estrés Psicológico/metabolismo , Adulto , Afecto/efectos de los fármacos , Análisis de Varianza , Femenino , Humanos , Masculino , Placebos , Saliva/efectos de los fármacos , Saliva/metabolismo , Encuestas y Cuestionarios
19.
Behav Pharmacol ; 22(5-6): 558-63, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21555938

RESUMEN

Ultralow-dose opioid antagonists augment the antinociceptive effect of morphine and block the development of tolerance to repeated morphine injections in rodents, but the effects are not reliably reproduced in humans. One explanation for this discrepancy is that preclinical studies of ultralow-dose antagonism in rodents generally use reflex-withdrawal tests of antinociception, which may be affected by cataleptic effects of morphine. We tested this hypothesis by examining whether ultralow-dose naltrexone alters the cataleptic effect of morphine or the development of tolerance to morphine-induced catalepsy. Rats (N=56) were randomly assigned to saline, morphine (10 mg/kg), cotreatments of morphine plus naltrexone (molar ratios of 1,000,000 : 1; 500,000 : 1; 100,000 : 1), or naltrexone-alone groups. Rats were injected with drug for 7 consecutive days; on each day, catalepsy and antinociception were assessed 30 and 60 min postinjection, using the bar and tail-flick tests, respectively. Ultralow-dose naltrexone (500,000 : 1) extended the antinociceptive effect of morphine within a session and attenuated the development of tolerance to the antinociceptive effect of morphine across sessions. Naltrexone alone had no effect on either test. These data show that the paradoxical effect of ultralow-dose naltrexone on antinociception is not the product of morphine-induced catalepsy, pointing to an important role for agonist-antagonist combinations in the clinical treatment of pain.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Analgésicos Opioides/administración & dosificación , Animales , Catalepsia/inducido químicamente , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Masculino , Morfina/administración & dosificación , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Dolor/tratamiento farmacológico , Ratas , Ratas Long-Evans , Factores de Tiempo
20.
Cannabis Cannabinoid Res ; 6(2): 137-147, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33912678

RESUMEN

Background: Cannabinoid receptors play a key role in regulating numerous physiological processes, including immune function and reward signaling. Originally, endocannabinoid contributions to central nervous system processes were attributed to CB1 receptors, but technological advances have confirmed the expression of CB2 receptors in both neurons and glia throughout the brain. Mapping of these receptors is less extensive than for CB1 receptors, and it is still not clear how CB2 receptors contribute to processes that involve endocannabinoid signaling. Objectives: The goal of our study was to assess the effects of peripheral nerve injury and chronic morphine administration, two manipulations that alter endocannabinoid system function, on CB2 receptor expression in the spinal dorsal horn of rats. Methods: Twenty-four male Sprague Dawley rats were assigned to chronic constriction injury (CCI), sham surgery, or pain naïve groups, with half of each group receiving once daily injections of morphine (5 mg/kg) for 10 days. On day 11, spinal cords were isolated and prepared for fluorescent immunohistochemistry. Separate sections from the deep and superficial dorsal horn were stained for neuronal nuclei (NeuN), CD11b, or 4',6-diamidino-2-phenylindole (DAPI) to mark neurons, microglia, and cell nuclei, respectively. Double labeling was used to assess colocalization of CB2 receptors with NeuN or microglial markers. Quantification of mean pixel intensity for each antibody was assessed using a fluorescent microscope, and CB2 receptor expressing cells were also counted manually. Results: Surgery increased DAPI cell counts in the deep and superficial dorsal horn, with CCI rats displaying increased CD11b labeling ipsilateral to the nerve injury. Surgery also decreased NeuN labeling in both regions, an effect that was blocked by morphine administration. CB2 receptors were expressed, predominantly, on NeuN-labeled cells with significant increases in CB2 receptor labeling across all surgery groups in both deep and superficial areas following morphine administration. Conclusions: Our findings provide supporting evidence for the expression of CB2 receptors on neurons and reveal upregulation of receptor expression in the dorsal spinal cord following surgery and chronic morphine administration, with the latter producing a larger effect. Synergistic effects of morphine-cannabinoid treatments, therefore, may involve CB2-mu opioid receptor interactions, pointing to novel therapeutic treatments for a variety of medical conditions.


Asunto(s)
Morfina , Traumatismos de los Nervios Periféricos , Animales , Masculino , Morfina/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB2 , Asta Dorsal de la Médula Espinal , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA