Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Learn Mem ; 30(1): 1-11, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36543385

RESUMEN

Posttraumatic stress disorder (PTSD) is associated with neural and behavioral alterations in response to trauma exposure, including working memory impairments. Rodent models of PTSD have not fully investigated chronic or reactive working memory deficits, despite clinical relevance. The present study uses footshock to induce a posttraumatic stress state in male and female rats and evaluates the effect of footshock and trauma-paired odor cues on working memory performance in the odor span task. Results demonstrate the emergence of chronic deficits in working memory among animals exposed to footshock by 3 wk after traumatic stress. The presentation of a trauma-paired odor cue was associated with further decrement in working memory performance for male animals. Furthermore, anxiety-like behaviors associated with the PTSD-like phenotype could predict the degree of working memory impairment in response to the trauma-paired odor cue. This study enhances validation of an existing rodent model of PTSD through replication of the clinical observations of working memory deficits associated with PTSD and provides novel insight into effects in female rodents. This will facilitate work to probe underlying mechanistic dysregulation of working memory following footshock trauma exposure and future development of novel treatment strategies.


Asunto(s)
Trastornos por Estrés Postraumático , Ratas , Masculino , Femenino , Animales , Odorantes , Ansiedad , Memoria a Corto Plazo/fisiología , Trastornos de la Memoria
2.
Alcohol Clin Exp Res ; 45(2): 480-489, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33351976

RESUMEN

BACKGROUND: Stress triggers alcohol use and relapse to drinking, with different effects by sex. Women are more susceptible to stress-related alcohol misuse, and most stressors in rodents produce sexually divergent effects. Female rodents are particularly sensitive to the stress produced by solitary housing, yet the impact of housing conditions on the establishment, escalation, and post-abstinence potentiation of intermittent access alcohol drinking in male and female rats, and the interaction of these factors with stress history are not well described. METHODS: Male (n = 62) and female (n = 64) Wistar rats were housed individually or in pairs separated by a perforated divider. Rats were exposed to light-cued footshock stress (stress history), or cues alone (control), once daily for 3 days, followed by 8 weeks' drinking under intermittent access to a 2-bottle choice (IA2BC), with 20% alcohol (v/v in water) available in addition to water for 24 hours on alternate days. After a 2-week forced abstinence, anxiety-like behavior was assessed via defensive withdrawal testing; then, IA2BC alcohol access was renewed for 2 weeks to model relapse-like behavior. RESULTS: Pair-housed female rats did not increase their alcohol intake across the 8-week drinking period, unlike all other groups, and stress history did not significantly change alcohol consumption. After abstinence, anxiety-like behavior was greatest in pair-housed stress history males, whereas alcohol intake was significantly elevated only in female rats, particularly those in solitary housing. CONCLUSIONS: Together, these findings suggest that paired housing differentially contributes to behavior in male and female rats, blunting alcohol intake in females, and unmasking stress history effects on anxiety-like behavior in males.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Ansiedad/psicología , Vivienda para Animales , Caracteres Sexuales , Aislamiento Social/psicología , Estrés Psicológico/psicología , Consumo de Bebidas Alcohólicas/efectos adversos , Animales , Femenino , Masculino , Ratas , Ratas Wistar , Recurrencia , Autoadministración
3.
J Neurosci ; 38(44): 9423-9432, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30381434

RESUMEN

Risk for stress-sensitive psychopathologies differs in men and women, yet little is known about sex-dependent effects of stress on cellular structure and function in corticolimbic regions implicated in these disorders. Determining how stress influences these regions in males and females will deepen our understanding of the mechanisms underlying sex-biased psychopathology. Here, we discuss sex differences in CRF regulation of arousal and cognition, glucocorticoid modulation of amygdalar physiology and alcohol consumption, the age-dependent impact of social stress on prefrontal pyramidal cell excitability, stress effects on the prefrontal parvalbumin system in relation to emotional behaviors, contributions of stress and gonadal hormones to stress effects on prefrontal glia, and alterations in corticolimbic structure and function after cessation of chronic stress. These studies demonstrate that, while sex differences in stress effects may be nuanced, nonuniform, and nonlinear, investigations of these differences are nonetheless critical for developing effective, sex-specific treatments for psychological disorders.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Emociones/fisiología , Motivación/fisiología , Resiliencia Psicológica , Caracteres Sexuales , Estrés Psicológico/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Humanos , Masculino , Trastornos Mentales/metabolismo , Trastornos Mentales/patología , Trastornos Mentales/psicología , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Factores de Riesgo , Estrés Psicológico/patología , Estrés Psicológico/psicología
4.
Addict Biol ; 24(3): 335-343, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-29726054

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) has been extensively studied for its role in the development and maintenance of the midbrain dopaminergic system, although evidence suggests that GDNF also plays a role in drug and alcohol addiction. This review focuses on the unique actions of GDNF in the mechanisms that prevent the transition from recreational alcohol use to abuse. Specifically, we describe studies in rodents suggesting that alcohol acutely increases GDNF expression in the ventral tegmental area, which enables the activation of the mitogen-activated protein kinase signaling pathway and the gating of alcohol intake. We further provide evidence to suggest that GDNF acts in the ventral tegmental area via both nongenomic and genomic mechanisms to suppress alcohol consumption. In addition, we describe findings indicating that when this endogenous protective pathway becomes dysregulated, alcohol intake levels escalate. Finally, we describe the potential use of GDNF inducers as a novel therapeutic approach to treat alcohol use disorder.


Asunto(s)
Alcoholismo/etiología , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Alcoholismo/fisiopatología , Depresores del Sistema Nervioso Central/farmacología , Neuronas Dopaminérgicas/fisiología , Etanol/farmacología , Humanos , Sistema Límbico/patología , Trastornos Mentales/etiología , Trastornos Mentales/fisiopatología , Núcleo Accumbens/fisiología , Transducción de Señal/fisiología , Tegmento Mesencefálico/fisiología
5.
J Neurosci ; 37(17): 4593-4603, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28363981

RESUMEN

L-type voltage-gated calcium channels (LTCCs) are implicated in several psychiatric disorders that are comorbid with alcoholism and involve amygdala dysfunction. Within the amygdala, the central nucleus (CeA) is critical in acute alcohol's reinforcing actions, and its dysregulation in human alcoholics drives their negative emotional state and motivation to drink. Here we investigated the specific role of CeA LTCCs in the effects of acute alcohol at the molecular, cellular physiology, and behavioral levels, and their potential neuroadaptation in alcohol-dependent rats. Alcohol increases CeA activity (neuronal firing rates and GABA release) in naive rats by engaging LTCCs, and intra-CeA LTCC blockade reduces alcohol intake in nondependent rats. Alcohol dependence reduces CeA LTCC membrane abundance and disrupts this LTCC-based mechanism; instead, corticotropin-releasing factor type 1 receptors (CRF1s) mediate alcohol's effects on CeA activity and drive the escalated alcohol intake of alcohol-dependent rats. Collectively, our data indicate that alcohol dependence functionally alters the molecular mechanisms underlying the CeA's response to alcohol (from LTCC- to CRF1-driven). This mechanistic switch contributes to and reflects the prominent role of the CeA in the negative emotional state that drives excessive drinking.SIGNIFICANCE STATEMENT The central amygdala (CeA) plays a critical role in the development of alcohol dependence. As a result, much preclinical alcohol research aims to identify relevant CeA neuroadaptions that promote the transition to dependence. Here we report that acute alcohol increases CeA neuronal activity in naive rats by engaging L-type calcium channels (LTCCs) and that intra-CeA LTCC blockade reduces alcohol intake in nondependent rats. Alcohol dependence disrupts this LTCC-based mechanism; instead, corticotropin-releasing factor type 1 receptors (CRF1s) mediate alcohol's effects on CeA activity and drive the escalated alcohol intake of alcohol-dependent rats. This switch reflects the important role of the CeA in the pathophysiology of alcohol dependence and represents a new potential avenue for therapeutic intervention during the transition period.


Asunto(s)
Alcoholismo/metabolismo , Canales de Calcio Tipo L/metabolismo , Núcleo Amigdalino Central/metabolismo , Consumo de Bebidas Alcohólicas/psicología , Alcoholismo/fisiopatología , Alcoholismo/psicología , Animales , Conducta Animal , Núcleo Amigdalino Central/fisiopatología , Depresores del Sistema Nervioso Central/farmacología , Emociones , Etanol/farmacología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/metabolismo
6.
J Neurosci ; 37(5): 1139-1155, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27986929

RESUMEN

Toll-like receptor 4 (TLR4) is a critical component of innate immune signaling and has been implicated in alcohol responses in preclinical and clinical models. Members of the Integrative Neuroscience Initiative on Alcoholism (INIA-Neuroimmune) consortium tested the hypothesis that TLR4 mediates excessive ethanol drinking using the following models: (1) Tlr4 knock-out (KO) rats, (2) selective knockdown of Tlr4 mRNA in mouse nucleus accumbens (NAc), and (3) injection of the TLR4 antagonist (+)-naloxone in mice. Lipopolysaccharide (LPS) decreased food/water intake and body weight in ethanol-naive and ethanol-trained wild-type (WT), but not Tlr4 KO rats. There were no consistent genotypic differences in two-bottle choice chronic ethanol intake or operant self-administration in rats before or after dependence. In mice, (+)-naloxone did not decrease drinking-in-the-dark and only modestly inhibited dependence-driven consumption at the highest dose. Tlr4 knockdown in mouse NAc did not decrease drinking in the two-bottle choice continuous or intermittent access tests. However, the latency to ethanol-induced loss of righting reflex increased and the duration decreased in KO versus WT rats. In rat central amygdala neurons, deletion of Tlr4 altered GABAA receptor function, but not GABA release. Although there were no genotype differences in acute ethanol effects before or after chronic intermittent ethanol exposure, genotype differences were observed after LPS exposure. Using different species and sexes, different methods to inhibit TLR4 signaling, and different ethanol consumption tests, our comprehensive studies indicate that TLR4 may play a role in ethanol-induced sedation and GABAA receptor function, but does not regulate excessive drinking directly and would not be an effective therapeutic target. SIGNIFICANCE STATEMENT: Toll-like receptor 4 (TLR4) is a key mediator of innate immune signaling and has been implicated in alcohol responses in animal models and human alcoholics. Members of the Integrative Neuroscience Initiative on Alcoholism (INIA-Neuroimmune) consortium participated in the first comprehensive study across multiple laboratories to test the hypothesis that TLR4 regulates excessive alcohol consumption in different species and different models of chronic, dependence-driven, and binge-like drinking. Although TLR4 was not a critical determinant of excessive drinking, it was important in the acute sedative effects of alcohol. Current research efforts are directed at determining which neuroimmune pathways mediate excessive alcohol drinking and these findings will help to prioritize relevant pathways and potential therapeutic targets.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/psicología , Alcoholismo/genética , Alcoholismo/psicología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/fisiología , Animales , Peso Corporal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Femenino , Técnicas de Inactivación de Genes , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Noqueados , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/metabolismo , Ratas , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/genética , Receptor Toll-Like 4/antagonistas & inhibidores
7.
J Neurosci ; 35(15): 6241-53, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878294

RESUMEN

Alcohol and nicotine are the two most co-abused drugs in the world. Previous studies have shown that nicotine can increase alcohol drinking in nondependent rats, yet it is unknown whether nicotine facilitates the transition to alcohol dependence. We tested the hypothesis that chronic nicotine will speed up the escalation of alcohol drinking in rats and that this effect will be accompanied by activation of sparsely distributed neurons (neuronal ensembles) throughout the brain that are specifically recruited by the combination of nicotine and alcohol. Rats were trained to respond for alcohol and made dependent using chronic, intermittent exposure to alcohol vapor, while receiving daily nicotine (0.8 mg/kg) injections. Identification of neuronal ensembles was performed after the last operant session, using immunohistochemistry. Nicotine produced an early escalation of alcohol drinking associated with compulsive alcohol drinking in dependent, but not in nondependent rats (air exposed), as measured by increased progressive-ratio responding and increased responding despite adverse consequences. The combination of nicotine and alcohol produced the recruitment of discrete and phenotype-specific neuronal ensembles (∼4-13% of total neuronal population) in the nucleus accumbens core, dorsomedial prefrontal cortex, central nucleus of the amygdala, bed nucleus of stria terminalis, and posterior ventral tegmental area. Blockade of nicotinic receptors using mecamylamine (1 mg/kg) prevented both the behavioral and neuronal effects of nicotine in dependent rats. These results demonstrate that nicotine and activation of nicotinic receptors are critical factors in the development of alcohol dependence through the dysregulation of a set of interconnected neuronal ensembles throughout the brain.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Encéfalo/metabolismo , Conducta Compulsiva/complicaciones , Nicotina/efectos adversos , Agonistas Nicotínicos/efectos adversos , Recompensa , Animales , Encéfalo/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Depresores del Sistema Nervioso Central/administración & dosificación , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Glutamato Descarboxilasa/metabolismo , Masculino , Fosfopiruvato Hidratasa/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Quinina/administración & dosificación , Ratas , Ratas Wistar , Autoadministración , Factores de Tiempo
8.
Front Neuroendocrinol ; 35(2): 234-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24456850

RESUMEN

Drug addiction is a chronically relapsing disorder characterized by loss of control over intake and dysregulation of stress-related brain emotional systems. Since the discovery by Wylie Vale and his colleagues of corticotropin-releasing factor (CRF) and the structurally-related urocortins, CRF systems have emerged as mediators of the body's response to stress. Relatedly, CRF systems have a prominent role in driving addiction via actions in the central extended amygdala, producing anxiety-like behavior, reward deficits, excessive, compulsive-like drug self-administration and stress-induced reinstatement of drug seeking. CRF neuron activation in the medial prefrontal cortex may also contribute to the loss of control. Polymorphisms in CRF system molecules are associated with drug use phenotypes in humans, often in interaction with stress history. Drug discovery efforts have yielded brain-penetrant CRF1 antagonists with activity in preclinical models of addiction. The results support the hypothesis that brain CRF-CRF1 systems contribute to the etiology and maintenance of addiction.


Asunto(s)
Conducta Adictiva/metabolismo , Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Estrés Psicológico/metabolismo , Trastornos Relacionados con Sustancias/etiología , Animales , Encéfalo/fisiopatología , Humanos , Estrés Fisiológico
9.
bioRxiv ; 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38260700

RESUMEN

A connection between stress-related illnesses and alcohol use disorders is extensively documented. Fear conditioning is a standard procedure used to study stress learning and links it to the activation of amygdala circuitry. However, the connection between the changes in amygdala circuit and function induced by alcohol and fear conditioning is not well established. We introduce a computational model to test the mechanistic relationship between amygdala functional and circuit adaptations during fear conditioning and the impact of acute vs. repeated alcohol exposure. In accordance with experiments, both acute and prior repeated alcohol decreases speed and robustness of fear extinction in our simulations. The model predicts that, first, the delay in fear extinction in alcohol is mostly induced by greater activation of the basolateral amygdala (BLA) after fear acquisition due to alcohol-induced modulation of synaptic weights. Second, both acute and prior repeated alcohol shifts the amygdala network away from the robust extinction regime by inhibiting the activity in the central amygdala (CeA). Third, our model predicts that fear memories formed in acute or after chronic alcohol are more connected to the context. Thus, the model suggests how circuit changes induced by alcohol may affect fear behaviors and provides a framework for investigating the involvement of multiple neuromodulators in this neuroadaptive process.

10.
J Neurosci ; 32(22): 7563-71, 2012 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-22649234

RESUMEN

Alcoholism is characterized by a compulsion to seek and ingest alcohol, loss of control over intake, and the emergence of a negative emotional state during abstinence. We hypothesized that sustained activation of neuroendocrine stress systems (e.g., corticosteroid release via the hypothalamic-pituitary-adrenal axis) by alcohol intoxication and withdrawal and consequent alterations in glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) activation drive compulsive alcohol drinking. Our results showed that rats exposed to alcohol vapor to the point of dependence displayed increased alcohol intake, compulsive drinking measured by progressive-ratio responding, and persistent alcohol consumption despite punishment, assessed by adding quinine to the alcohol solution, compared with control rats that were not exposed to alcohol vapor. No group differences were observed in the self-administration of saccharin-sweetened water. Acute alcohol withdrawal was accompanied by downregulated GR mRNA in various stress/reward-related brain regions [i.e., prefrontal cortex, nucleus accumbens (NAc), and bed nucleus of the stria terminalis (BNST)], whereas protracted alcohol abstinence was accompanied by upregulated GR mRNA in the NAc core, ventral BNST, and central nucleus of the amygdala. No significant alterations in MR mRNA levels were found. Chronic GR antagonism with mifepristone (RU38486) prevented the escalation of alcohol intake and compulsive responding induced by chronic, intermittent alcohol vapor exposure. Chronic treatment with mifepristone also blocked escalated alcohol drinking and compulsive responding during protracted abstinence. Thus, the GR system appears to be involved in the development of alcohol dependence and may represent a potential pharmacological target for the treatment of alcoholism.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Encéfalo/metabolismo , Receptores de Glucocorticoides/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Regulación hacia Arriba , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/patología , Análisis de Varianza , Animales , Conducta Adictiva/tratamiento farmacológico , Conducta Adictiva/metabolismo , Depresores del Sistema Nervioso Central/administración & dosificación , Conducta Compulsiva/fisiopatología , Condicionamiento Operante/efectos de los fármacos , Etanol/administración & dosificación , Antagonistas de Hormonas/uso terapéutico , Masculino , Mifepristona/uso terapéutico , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Esquema de Refuerzo , Autoadministración , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos
11.
Eur J Neurosci ; 37(4): 607-12, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23189980

RESUMEN

We previously found that the brain-derived neurotrophic factor (BDNF) in the dorsolateral striatum (DLS) is part of a homeostatic pathway that gates ethanol self-administration [Jeanblanc et al. (2009). J Neurosci, 29, 13494-13502)]. Specifically, we showed that moderate levels (10%) of ethanol consumption increase BDNF expression within the DLS, and that direct infusion of BDNF into the DLS decreases operant self-administration of a 10% ethanol solution. BDNF binding to its receptor, TrkB, activates the mitogen-activated protein kinase (MAPK), phospholipase C-γ (PLC-γ) and phosphatidylinositol 3-kinase (PI3K) pathways. Thus, here, we set out to identify which of these intracellular pathway(s) plays a role in the regulation of ethanol consumption by BDNF. We found that inhibition of the MAPK, but not PLC-γ or PI3K, activity blocks the BDNF-mediated reduction of ethanol consumption. As activation of the MAPK pathway leads to the initiation of transcription and/or translation events, we tested whether the BDNF-mediated reduction of ethanol self-administration requires de novo protein synthesis. We found that the inhibitory effect of BDNF on ethanol intake is blocked by the protein synthesis inhibitor cycloheximide. Together, our results show that BDNF attenuates ethanol drinking via activation of the MAPK pathway in a protein synthesis-dependent manner within the DLS.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Núcleo Talámico Mediodorsal/metabolismo , Animales , Depresores del Sistema Nervioso Central/farmacología , Condicionamiento Operante , Etanol/farmacología , Masculino , Núcleo Talámico Mediodorsal/efectos de los fármacos , Ratas , Ratas Long-Evans
12.
eNeuro ; 10(3)2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36808099

RESUMEN

Women are twice as likely as men to experience emotional dysregulation after stress, resulting in substantially higher psychopathology for equivalent lifetime stress exposure, yet the mechanisms underlying this vulnerability remain unknown. Studies suggest changes in medial prefrontal cortex (mPFC) activity as a potential contributor. Whether maladaptive changes in inhibitory interneurons participate in this process, and whether adaptations in response to stress differ between men and women, producing sex-specific changes in emotional behaviors and mPFC activity, remained undetermined. This study examined whether unpredictable chronic mild stress (UCMS) in mice differentially alters behavior and mPFC parvalbumin (PV) interneuron activity by sex, and whether the activity of these neurons drives sex-specific behavioral changes. Four weeks of UCMS increased anxiety-like and depressive-like behaviors associated with FosB activation in mPFC PV neurons, particularly in females. After 8 weeks of UCMS, both sexes displayed these behavioral and neural changes. Chemogenetic activation of PV neurons in UCMS-exposed and nonstressed males induced significant changes in anxiety-like behaviors. Importantly, patch-clamp electrophysiology demonstrated altered excitability and basic neural properties on the same timeline as the emergence of behavioral effects: changes in females after 4 weeks and in males after 8 weeks of UCMS. These findings show, for the first time, that sex-specific changes in the excitability of prefrontal PV neurons parallel the emergence of anxiety-like behavior, revealing a potential novel mechanism underlying the enhanced vulnerability of females to stress-induced psychopathology and supporting further investigation of this neuronal population to identify new therapeutic targets for stress disorders.


Asunto(s)
Ansiedad , Parvalbúminas , Masculino , Ratones , Femenino , Animales , Parvalbúminas/metabolismo , Ansiedad/patología , Neuronas/metabolismo , Trastornos de Ansiedad , Emociones , Interneuronas/fisiología , Corteza Prefrontal/metabolismo , Estrés Psicológico/patología
13.
Addict Biol ; 17(5): 920-33, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22741603

RESUMEN

Stressful experiences can result in elevated alcohol drinking, as exemplified in many individuals with post-traumatic stress disorder. However, how stress history, rather than acute stressors, influences alcohol intake remains uncertain. To model the protracted effects of past stress, male Wistar rats were subjected to light-cued footshock (stress history) or light cues alone (control) prior to acquisition of alcohol self-administration (1-hour sessions, fixed ratio 1-3, 100 µl of 10% v/v alcohol as reinforcer). Stress history did not alter mean alcohol intake during acquisition of self-administration, but it increased preference for the alcohol-paired lever over the inactive lever. Following an extinction period, rats with a history of stress exposure and low baseline alcohol intake showed a twofold elevation in alcohol self-administration, as compared with low-drinking rats with no stress history. Similar effects were not seen in rats self-administering 0.1% sucrose. Analysis of mRNA levels of phosphodiesterase 10A (PDE10A), a dual-specificity cyclic adenosine monophosphate and cyclic guanosine monophosphate hydrolyzing enzyme, showed that stress history increased Pde10a mRNA levels in the basolateral amygdala and, in low-drinking rats, the prelimbic prefrontal cortex (plPFC). Pde10a mRNA levels in the plPFC correlated directly with greater alcohol self-administration during the relapse-like phase, and greater BLA Pde10a mRNA levels correlated with increased ethanol preference after acquisition. The data demonstrate that stress history sensitizes otherwise low alcohol drinkers to consume more alcohol in a relapse-like situation and identify stress-induced neuroadaptations in amygdala and prefrontal cortical Pde10a expression as changes that may drive heightened alcohol intake and preference in susceptible individuals.


Asunto(s)
Alcoholismo/etiología , Hidrolasas Diéster Fosfóricas/metabolismo , Estrés Psicológico/psicología , Alcoholismo/enzimología , Amígdala del Cerebelo/enzimología , Análisis de Varianza , Animales , Depresores del Sistema Nervioso Central/farmacología , Condicionamiento Operante , Etanol/farmacología , Extinción Psicológica , Masculino , Estimulación Luminosa , Corteza Prefrontal/enzimología , Ratas , Ratas Wistar , Recurrencia , Estrés Psicológico/enzimología
14.
Neuropharmacology ; 190: 108564, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33857521

RESUMEN

Genetic predisposition to heavy drinking is a risk factor for alcohol misuse. We used selectively bred crossed high alcohol-preferring (cHAP) mice to study sex differences in alcohol drinking and its effect on glutamatergic activity in dorsolateral (DLS) and dorsomedial (DMS) striatum. We performed whole-cell patch-clamp recording in neurons from male and female cHAP mice with 5-week alcohol drinking history and alcohol-naïve controls. In DMS, alcohol-naïve males' neurons displayed lower cell capacitance and higher membrane resistance than females' neurons, both effects reversed by drinking. Conversely, in DLS neurons, drinking history increased capacitance only in males and changed membrane resistance only in females. Altered biophysical membrane properties were accompanied by disrupted glutamatergic transmission. Drinking history increased spontaneous excitatory postsynaptic current (sEPSC) amplitude in DMS and frequency in DLS female neurons, compared to alcohol-naïve females, without effect in males. Acute ethanol differentially impacted DMS and DLS neurons by sex and drinking history. In DMS, acute alcohol significantly increased sEPSC frequency only in neurons from alcohol-naïve females, an effect that disappeared after drinking history. In DLS, acute alcohol had opposing effects in males and females based on drinking history. Estrous cycle also impacted DMS and DLS neurons differently: sEPSC amplitudes were higher in DMS cells from drinking history than alcohol-naïve females, whereas estrous cycle, not drinking history, modified DLS firing rate. Our data show sex differences in cHAP ethanol consumption and neurophysiology, suggesting differential dysregulation of glutamatergic drive onto DMS and DLS after chronic ethanol consumption.


Asunto(s)
Alcoholismo/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Ácido Glutámico/metabolismo , Neostriado/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Conducta Animal , Depresores del Sistema Nervioso Central/administración & dosificación , Ciclo Estral/metabolismo , Etanol/administración & dosificación , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ratones , Neostriado/metabolismo , Neuronas/metabolismo , Autoadministración , Caracteres Sexuales , Factores Sexuales
15.
Neuropharmacology ; 164: 107857, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31756338

RESUMEN

Stress responses differ by sex, and females are more susceptible to developing mental illnesses because of past stress, including alcohol use disorder. Investigation of neuroadaptations governing the interaction between past stress and future alcohol intake remains understudied in females. A history of footshock stress previously was shown to increase alcohol self-administration under relapse-like conditions in male rats, associated with elevated phosphodiesterase 10A (PDE10A) mRNA expression in the dorsomedial prefrontal cortex and basolateral amygdala. To identify sex differences in long-term stress effects, male and female Wistar rats were exposed to light-cued footshock stress prior to alcohol self-administration training. While past stress did not alter acquisition or extinction, reacquisition self-administration was oppositely impacted by past stress. Stress history slightly increased reacquisition self-administration in males, but reduced alcohol self-administration in females, relative to same-sex controls. Control females self-administered less alcohol following glucocorticoid receptor inhibition by mifepristone, which did not significantly alter alcohol consumption in the other groups. PDE10A expression in synaptically enriched fractions also differed by sex and stress history in a brain region-specific manner. Females expressed more synaptic PDE10A than males in basolateral amygdala and dorsolateral striatum, regardless of stress history, whereas dorsomedial prefrontal cortex PDE10A protein levels matched group differences in reacquisition drinking, but also were expressed at much lower levels than all other regions examined. Together, these data show stress history differentially impacts alcohol self-administration and PDE10A expression by sex, with control females consuming alcohol in a glucocorticoid receptor-sensitive fashion that may relate to sex differences in PDE10A expression.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Glucocorticoides/farmacología , Hidrolasas Diéster Fosfóricas/biosíntesis , Estrés Psicológico/psicología , Animales , Química Encefálica/efectos de los fármacos , Electrochoque , Extinción Psicológica , Femenino , Masculino , Mifepristona/farmacología , Ratas , Ratas Wistar , Autoadministración , Caracteres Sexuales
16.
J Neurochem ; 109(5): 1459-68, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19453942

RESUMEN

Alcoholism is a chronically relapsing condition, indicative of long-term neuronal adaptations maintaining the disease even after prolonged abstinence. Previously, we identified brain-derived neurotrophic factor (BDNF) in the dorsal striatum as the central mediator of a homeostatic mechanism which is activated by acute alcohol (ethanol) exposure and functions to decrease the sensitivity of rodents to ethanol-related behaviors. We hypothesized that extensive exposure to ethanol would result in dysregulation of this BDNF-mediated protective mechanism, accompanied by heightened ethanol intake. In this study, we demonstrate that while a single bout of ethanol intake increases BDNF mRNA expression in the dorsal striatum, this effect is no longer observed after 6 weeks of daily ethanol access. Additionally, 6 weeks of ethanol consumption decreases BDNF in the cortex, a main source of BDNF for the striatum. Importantly, these ethanol-induced changes in BDNF levels are not ameliorated by 2 weeks' abstinence. Together, these data suggest that the BDNF pathway, which is activated following a single bout of ethanol drinking, breaks down by the end of 6 weeks of access and does not recover its protective function after a 2-week deprivation period. These results suggest that the persistence of altered BDNF signaling may contribute to the inflexibility of addictive behaviors.


Asunto(s)
Consumo de Bebidas Alcohólicas/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Regulación de la Expresión Génica/fisiología , Consumo de Bebidas Alcohólicas/fisiopatología , Análisis de Varianza , Animales , Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/genética , Conducta de Elección/fisiología , Preferencias Alimentarias/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Autoadministración
17.
FASEB J ; 22(7): 2393-404, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18310464

RESUMEN

We recently identified brain-derived neurotrophic factor (BDNF) in the dorsal striatum to be a major component of a homeostatic pathway controlling ethanol consumption. We hypothesized that ethanol-mediated activation of the BDNF signaling cascade is required for the ethanol-related function of the neurotrophic factor. Here, we demonstrate that exposure of striatal neurons to ethanol results in the activation of the BDNF receptor TrkB, leading to the activation of the mitogen-activated protein kinase (MAP kinase) signaling pathway and the subsequent increase in the expression of preprodynorphin (Pdyn) via BDNF. Finally, we show that activation of the dynorphin receptor, the kappa opioid receptor (KOR), is required for the BDNF-mediated decrease in ethanol intake, illustrating a function of dynorphin in BDNF's homeostatic control of ethanol consumption. Taken together, these results demonstrate that BDNF regulates ethanol intake by initiation of MAP kinase signaling and the ensuing production of downstream gene products, including Pdyn.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/genética , Cuerpo Estriado/fisiopatología , Dinorfinas/fisiología , Neuronas/fisiología , Animales , Técnicas de Cultivo de Célula , Cuerpo Estriado/fisiología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/patología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Alcohol ; 74: 3-9, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30033149

RESUMEN

Alcohol use disorder (AUD) is a pervasive societal problem, marked by high levels of alcohol intake and recidivism. Despite these common disease traits, individuals diagnosed with AUD display a range of disordered drinking and alcohol-related behaviors. The diversity in disease presentation, as well as the established polygenic nature of the disorder and complex neurocircuitry, speaks to the variety of neurochemical changes resulting from alcohol intake that may differentially regulate alcohol-related behaviors. Investigations into the molecular adaptations responsible for maladaptive alcohol-related behavioral outcomes require an ever-evolving set of molecular tools to elucidate with increasing precision how alcohol alters behavior through neurochemical changes. This review highlights recent advances in molecular methodology, addressing how incorporation of these cutting-edge techniques not only may enhance current knowledge of the molecular bases of AUD, but also may facilitate identification of improved treatment targets that may be therapeutic in specific subpopulations of AUD individuals.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Alcoholismo/genética , Alcoholismo/psicología , Alcoholismo/terapia , Animales , Proteína 9 Asociada a CRISPR/genética , Edición Génica , Regulación de la Expresión Génica , Humanos , Interferencia de ARN , ARN Mensajero/aislamiento & purificación
19.
Alcohol ; 72: 49-59, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30227988

RESUMEN

Alcohol use disorder is a widespread mental illness characterized by periods of abstinence followed by recidivism, and stress is the primary trigger of relapse. Despite the higher prevalence of alcohol use disorder in males, the relationship between stress and behavioral features of relapse, such as craving, is stronger in females. Given the greater susceptibility of females to stress-related psychiatric disorders, understanding sexual dimorphism in the relationship between stress and alcohol use is essential to identifying better treatments for both male and female alcoholics. This review addresses sex differences in the impact of stressors on alcohol drinking and seeking in rodents and humans. As these behavioral differences in alcohol use and relapse originate from sexual dimorphism in neuronal function, the impact of stressors and alcohol, and their interaction, on molecular adaptations and neural activity in males and females will also be discussed. Together, the data reviewed herein, arising from a symposium titled "Sex matters in stress-alcohol interactions" presented at the Fourth Volterra Conference on Stress and Alcohol, will highlight the importance of identifying sex differences to improve treatments for comorbid stress and alcohol use disorder in both sexes.


Asunto(s)
Alcoholismo/metabolismo , Encéfalo/metabolismo , Estrés Fisiológico/fisiología , Estrés Psicológico/metabolismo , Animales , Trastornos de Ansiedad/metabolismo , Corticosterona/metabolismo , Femenino , Humanos , Hidrocortisona/metabolismo , Masculino , Vías Nerviosas , Caracteres Sexuales , Factores Sexuales , Trastornos por Estrés Postraumático/metabolismo
20.
Front Psychiatry ; 9: 28, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29497387

RESUMEN

Alcoholism is a pervasive societal problem, yet available pharmacotherapies fail to treat most sufferers. The type 1 corticotropin-releasing factor (CRF1) receptor has received much attention for its putative role in the progression to alcohol dependence, although at present its success in clinical trials has been limited. Two single-nucleotide polymorphisms in the rat Crhr1 promoter have been identified in the Marchigian substrain of Sardinian alcohol-preferring (msP) rats. Unlike other Wistar-derived alcohol-preferring lines, nondependent msP rats reduce their alcohol self-administration in response to CRF1 antagonists and show increased brain CRF1 expression. The current study tested the hypotheses that the A alleles in the Crhr1 promoter polymorphisms are: (1) unique to msP (vs. CRF1 antagonist-insensitive) alcohol-preferring lines and (2) associate with greater alcohol preference or intake. Two related polymorphisms were observed in which both loci on a given chromosome were either mutant variant (A) or wild-type (G) alleles within the distal Crhr1 promoter of 17/25 msP rats (68%), as compared to 0/23 Indiana P rats, 0/20 Sardinian alcohol-preferring rats bred at Scripps (Scr:sP) and 0/21 outbred Wistar rats. Alcohol consumption in msP rats did not differ according to the presence of Crhr1 A alleles, but greater alcohol preference (98%) was observed in A allele homozygous msP rats (AA) compared to msP rats with wild-type (GG, 91%) or heterozygous (GA, 91%) genotypes. The greater alcohol preference reflected decreased water intake, accompanied by reduced total calories consumed by AA rats. The data show that msP rats differentially possess mutant A variant alleles in the polymorphic promoter region of the Crhr1 gene that may differentially regulate consumption.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA