Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
bioRxiv ; 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38260700

ABSTRACT

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.

2.
eNeuro ; 10(3)2023 03.
Article in English | MEDLINE | ID: mdl-36808099

ABSTRACT

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.


Subject(s)
Anxiety , Parvalbumins , Male , Mice , Female , Animals , Parvalbumins/metabolism , Anxiety/pathology , Neurons/metabolism , Anxiety Disorders , Emotions , Interneurons/physiology , Prefrontal Cortex/metabolism , Stress, Psychological/pathology
3.
Learn Mem ; 30(1): 1-11, 2023 01.
Article in English | MEDLINE | ID: mdl-36543385

ABSTRACT

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.


Subject(s)
Stress Disorders, Post-Traumatic , Rats , Male , Female , Animals , Odorants , Anxiety , Memory, Short-Term/physiology , Memory Disorders
4.
Neuropharmacology ; 190: 108564, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33857521

ABSTRACT

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.


Subject(s)
Alcoholism/metabolism , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Glutamic Acid/metabolism , Neostriatum/drug effects , Neurons/drug effects , Animals , Behavior, Animal , Central Nervous System Depressants/administration & dosage , Estrous Cycle/metabolism , Ethanol/administration & dosage , Excitatory Postsynaptic Potentials/drug effects , Mice , Neostriatum/metabolism , Neurons/metabolism , Self Administration , Sex Characteristics , Sex Factors
5.
Alcohol Clin Exp Res ; 45(2): 480-489, 2021 02.
Article in English | MEDLINE | ID: mdl-33351976

ABSTRACT

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.


Subject(s)
Alcohol Drinking/psychology , Anxiety/psychology , Housing, Animal , Sex Characteristics , Social Isolation/psychology , Stress, Psychological/psychology , Alcohol Drinking/adverse effects , Animals , Female , Male , Rats , Rats, Wistar , Recurrence , Self Administration
6.
Neuropharmacology ; 164: 107857, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31756338

ABSTRACT

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.


Subject(s)
Alcohol Drinking/psychology , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Glucocorticoids/pharmacology , Phosphoric Diester Hydrolases/biosynthesis , Stress, Psychological/psychology , Animals , Brain Chemistry/drug effects , Electroshock , Extinction, Psychological , Female , Male , Mifepristone/pharmacology , Rats , Rats, Wistar , Self Administration , Sex Characteristics
7.
Addict Biol ; 24(3): 335-343, 2019 05.
Article in English | MEDLINE | ID: mdl-29726054

ABSTRACT

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.


Subject(s)
Alcoholism/etiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Alcoholism/physiopathology , Central Nervous System Depressants/pharmacology , Dopaminergic Neurons/physiology , Ethanol/pharmacology , Humans , Limbic System/pathology , Mental Disorders/etiology , Mental Disorders/physiopathology , Nucleus Accumbens/physiology , Signal Transduction/physiology , Tegmentum Mesencephali/physiology
8.
Alcohol ; 74: 3-9, 2019 02.
Article in English | MEDLINE | ID: mdl-30033149

ABSTRACT

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.


Subject(s)
Alcohol Drinking/genetics , Alcoholism/genetics , Alcoholism/psychology , Alcoholism/therapy , Animals , CRISPR-Associated Protein 9/genetics , Gene Editing , Gene Expression Regulation , Humans , RNA Interference , RNA, Messenger/isolation & purification
9.
J Neurosci ; 38(44): 9423-9432, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30381434

ABSTRACT

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.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Emotions/physiology , Motivation/physiology , Resilience, Psychological , Sex Characteristics , Stress, Psychological/metabolism , Animals , Brain/metabolism , Brain/pathology , Female , Humans , Male , Mental Disorders/metabolism , Mental Disorders/pathology , Mental Disorders/psychology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Risk Factors , Stress, Psychological/pathology , Stress, Psychological/psychology
10.
Alcohol ; 72: 49-59, 2018 11.
Article in English | MEDLINE | ID: mdl-30227988

ABSTRACT

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.


Subject(s)
Alcoholism/metabolism , Brain/metabolism , Stress, Physiological/physiology , Stress, Psychological/metabolism , Animals , Anxiety Disorders/metabolism , Corticosterone/metabolism , Female , Humans , Hydrocortisone/metabolism , Male , Neural Pathways , Sex Characteristics , Sex Factors , Stress Disorders, Post-Traumatic/metabolism
11.
Front Psychiatry ; 9: 28, 2018.
Article in English | MEDLINE | ID: mdl-29497387

ABSTRACT

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.

12.
J Neurosci ; 37(17): 4593-4603, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28363981

ABSTRACT

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.


Subject(s)
Alcoholism/metabolism , Calcium Channels, L-Type/metabolism , Central Amygdaloid Nucleus/metabolism , Alcohol Drinking/psychology , Alcoholism/physiopathology , Alcoholism/psychology , Animals , Behavior, Animal , Central Amygdaloid Nucleus/physiopathology , Central Nervous System Depressants/pharmacology , Emotions , Ethanol/pharmacology , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/metabolism
13.
J Neurosci ; 37(5): 1139-1155, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27986929

ABSTRACT

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.


Subject(s)
Alcohol Drinking/genetics , Alcohol Drinking/psychology , Alcoholism/genetics , Alcoholism/psychology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology , Animals , Body Weight/drug effects , Conditioning, Operant/drug effects , Female , Gene Knockout Techniques , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Knockout , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/metabolism , Rats , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics , Toll-Like Receptor 4/antagonists & inhibitors
14.
Neuropharmacology ; 114: 123-134, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27899281

ABSTRACT

Alcohol use disorders are chronically relapsing conditions that pose significant health challenges for our society. Stress is a prevalent trigger of relapse, particularly for women, yet the mechanisms by which alcohol and stress interact, and how this differs between males and females, remain poorly understood. The glutamatergic circuit connecting the basolateral (BLA) and central (CeA) nuclei of the amygdala is a likely locus for such adaptations, yet the impact of alcohol, corticosterone and their interaction on this circuit has been understudied. In particular, no studies have addressed sex differences in these effects or potential differential responses between the lateral and medial subdivisions of the central nucleus. Thus, we assessed the effects of alcohol and corticosterone treatments on BLA-evoked compound glutamatergic responses in medial and lateral CeA neurons from male and female rats. We observed minimal differences between medial and lateral CeA responses to alcohol and corticosterone in male rats, which were primarily sensitive to alcohol-induced inhibition of glutamatergic postsynaptic potentials. Unlike male neurons, cells from female rats displayed reduced sensitivity to alcohol's inhibitory effects. In addition, female neurons diverged in their sensitivity to corticosterone, with lateral CeA neuronal responses significantly blunted following corticosterone treatment and medial CeA neurons largely unchanged by corticosterone or subsequent co-application of alcohol. Together these data highlight striking differences in how male and female amygdala respond to alcohol and the stress hormone corticosterone, factors which may impact differential susceptibility of the sexes to alcohol- and stress-related disorders.


Subject(s)
Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/physiology , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/physiology , Corticosterone/administration & dosage , Ethanol/administration & dosage , Neurons/drug effects , Neurons/physiology , Animals , Excitatory Postsynaptic Potentials/drug effects , Female , Male , Rats , Rats, Wistar , Sex Characteristics
15.
Alcohol ; 49(8): 795-802, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26095589

ABSTRACT

Alcohol use disorders are chronically relapsing conditions characterized by persistent drinking despite the negative impact on one's life. The difficulty of achieving and maintaining sobriety suggests that current treatments fail to fully address the underlying causes of alcohol use disorders. Identifying additional pathways controlling alcohol consumption may uncover novel targets for medication development to improve treatment options. One family of proteins recently implicated in the regulation of alcohol consumption is the cyclic nucleotide phosphodiesterases (PDEs). As an integral component in the regulation of the second messengers cyclic AMP and cyclic GMP, and thus their cognate signaling pathways, PDEs present intriguing targets for pharmacotherapies to combat alcohol use disorders. As activation of cAMP/cGMP-dependent signaling cascades can dampen alcohol intake, PDE inhibitors may provide a novel target for reducing excessive alcohol consumption, as has been proposed for PDE4 and PDE10A. This review highlights preclinical literature demonstrating the involvement of cyclic nucleotide-dependent signaling in neuronal and behavioral responses to alcohol, as well as detailing the capacity of various PDE inhibitors to modulate alcohol intake. Together these data provide a framework for evaluating the potential utility of PDE inhibitors as novel treatments for alcohol use disorders.


Subject(s)
Alcohol Drinking/metabolism , Behavior, Animal/drug effects , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Adenylyl Cyclases/metabolism , Animals , Behavior, Animal/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Guanylate Cyclase/metabolism , Mice , Phosphodiesterase 4 Inhibitors/pharmacology , Rats , Signal Transduction
16.
J Clin Invest ; 125(8): 3193-7, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26121746

ABSTRACT

Alcoholism, or alcohol use disorder, is a major public health concern that is a considerable risk factor for morbidity and disability; therefore, effective treatments are urgently needed. Here, we demonstrated that the glucocorticoid receptor (GR) antagonist mifepristone reduces alcohol intake in alcohol-dependent rats but not in nondependent animals. Both systemic delivery and direct administration into the central nucleus of the amygdala, a critical stress-related brain region, were sufficient to reduce alcohol consumption in dependent animals. We also tested the use of mifepristone in 56 alcohol-dependent human subjects as part of a double-blind clinical and laboratory-based study. Relative to placebo, individuals who received mifepristone (600 mg daily taken orally for 1 week) exhibited a substantial reduction in alcohol-cued craving in the laboratory, and naturalistic measures revealed reduced alcohol consumption during the 1-week treatment phase and 1-week post-treatment phase in mifepristone-treated individuals. Mifepristone was well tolerated and improved liver-function markers. Together, these results support further exploration of GR antagonism via mifepristone as a therapeutic strategy for alcoholism.


Subject(s)
Alcohol Drinking/drug therapy , Alcoholism/drug therapy , Hormone Antagonists/administration & dosage , Mifepristone/administration & dosage , Receptors, Glucocorticoid/antagonists & inhibitors , Administration, Oral , Adult , Alcohol Drinking/physiopathology , Alcoholism/physiopathology , Animals , Female , Hormone Antagonists/adverse effects , Humans , Male , Mifepristone/adverse effects , Rats
17.
J Neurosci ; 35(15): 6241-53, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878294

ABSTRACT

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.


Subject(s)
Alcohol Drinking/physiopathology , Brain/metabolism , Compulsive Behavior/complications , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Reward , Animals , Brain/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Central Nervous System Depressants/administration & dosage , Conditioning, Operant/drug effects , Disease Models, Animal , Ethanol/administration & dosage , Glutamate Decarboxylase/metabolism , Male , Phosphopyruvate Hydratase/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Quinine/administration & dosage , Rats , Rats, Wistar , Self Administration , Time Factors
18.
Brain Res ; 1628(Pt A): 60-7, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-25801118

ABSTRACT

Growth factors, long studied for their involvement in neuronal development and plasticity, also regulate responses to drugs of abuse, including alcohol. This review details the intricate interaction between the Brain-Derived Neurotrophic Factor (BDNF) and alcohol, and provides evidence to suggest that corticostriatal BDNF signaling acts to keep alcohol drinking in moderation. Specifically, we describe studies in rodent models suggesting that moderate consumption of alcohol increases BDNF levels in the dorsal striatum, which in turn act to suppress alcohol intake by activating a Mitogen-Activated Protein Kinase (MAPK)-dependent genomic mechanism. We further provide data to suggest that alcohol intake levels escalate when the endogenous corticostriatal BDNF pathway becomes dysregulated. Finally, we summarize recent studies suggesting that specific microRNAs targeting BDNF mRNA in the medial prefrontal cortex (mPFC) regulate the breakdown of the protective corticostriatal BDNF pathway.


Subject(s)
Alcoholism/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Animals , Central Nervous System Depressants/pharmacology , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Ethanol/pharmacology , Neural Pathways/drug effects , Neural Pathways/metabolism
19.
Article in English | MEDLINE | ID: mdl-24782725

ABSTRACT

Alcohol use disorders are persistent problems with high recidivism rates despite repeated efforts to quit drinking. Neuroadaptations that result from alcohol exposure and that persist during periods of abstinence represent putative molecular determinants of the propensity to relapse. Previously we demonstrated a positive association between phosphodiesterase 10A (PDE10A) gene expression and elevations in relapse-like alcohol self-administration in rats with a history of stress exposure. Because alcohol withdrawal is characterized by heightened anxiety-like behavior, activation of stress-responsive brain regions and an elevated propensity to self-administer alcohol, we hypothesized that Pde10a expression also would be upregulated in reward- and stress-responsive brain regions during periods of acute (8-10 h) and protracted (6 weeks) alcohol withdrawal. During acute withdrawal, elevated Pde10a mRNA expression was found in the medial and basolateral amygdala (BLA), as well as the infralimbic and anterior cingulate subdivisions of the medial prefrontal cortex, relative to alcohol-naïve controls. The BLA was the only region with elevated Pde10a mRNA expression during both acute and protracted withdrawal. In contrast to the elevations, Pde10a mRNA levels tended to be reduced during protracted withdrawal in the dorsal striatum, prelimbic prefrontal cortex, and medial amygdala. Together these results implicate heightened PDE10A expression in the BLA as a lasting neuroadaptation associated with alcohol dependence.

20.
Neuropsychopharmacology ; 39(7): 1722-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24549104

ABSTRACT

A history of stress produces increases in rodent relapse-like alcohol self-administration behavior and regional brain gene expression of phosphodiesterase 10A (PDE10A), a dual-specificity cyclic adenosine monophosphate/cyclic guanosine monophosphate-inhibiting enzyme. Here, we tested the hypothesis that administration of TP-10, a specific PDE10A inhibitor, would reduce alcohol self-administration in conditions predisposing to elevated self-administration. TP-10 administration dose-dependently (0.562, 1.0 mg/kg; subcutaneously) reduced relapse-like alcohol self-administration regardless of stress history enhancement of relapse-like behavior. TP-10 also reduced alcohol self-administration in genetically alcohol-preferring rats, as well as in alcohol-non-dependent and -dependent rats. Effective systemic TP-10 doses did not alter alcohol pharmacokinetics, significantly reduce motor activity or intrabout operant response speed, or promote a conditioned place aversion. TP-10 also reduced saccharin self-administration, suggesting a general role for PDE10A in the self-administration of reinforcing substances. PDE10A inhibition in the dorsolateral striatum, but not the nucleus accumbens, reduced alcohol self-administration. Taken together, the results implicate dorsolateral striatum PDE10A in facilitating alcohol intake and support further investigation of PDE10A systems in the pathophysiology and potential treatment of substance use disorders.


Subject(s)
Alcohols/administration & dosage , Conditioning, Operant/drug effects , Phosphoric Diester Hydrolases/metabolism , Reinforcement, Psychology , Saccharin/administration & dosage , Sweetening Agents/administration & dosage , 2-Hydroxypropyl-beta-cyclodextrin , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Drug Administration Routes , Enzyme Inhibitors/pharmacology , Male , Motor Activity/drug effects , Pyrazoles/pharmacology , Quinolines/pharmacology , Rats , Rats, Wistar , Reinforcement Schedule , Self Administration , beta-Cyclodextrins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...