Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Handb Exp Pharmacol ; 248: 311-343, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29374839

RESUMEN

Neural mechanisms underlying alcohol use disorder remain elusive, and this lack of understanding has slowed the development of efficacious treatment strategies for reducing relapse rates and prolonging abstinence. While synaptic adaptations produced by chronic alcohol exposure have been extensively characterized in a variety of brain regions, changes in intrinsic excitability of critical projection neurons are understudied. Accumulating evidence suggests that prolonged alcohol drinking and alcohol dependence produce plasticity of intrinsic excitability as measured by changes in evoked action potential firing and after-hyperpolarization amplitude. In this chapter, we describe functional changes in cell firing of projection neurons after long-term alcohol exposure that occur across species and in multiple brain regions. Adaptations in calcium-activated (KCa2), voltage-dependent (KV7), and G protein-coupled inwardly rectifying (Kir3 or GIRK) potassium channels that regulate the evoked firing and after-hyperpolarization parallel functional changes in intrinsic excitability induced by chronic alcohol. Moreover, there are strong genetic links between alcohol-related behaviors and genes encoding KCa2, KV7, and GIRK channels, and pharmacologically targeting these channels reduces alcohol consumption and alcohol-related behaviors. Together, these studies demonstrate that chronic alcohol drinking produces adaptations in KCa2, KV7, and GIRK channels leading to impaired regulation of the after-hyperpolarization and aberrant cell firing. Correcting the deficit in the after-hyperpolarization with positive modulators of KCa2 and KV7 channels and altering the GIRK channel binding pocket to block the access of alcohol represent a potentially highly effective pharmacological approach that can restore changes in intrinsic excitability and reduce alcohol consumption in affected individuals.


Asunto(s)
Potenciales de Acción , Alcoholismo , Neuronas/efectos de los fármacos , Canales de Potasio/fisiología , Consumo de Bebidas Alcohólicas , Etanol/farmacología , Humanos
2.
Handb Exp Pharmacol ; 248: 619, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30810861

RESUMEN

In section 8.1 on the 10th line in first paragraph the reference citation Mateos-Aparicio et al. 2014 is incorrect.

3.
Brain Behav Immun ; 51: 258-267, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26365025

RESUMEN

Proinflammatory cytokines have been implicated in alcohol-induced neurodegeneration, but the role of the neuroimmune system in alcohol related behaviors has only recently come to the forefront. Herein, the effects of binge-like drinking on IL-1ß mRNA and immunoreactivity within the amygdala were measured following the "drinking in the dark" (DID) paradigm, a model of binge-like ethanol drinking in C57BL/6J mice. Moreover, the role of IL-1 receptor signaling in the amygdala on ethanol consumption was assessed. Results indicated that a history of binge-like ethanol drinking promoted a significant increase of IL-1ß mRNA expression within the amygdala, and immunohistochemistry analyses revealed that the basolateral amygdala (BLA), but not central amygdala (CeA), exhibited significantly increased IL-1ß immunoreactivity. However, Fluoro-Jade® C labeling indicated that multiple cycles of the DID paradigm were not sufficient to elicit neuronal death. Bilateral infusions of IL-1 receptor antagonist (IL-1Ra) reduced ethanol consumption when infused into the BLA but not the CeA. These observations were specific to ethanol drinking as the IL-1Ra did not alter either sucrose drinking or open-field locomotor activity. The current findings highlight a specific role for IL-1 receptor signaling in modulating binge-like ethanol consumption and indicate that proinflammatory cytokines can be induced prior to dependence or any evidence of neuronal cell death. These findings provide a framework in which to understand how neuroimmune adaptations may alter ethanol consumption and therein contribute to alcohol abuse.


Asunto(s)
Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Etanol/administración & dosificación , Interleucina-1beta/metabolismo , Receptores de Interleucina-1/metabolismo , Animales , Encefalitis/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Receptores de Interleucina-1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
4.
Addict Biol ; 21(4): 835-46, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-25975524

RESUMEN

Binge ethanol drinking is a highly pervasive and destructive behavior yet the underlying neurobiological mechanisms remain poorly understood. Recent work suggests that overlapping neurobiological mechanisms modulate feeding disorders and excessive ethanol intake, and converging evidence indicates that the melanocortin (MC) system may be a promising candidate. The aims of the present work were to examine how repeated binge-like ethanol drinking, using the 'drinking in the dark' (DID) protocol, impacts key peptides within the MC system and if site-specific manipulation of MC receptor (MCR) signaling modulates binge-like ethanol drinking. Male C57BL/6J mice were exposed to one, three or six cycles of binge-like ethanol, sucrose or water drinking, after which brain tissue was processed via immunohistochemistry (IHC) for analysis of key MC peptides, including alpha-melanocyte stimulating hormone (α-MSH) and agouti-related protein (AgRP). Results indicated that α-MSH expression was selectively decreased, while AgRP expression was selectively increased, within specific hypothalamic subregions following repeated binge-like ethanol drinking. To further explore this relationship, we used site-directed drug delivery techniques to agonize or antagonize MCRs within the lateral hypothalamus (LH). We found that the nonselective MCR agonist melanotan-II (MTII) blunted, while the nonselective MCR antagonist AgRP augmented, binge-like ethanol consumption when delivered into the LH. As these effects were region-specific, the present results suggest that a more thorough understanding of the MC neurocircuitry within the hypothalamus will help provide novel insight into the mechanisms that modulate excessive binge-like ethanol intake and may help uncover new therapeutic targets aimed at treating alcohol abuse disorders.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Hipotálamo/efectos de los fármacos , Receptores de Melanocortina/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
5.
Alcohol Clin Exp Res ; 39(8): 1406-16, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26110576

RESUMEN

BACKGROUND: In recent years, much attention has been given to the lack of reproducibility in biomedical research, particularly in preclinical animal studies. This is a problem that also plagues the alcohol research field, particularly in consistent consumption in animal models of alcohol use disorders. One often overlooked factor that could affect reproducibility is the maintenance diet used in preclinical studies. METHODS: Herein, 2 well-established models of alcohol consumption, the "drinking in the dark" (DID) procedure and the continuous 2-bottle choice (C2BC) paradigm, were employed to determine the effects of diet on ethanol (EtOH) consumption. Male C57BL/6J mice were given 1 of 6 standard rodent chow diets obtained from Purina LabDiet(®) , Inc. (Prolab(®) RMH 3000) or Harlan(®) Laboratories, Inc. (Teklad Diets T.2916, T.2918, T.2920X, T.7912, or T.8940). A separate group of animals were used to test dietary effects on EtOH pharmacokinetics and behavioral measures following intraperitoneal (IP) injections of various doses of EtOH. RESULTS: Mice eating Harlan diets T.2916 (H2916) and T.2920X (H2920) consumed significantly less EtOH and exhibited lower blood EtOH concentrations (BECs) during DID; however, during C2BC, animals maintained on Harlan T.7912 (H7912) consumed more EtOH and had a higher EtOH preference than the other diet groups. EtOH consumption levels did not stem from changes in alcohol pharmacokinetics, as a separate group of animals administered EtOH IP showed no difference in BECs. However, animals on Harlan diet T.2920X (H2920) were more sensitive to alcohol-induced locomotor activity in an open-field task. No diet-dependent differences were seen in alcohol-induced sedation as measured with loss of righting reflex. CONCLUSIONS: Although these data do not identify a specific mechanism, together, they clearly show that the maintenance diet impacts EtOH consumption. It is incumbent upon the research community to consider the importance of describing nutritional information in methods, which may help decrease interlaboratory reproducibility issues.


Asunto(s)
Consumo de Bebidas Alcohólicas , Alimentación Animal , Consumo Excesivo de Bebidas Alcohólicas , Conducta de Elección , Dieta , Etanol/administración & dosificación , Consumo de Bebidas Alcohólicas/fisiopatología , Alimentación Animal/normas , Animales , Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Dieta/normas , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Alcohol Clin Exp Res ; 38(9): 2377-86, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25130590

RESUMEN

BACKGROUND: A growing body of literature suggests that epigenetic mechanisms, including histone acetylation, may play key roles in drug abuse and the development of addiction. Experiments in this study were designed to investigate the role of histone acetylation in ethanol (EtOH)-induced locomotor sensitization. METHODS: Immunohistochemical, Western blotting, and site-directed pharmacological techniques were used to explore the roles of histone acetylation at histone H3 (acH3K9) in both the expression of and acquisition of EtOH-induced locomotor sensitization. A commonly used sensitization protocol, in which animals were exposed to repeated injections of a low dose of EtOH while in their home cage, was used to examine this behavioral phenomenon. Additionally, site-directed administration of the histone deacetylase inhibitor (HDACi) Trichostatin A (TSA), in the absence of repeated EtOH injections, was used to examine the role of hyperacetylation in the nucleus accumbens (NAC) shell in EtOH-induced locomotor sensitization. RESULTS: Sensitized mice displayed elevated acH3K9 immunoreactivity (IR) localized to the shell of the NAC. This augmentation in acH3K9 IR was confirmed, in a separate experiment, using Western blot analyses. Next, repeated intra-accumbal infusions of TSA, in the absence of repeated EtOH injections, were sufficient to induce an augmented locomotor response to a later injection of a low dose (2.0 g/kg, intraperitoneally) of EtOH, indicative of cross-sensitization to this locomotor stimulation between TSA and EtOH. Finally, a local infusion of TSA into the shell of the accumbens was also associated with a significant increase in acH3K9 IR within this region. CONCLUSIONS: Together, the present observations suggest that histone acetylation, particularly within the shell of the NAC, is important for the development and expression of EtOH-induced locomotor sensitization.


Asunto(s)
Etanol/farmacología , Histonas/metabolismo , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Acetilación/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Endogámicos DBA
7.
Neuron ; 112(5): 772-785.e9, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38141605

RESUMEN

Lack of behavioral suppression typifies substance use disorders, yet the neural circuit underpinnings of drug-induced behavioral disinhibition remain unclear. Here, we employ deep-brain two-photon calcium imaging in heroin self-administering mice, longitudinally tracking adaptations within a paraventricular thalamus to nucleus accumbens behavioral inhibition circuit from the onset of heroin use to reinstatement. We find that select thalamo-accumbal neuronal ensembles become profoundly hypoactive across the development of heroin seeking and use. Electrophysiological experiments further reveal persistent adaptations at thalamo-accumbal parvalbumin interneuronal synapses, whereas functional rescue of these synapses prevents multiple triggers from initiating reinstatement of heroin seeking. Finally, we find an enrichment of µ-opioid receptors in output- and cell-type-specific paraventricular thalamic neurons, which provide a mechanism for heroin-induced synaptic plasticity and behavioral disinhibition. These findings reveal key circuit adaptations that underlie behavioral disinhibition in opioid dependence and further suggest that recovery of this system would reduce relapse susceptibility.


Asunto(s)
Heroína , Trastornos Relacionados con Opioides , Ratas , Ratones , Animales , Heroína/farmacología , Ratas Sprague-Dawley , Autoadministración/métodos , Neuronas , Núcleo Accumbens/fisiología
8.
J Neurosci ; 32(10): 3405-13, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22399763

RESUMEN

A well established body of work indicates a crucial role for corticotropin-releasing factor (CRF) in neurobiological responses associated with excessive dependence-like ethanol drinking in ethanol-vapor-exposed rodents. Recent evidence demonstrates a role for CRF in the modulation of binge-like ethanol consumption by nondependent mice, a behavior that can precede ethanol dependence. The CRF circuitry that is engaged by binge-like ethanol exposure, however, is unknown. Using converging approaches, we provide evidence that, similar to ethanol-vapor-induced increases in ethanol intake, CRF signaling in the central nucleus of the amygdala (CeA) is engaged during binge-like ethanol consumption by C57BL/6J mice. Specifically, we found that binge-like consumption of an ethanol solution (20% ethanol v/v) was attenuated by pretreatment with the CRF1R antagonists antalarmin, 4-ethyl-[2,5,6-trimethyl-7-(2,4,6-trimethylphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]amino-1-butanol, and NBI-27914 at doses (30 mg/kg, i.p.) that did not alter nonbinge-like ethanol consumption. Binge-like ethanol consumption resulted in significant increases of CRF immunoreactivity in the CeA immediately following ethanol drinking and 18-24 h following ethanol removal and also blocked the ability of CRF to enhance GABAergic transmission in the CeA 18-24 h following ethanol removal. Pretreatment with bilateral injections of antalarmin (1 µg/0.5 µl per side) into the CeA, but not the adjacent basolateral amygdala, significantly attenuated binge-like ethanol consumption. These findings suggest that CRF signaling in the CeA is recruited during excessive ethanol intake, before the development of dependence. We hypothesize that plastic changes in CRF signaling develop with repeated binge-like drinking episodes, contributing to the transition to dependence.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Hormona Liberadora de Corticotropina/fisiología , Etanol/envenenamiento , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Alcohol Clin Exp Res ; 37(10): 1688-95, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23647551

RESUMEN

BACKGROUND: Recently, procedures have been developed to model specific facets of human alcohol abuse disorders, including those that model excessive binge-like drinking (i.e., "drinking-in-the-dark," or DID procedures) and excessive dependence-like drinking (i.e., intermittent ethanol [EtOH] vapor exposure). Similar neuropeptide systems modulate excessive EtOH drinking stemming from both procedures, raising the possibility that both paradigms are actually modeling the same phenotypes and triggering the same central neuroplasticity. Therefore, the goal of this present project was to study the effects of a history of binge-like EtOH drinking, using DID procedures, on phenotypes that have previously been described with procedures to model dependence-like drinking. METHODS: Male C57BL/6J mice first experienced 0 to 10 four-day binge-like drinking episodes (3 days of rest between episodes). Beginning 24 hours after the final binge-like drinking session, mice were tested for anxiety-like behaviors (with elevated plus maze [EPM] and open-field locomotor activity tests), ataxia with the rotarod test, and sensitivity to handling-induced convulsions (HICs). One week later, mice began a 40-day 2-bottle (water vs. EtOH) voluntary consumption test with concentration ranging from 10 to 20% (v/v) EtOH. RESULTS: A prior history of binge-like EtOH drinking significantly increased subsequent voluntary EtOH consumption and preference, effects most robust in groups that initially experienced 6 or 10 binge-like drinking episodes and completely absent in mice that experienced 1 binge-like drinking episode. Conversely, a history of binge-like EtOH drinking did not influence anxiety-like behaviors, ataxia, or HICs. CONCLUSIONS: Excessive EtOH drinking stemming from DID procedures does not initially induce phenotypes consistent with a dependence-like state. However, the subsequent increases in voluntary EtOH consumption and preference that become more robust following repeated episodes of binge-like EtOH drinking may reflect the early stages of EtOH dependence, suggesting that DID procedures may be ideal for studying the transition to EtOH dependence.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/diagnóstico , Consumo Excesivo de Bebidas Alcohólicas/psicología , Oscuridad , Etanol/administración & dosificación , Fenotipo , Consumo de Bebidas Alcohólicas/psicología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
10.
Alcohol ; 100: 41-56, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35181404

RESUMEN

Alcohol use disorder (AUD) is frequently comorbid with mood disorders, and these co-occurring neuropsychiatric disorders contribute to the development and maintenance of alcohol dependence and relapse. In preclinical models, mice chronically exposed to alcohol display anxiety-like and depressive-like behaviors during acute withdrawal and protracted abstinence. However, in total, results from studies using voluntary alcohol-drinking paradigms show variable behavioral outcomes in assays measuring negative affective behaviors. Thus, the main objective of this review is to summarize the literature on the variability of negative affective behaviors in mice after chronic alcohol exposure. We compare the behavioral phenotypes that emerge during abstinence across different exposure models, including models of alcohol and stress interactions. The complicated outcomes from these studies highlight the difficulties of assessing negative affective behaviors in mouse models designed for the study of AUD. We discuss new behavioral assays, comprehensive platforms, and unbiased machine-learning algorithms as promising approaches to better understand the interaction between alcohol and negative affect in mice. New data-driven approaches in the understanding of mouse behavior hold promise for improving the identification of mechanisms, cell subtypes, and neurocircuits that mediate negative affect. In turn, improving our understanding of the neurobehavioral basis of alcohol-associated negative affect will provide a platform to test hypotheses in mouse models that aim to improve the development of more effective strategies for treating individuals with AUD and co-occurring mood disorders.


Asunto(s)
Consumo de Bebidas Alcohólicas , Alcoholismo , Afecto , Abstinencia de Alcohol , Consumo de Bebidas Alcohólicas/psicología , Alcoholismo/psicología , Animales , Ansiedad/psicología , Trastornos de Ansiedad/psicología , Etanol , Ratones
11.
Nat Commun ; 13(1): 6865, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36369508

RESUMEN

Suppression of dangerous or inappropriate reward-motivated behaviors is critical for survival, whereas therapeutic or recreational opioid use can unleash detrimental behavioral actions and addiction. Nevertheless, the neuronal systems that suppress maladaptive motivated behaviors remain unclear, and whether opioids disengage those systems is unknown. In a mouse model using two-photon calcium imaging in vivo, we identify paraventricular thalamostriatal neuronal ensembles that are inhibited upon sucrose self-administration and seeking, yet these neurons are tonically active when behavior is suppressed by a fear-provoking predator odor, a pharmacological stressor, or inhibitory learning. Electrophysiological, optogenetic, and chemogenetic experiments reveal that thalamostriatal neurons innervate accumbal parvalbumin interneurons through synapses enriched with calcium permeable AMPA receptors, and activity within this circuit is necessary and sufficient for the suppression of sucrose seeking regardless of the behavioral suppressor administered. Furthermore, systemic or intra-accumbal opioid injections rapidly dysregulate thalamostriatal ensemble dynamics, weaken thalamostriatal synaptic innervation of downstream neurons, and unleash reward-seeking behaviors in a manner that is reversed by genetic deletion of thalamic µ-opioid receptors. Overall, our findings reveal a thalamostriatal to parvalbumin interneuron circuit that is both required for the suppression of reward seeking and rapidly disengaged by opioids.


Asunto(s)
Analgésicos Opioides , Parvalbúminas , Ratones , Animales , Analgésicos Opioides/farmacología , Calcio , Recompensa , Sacarosa
12.
Neuropharmacology ; 182: 108396, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33181147

RESUMEN

Stress is a risk factor that plays a considerable role in the development and maintenance of alcohol (ethanol) abuse and relapse. Preclinical studies examining ethanol-stress interactions have demonstrated elevated ethanol drinking, cognitive deficits, and negative affective behaviors in mice. However, the neural adaptations in prefrontal cortical regions that drive these aberrant behaviors produced by ethanol-stress interactions are unknown. In this study, male C57BL/6J mice were exposed to chronic intermittent ethanol (CIE) and repeated forced swim stress (FSS). After two cycles of CIE x FSS, brain slices containing the prelimbic (PrL) and infralimbic (IfL) cortex were prepared for analysis of adaptations in dendritic spines and synaptic plasticity. In the PrL cortex, total spine density was increased in mice exposed to CIE. Immediately following induction of long-term potentiation (LTP), the fEPSP slope was increased in the PrL of CIE x FSS treated mice, indicative of a presynaptic adaptation on post-tetanic potentiation (PTP). In the IfL cortex, CIE exposure regardless of FSS experience resulted in an increase in spine density. FSS alone or when combined with CIE exposure increased PTP following LTP induction. Repeated FSS episodes increased IfL cortical paired-pulse facilitation, a second measure of presynaptic plasticity. In summary, CIE exposure resulted in structural adaptations while repeated stress exposure drove metaplastic changes in presynaptic function, demonstrating distinct morphological and functional changes in PrL and IfL cortical neurons. Thus, the structural and functional adaptations may be one mechanism underlying the development of excessive drinking and cognitive deficits associated with ethanol-stress interactions.


Asunto(s)
Etanol/administración & dosificación , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Estrés Psicológico/patología , Administración por Inhalación , Animales , Etanol/toxicidad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Estrés Psicológico/psicología
13.
Nat Commun ; 12(1): 5080, 2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34426574

RESUMEN

Bed nucleus of the stria terminalis (BNST) neurons that synthesize corticotropin-releasing factor (CRF) drive binge alcohol drinking and anxiety. Here, we found that female C57BL/6J mice binge drink more than males and have greater basal BNSTCRF neuron excitability and synaptic excitation. We identified a dense VGLUT2 + synaptic input from the paraventricular thalamus (PVT) that releases glutamate directly onto BNSTCRF neurons but also engages a large BNST interneuron population to ultimately inhibit BNSTCRF neurons, and this polysynaptic PVTVGLUT2-BNSTCRF circuit is more robust in females than males. Chemogenetic inhibition of the PVTBNST projection promoted binge alcohol drinking only in female mice, while activation reduced avoidance behavior in both sexes. Lastly, repeated binge drinking produced a female-like phenotype in the male PVT-BNSTCRF excitatory synapse without altering the function of PVTBNST neurons per se. Our data describe a complex, feedforward inhibitory PVTVGLUT2-BNSTCRF circuit that is sex-dependent in its function, behavioral roles, and alcohol-induced plasticity.


Asunto(s)
Consumo de Bebidas Alcohólicas/patología , Reacción de Prevención , Hormona Liberadora de Corticotropina/metabolismo , Sistema Límbico/patología , Neuronas/patología , Sinapsis/patología , Tálamo/patología , Consumo de Bebidas Alcohólicas/fisiopatología , Animales , Ansiedad/fisiopatología , Conducta Animal , Potenciales Postsinápticos Excitadores , Femenino , Ácido Glutámico/metabolismo , Potenciales Postsinápticos Inhibidores , Integrasas/metabolismo , Sistema Límbico/fisiopatología , Masculino , Ratones Endogámicos C57BL , Fenotipo , Núcleos Septales/patología , Núcleos Septales/fisiopatología , Caracteres Sexuales , Tálamo/fisiopatología
14.
Alcohol ; 88: 91-99, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32777473

RESUMEN

Alcohol use disorder is highly comorbid with other neuropsychiatric disorders such as depression and anxiety. Importantly, women and men are affected differentially by heavy drinking, with women experiencing longer negative affective states after intoxication and increased likelihood to present with comorbid mood or anxiety disorders. In rodents, several studies using different alcohol administration models have shown the development of depressive-like or anxiety-like phenotypes that emerge during abstinence. In this study, we compared the emergence of negative affective behaviors during abstinence from 7 weeks of two-bottle choice intermittent access to 20% alcohol in male and female C57BL/6J mice, a drinking paradigm little studied in this context. Half of the mice were tested 24 hours into abstinence on the elevated zero maze and 19-20 days into abstinence in a novel object in the home cage encounter test. The other half of the mice were tested 27-28 days into abstinence with the novelty-suppressed feeding test. As expected, females drank more than males across the 7 weeks of access to alcohol. Drinking history did not affect performance on these tasks, with the exception of increasing the number of open arm entries on the elevated zero maze. Interestingly, in alcohol-naïve mice, females showed fewer anxiety-like behaviors than males in the elevated zero maze and the novelty-suppressed feeding test. Our results suggest that the intermittent access model does not reliably induce negative affective behaviors on these tasks, and that behavior in female and male mice differs across these tests. Rather, intermittent alcohol drinking may induce a mild form of behavioral disinhibition. Thus, the model of alcohol access is a critical factor in determining the appearance of behavioral disturbances that emerge during abstinence.


Asunto(s)
Alcoholismo , Ansiedad , Etanol/efectos adversos , Trastornos del Humor/inducido químicamente , Consumo de Bebidas Alcohólicas , Alcoholismo/complicaciones , Animales , Ansiedad/inducido químicamente , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Transl Psychiatry ; 10(1): 414, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33247097

RESUMEN

Mood disorders are often comorbid with alcohol use disorder (AUD) and play a considerable role in the development and maintenance of alcohol dependence and relapse. Because of this high comorbidity, it is necessary to determine shared and unique genetic factors driving heavy drinking and negative affective behaviors. In order to identify novel pharmacogenetic targets, a bioinformatics analysis was used to quantify the expression of amygdala K+ channel genes that covary with anxiety-related phenotypes in the well-phenotyped and fully sequenced family of BXD strains. We used a model of stress-induced escalation of drinking in alcohol-dependent mice to measure negative affective behaviors during abstinence. A pharmacological approach was used to validate the key bioinformatics findings in alcohol-dependent, stressed mice. Amygdalar expression of Kcnn3 correlated significantly with 40 anxiety-associated phenotypes. Further examination of Kcnn3 expression revealed a strong eigentrait for anxiety-like behaviors and negative correlations with binge-like and voluntary alcohol drinking. Mice treated with chronic intermittent alcohol exposure and repeated swim stress consumed more alcohol in their home cages and showed hypophagia on the novelty-suppressed feeding test during abstinence. Pharmacologically targeting Kcnn gene products with the KCa2 (SK) channel-positive modulator 1-EBIO decreased drinking and reduced feeding latency in alcohol-dependent, stressed mice. Collectively, these validation studies provide central nervous system links into the covariance of stress, negative affective behaviors, and AUD in the BXD strains. Further, the bioinformatics discovery tool is effective in identifying promising targets (i.e., KCa2 channels) for treating alcohol dependence exacerbated by comorbid mood disorders.


Asunto(s)
Alcoholismo , Conducta Animal , Biología Computacional , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Afecto , Consumo de Bebidas Alcohólicas , Animales , Ansiedad , Etanol , Ratones
16.
Psychopharmacology (Berl) ; 197(3): 409-19, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18204997

RESUMEN

RATIONALE: Overall drug acceptability is thought to be a function of the balance between its rewarding and aversive effects, the latter of which is reportedly affected by polydrug use. OBJECTIVES: Given that nicotine and alcohol are commonly co-used, the present experiments sought to assess nicotine's impact on ethanol's aversive effects within a conditioned taste aversion design. MATERIALS AND METHODS: Experiment 1 examined various doses of nicotine (0, 0.4, 0.8, 1.2 mg/kg) to determine a behaviorally active dose, and experiment 2 examined various doses of ethanol (0, 0.5, 1.0, 1.5 g/kg) to determine a dose that produced intermediate aversions. Experiment 3 then examined the aversive effects of nicotine (0.8 mg/kg) and ethanol (1.0 g/kg) alone and in combination. Additionally, nicotine's effects on blood alcohol concentrations (BAC) and ethanol-induced hypothermia were examined. RESULTS: Nicotine and ethanol combined produced aversions significantly greater than those produced by either drug alone or the summed aversive effects of the individual compounds. These effects were unrelated to changes in BAC, but nicotine and ethanol combined produced a prolonged hypothermic effect which may contribute to the increased aversions induced by the combination. CONCLUSIONS: These data demonstrate that nicotine may interact with ethanol, increasing ethanol's aversive effects. Although the rewarding effects of concurrently administered nicotine and ethanol were not assessed, these data do indicate that the reported high incidence of nicotine and ethanol co-use is unlikely due to reductions in the aversiveness of ethanol with concurrently administered nicotine. It is more likely attributable to nicotine-related changes in ethanol's rewarding effects.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Etanol/toxicidad , Nicotina/farmacología , Gusto/efectos de los fármacos , Animales , Aprendizaje por Asociación/efectos de los fármacos , Regulación de la Temperatura Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Etanol/sangre , Femenino , Inyecciones Intravenosas , Nicotina/toxicidad , Ratas , Ratas Long-Evans
17.
Pharmacol Biochem Behav ; 88(4): 427-31, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17945339

RESUMEN

Sex differences in taste aversion learning have been reported for a number of different compounds. It is unknown, however, to what degree, if any, such differences exist when nicotine is the aversion-inducing agent. To address this issue, in the present experiment male and female rats were given limited access to saccharin followed by an intraperitoneal (i.p.) injection of either vehicle or nicotine (0.4, 0.8 or 1.2 mg/kg). Although nicotine induced significant taste aversions in both males and females, the aversions were generally weak at all doses tested. There were no sex differences in the acquisition or strength of the aversions induced by nicotine. The vulnerability to drug abuse has been suggested to be a function of the balance of the rewarding and aversive effects of a drug. Given the relatively weak aversions induced in both sexes and the absence of differences between males and females, it is unlikely that the reported sex difference in the self-administration of nicotine is a function of differences in nicotine's aversive effects. The reported difference in the self-administration of nicotine by males and females is more likely a function of differences in the sensitivity to the rewarding effects of the drug.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Gusto/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratas , Ratas Long-Evans , Sacarina/farmacología , Autoadministración , Caracteres Sexuales , Edulcorantes/farmacología
18.
Pharmacol Biochem Behav ; 91(1): 134-9, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18639579

RESUMEN

Genetic and early environmental factors interact to influence ethanol's motivational effects. To explore these issues, a reciprocal cross-fostering paradigm was applied to Fischer and Lewis rats. The adult female offspring received vehicle or the kappa opioid antagonist nor-BNI (1 mg/kg) followed by assessments of conditioned taste aversion (CTA), blood alcohol concentrations (BACs) and hypothermia induced by 1.25 g/kg intraperitoneal ethanol. CTA acquisition in the in-fostered Fischer and Lewis animals did not differ; however, the Fischer maternal environment produced stronger acquisition in the cross-fostered Lewis rats versus their in-fostered counterparts. CTAs in the Fischer rats were not affected by cross-fostering. In extinction, the in-fostered Lewis animals displayed stronger aversions than the Fischer groups on two trials (of 12) whereas the cross-fostered Lewis differed from the Fischer groups on nine trials. Despite these CTA effects, Lewis rats exhibited higher BACs and stronger hypothermic responses than Fischer with no cross-fostering effects in either strain. No phenotypes were affected by nor-BNI. These data extend previous findings dissociating the aversive and peripheral physiological effects of ethanol in female Fischer and Lewis rats, and highlight the importance of genetic and early environmental factors in shaping subsequent responses to alcohol's motivational effects in adulthood.


Asunto(s)
Depresores del Sistema Nervioso Central/antagonistas & inhibidores , Depresores del Sistema Nervioso Central/farmacología , Etanol/antagonistas & inhibidores , Etanol/farmacología , Motivación , Animales , Reacción de Prevención/efectos de los fármacos , Depresores del Sistema Nervioso Central/sangre , Condicionamiento Operante/efectos de los fármacos , Interpretación Estadística de Datos , Dinorfinas/administración & dosificación , Dinorfinas/farmacología , Etanol/sangre , Extinción Psicológica/efectos de los fármacos , Femenino , Hipotermia/inducido químicamente , Hipotermia/fisiopatología , Conducta Materna/efectos de los fármacos , Naltrexona/administración & dosificación , Naltrexona/análogos & derivados , Naltrexona/farmacología , Ratas , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides kappa/genética , Gusto/efectos de los fármacos
19.
Neuropharmacology ; 138: 10-19, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29775679

RESUMEN

Currently available pharmacotherapies for treating alcohol use disorder (AUD) suffer from deleterious side effects and are not efficacious in diverse populations. Clinical and preclinical studies provide evidence that the Kcnq family of genes that encode KV7 channels influence alcohol intake and dependence. KV7 channels are a class of slowly activating voltage-dependent K+ channels that regulate neuronal excitability. Studies indicate that the KV7 channel positive modulator retigabine can decrease dopaminergic neuron firing, alter dopamine (DA) release, and reduce alcohol intake in heavy drinking rodents. Given the critical nature of ventral tegmental area (VTA) DA to the addiction process and predominant expression of Kcnq4 in DA neurons, we investigated the role of midbrain Kcnq genes and KV7 channels in the VTA of genetically diverse mice and long-term heavy drinking rats, respectively. Integrative bioinformatics analysis identified negative correlations between midbrain Kcnq4 expression and alcohol intake and seeking behaviors. Kcnq4 expression levels were also correlated with dopaminergic-related phenotypes in BXD strains, and Kcnq4 was present in support intervals for alcohol sensitivity and alcohol withdrawal severity QTLs in rodents. Pharmacological validation studies revealed that VTA KV7 channels regulate excessive alcohol intake in rats with a high-drinking phenotype. Administration of a novel and selective KV7.2/4 channel positive modulator also reduced alcohol drinking in rats. Together, these findings indicate that midbrain Kcnq4 expression regulates alcohol-related behaviors in genetically diverse mice and provide evidence that KV7.4 channels are a critical mediator of excessive alcohol drinking.


Asunto(s)
Trastornos Relacionados con Alcohol/metabolismo , Canales de Potasio KCNQ/metabolismo , Área Tegmental Ventral/metabolismo , Disuasivos de Alcohol/farmacología , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/metabolismo , Trastornos Relacionados con Alcohol/tratamiento farmacológico , Animales , Dopamina/metabolismo , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/fisiología , Femenino , Predisposición Genética a la Enfermedad , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Ratas Wistar , Área Tegmental Ventral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA