1
|
Griffith AK, Martel MM, Fillmore MT. Effect of menstrual cycle on rewarding properties of alcohol cues in women. PSYCHOLOGY OF ADDICTIVE BEHAVIORS 2024; 38:676-687. [PMID: 38059946 PMCID: PMC11156796 DOI: 10.1037/adb0000978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
OBJECTIVE Compared with men, women are disproportionately affected by alcohol, including greater risks of behavioral impairment and relapse from abstinence-based treatments. One potential mechanism underlying this disparity is ovarian hormone fluctuations across menstrual cycle phases, particularly estradiol (E2). Preclinical and clinical studies have shown that E2 levels positively correlate with alcohol consumption, suggesting E2 modulates drinking. Rewarding properties of alcohol are thought to mediate this relationship. The present study tested the degree to which women report increased rewarding effects from alcohol and heightened attention to alcohol-related cues when E2 was elevated during the late follicular phase of the menstrual cycle. METHOD Fifty women aged 21-29 participated in a within-subjects placebo-controlled study examining how menstrual cycle phase alters the rewarding properties of alcohol and alcohol-associated cues when sober and intoxicated, as measured by their attentional bias toward alcohol-associated cues and subjective reports. Measures were obtained following 0.60 g/kg alcohol and placebo during the early follicular phase when E2 was low and the late follicular phase (i.e., ovulation) when E2 was elevated. RESULTS Attentional bias to alcohol-associated cues was greater during the late follicular phase in both sober and intoxicated states. Women reported rewarding effects from alcohol, but no effects of phase were observed. CONCLUSIONS The findings suggest that the rewarding properties of alcohol-associated cues might be enhanced during the late follicular phase of the menstrual cycle when E2 is elevated, possibly increasing the risk for excessive drinking in women during this phase. (PsycInfo Database Record (c) 2024 APA, all rights reserved).
Collapse
Affiliation(s)
- Annie K. Griffith
- Department of Psychology, University of Kentucky College of Arts and Sciences, 110 Kastle Hall, Lexington, KY 40506-0044, U.S.A
| | - Michelle M. Martel
- Department of Psychology, University of Kentucky College of Arts and Sciences, 110 Kastle Hall, Lexington, KY 40506-0044, U.S.A
| | - Mark T. Fillmore
- Department of Psychology, University of Kentucky College of Arts and Sciences, 110 Kastle Hall, Lexington, KY 40506-0044, U.S.A
| |
Collapse
|
2
|
Griffith AK, Martel MM, Eisenlohr-Moul T, Fillmore MT. Heightened sensitivity to the disinhibiting effect of alcohol in women during the late follicular phase of the menstrual cycle. Exp Clin Psychopharmacol 2023; 31:839-848. [PMID: 36265052 PMCID: PMC10302159 DOI: 10.1037/pha0000611] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Compared with men, women are disproportionately affected by alcohol, including greater risks of physiological damage, behavioral impairment, and relapse. One likely mechanism underlying the sexual disparity in this vulnerability is the fluctuation of ovarian hormones, particularly estradiol (E2), across phases of the menstrual cycle. Several preclinical and clinical studies have shown that higher E2 levels positively correlate with drinking, suggesting E2 may play a significant role in modulating drinking. Inhibitory control also modulates drinking; when it is reduced or compromised by alcohol, the drinker's ability to stop the self-administration of alcohol could be impaired, leading to a binge episode. The present study aimed to examine the degree to which menstrual cycle phase can influence the disinhibiting effect of alcohol. Twenty-four healthy young adult women participated in a within-subjects placebo-controlled study of the acute disinhibiting effect of 0.60 g/kg alcohol over the course of two test sessions. A cued go/no-go task measured the disinhibiting effects of alcohol and placebo beverages during the early follicular phase of the cycle when E2 levels were low and the late follicular phase (i.e., ovulation) when E2 was elevated. Results showed that the disinhibiting effect of alcohol increased nearly twofold during the late follicular phase when E2 was elevated. These findings highlight the role of alcohol-induced disinhibition as a potential behavioral mechanism by which fluctuations in ovarian hormones as a function of the menstrual cycle contribute to increased risk for excessive alcohol use in women. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Annie K Griffith
- Department of Psychology, University of Kentucky, College of Arts and Sciences
| | - Michelle M Martel
- Department of Psychology, University of Kentucky, College of Arts and Sciences
| | - Tory Eisenlohr-Moul
- Neuropsychiatric Institute, Department of Psychiatry, University of Illinois
| | - Mark T Fillmore
- Department of Psychology, University of Kentucky, College of Arts and Sciences
| |
Collapse
|
3
|
Li X, Shi Z, Byanyima J, Morgan PT, van der Veen JW, Zhang R, Deneke E, Wang GJ, Volkow ND, Wiers CE. Brain glutamate and sleep efficiency associations following a ketogenic diet intervention in individuals with Alcohol Use Disorder. DRUG AND ALCOHOL DEPENDENCE REPORTS 2022; 5:100092. [PMID: 36311277 PMCID: PMC9601174 DOI: 10.1016/j.dadr.2022.100092] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background We previously showed that ketogenic diet (KD) was effective in curbing alcohol withdrawal and craving in individuals with alcohol use disorder (AUD). We hypothesized that the clinical benefits were due to improvements in sleep. To test this, we performed a secondary analysis on the KD trial data to (1) examine the effects of KD on total sleep time (TST) and sleep quality and (2) investigate the association between KD-induced alterations in cingulate glutamate concentration and changes in TST and sleep quality. Methods AUD individuals undergoing alcohol detoxification were randomized to receive KD (n=19) or standard American diet (SA; n=14) for three weeks. TST was measured weekly by self-report, GENEActive sleep accelerometer, and X4 Sleep Profiler ambulatory device. Sleep quality was assessed using subjectively ratings of sleep depth and restedness and Sleep Profiler (Sleep Efficiency [%]). Weekly 1H magnetic resonance spectroscopy scans measured cingulate glutamate levels. Results TST was lower in KD than SA and increased with effect of time. Sleep depth, restedness, and Sleep Efficiency improved with time, but exhibited no effect of diet. In KD and SA combined, week 1 cingulate glutamate levels correlated positively with Sleep Efficiency, but not with TST. Conclusions Although cingulate glutamate levels correlated positively with Sleep Efficiency in week 1, KD-induced glutamate elevation did not produce significant sleep improvements. Rather, KD was associated with lower TST than SA. Given the well-established associations between sleep and alcohol relapse, longer follow up assessment of KD's impact on sleep in AUD is warranted.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Zhenhao Shi
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Juliana Byanyima
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Peter T. Morgan
- Department of Psychiatry, Yale University, New Haven, CT, 06519, USA
- Department of Psychiatry, Bridgeport Hospital, Bridgeport, CT, 06610, USA
| | | | - Rui Zhang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Erin Deneke
- Caron Treatment Centers, 243N Galen Hall Rd, Wernersville, PA, 19565, USA
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Corinde E. Wiers
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
- Corresponding author at: 3535 Market St Ste 500, Philadelphia, PA 19104.
| |
Collapse
|
4
|
Quijano Cardé NA, Shaw J, Carter C, Kim S, Stitzel JA, Venkatesh SK, Ramchandani VA, De Biasi M. Mutation of the α5 nicotinic acetylcholine receptor subunit increases ethanol and nicotine consumption in adolescence and impacts adult drug consumption. Neuropharmacology 2022; 216:109170. [PMID: 35752273 PMCID: PMC9308728 DOI: 10.1016/j.neuropharm.2022.109170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/16/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
Alcohol and nicotine are commonly used during adolescence, establishing long-lasting neuroplastic alterations that influence subsequent drug use and abuse. Drinking- and smoking-related traits have been extensively associated with variation in CHRNA5 - the gene that encodes the α5 subunit of neuronal nicotinic acetylcholine receptors (nAChRs). The single nucleotide polymorphism (SNP) rs16969968 in CHRNA5 encodes an amino acid substitution (D398N) that alters the function and pharmacokinetics of α5-containing nAChR. When expressed in rodents, this variant results in increased ethanol and nicotine operant self-administration. How disruption of α5-containing nAChRs influences adolescent ethanol and nicotine intake, and how it modulates interactions between these drugs has not been previously explored. In the present study, we examined volitional ethanol and nicotine consumption in adolescent mice (post-natal day 30-43) of both sexes with mutated (SNP) or lacking (KO) the α5 nAChR subunit. The effect of adolescent alcohol or nicotine exposure on home cage consumption of the opposite drug in adulthood and its modulation by Chrna5 mutation and sex were examined. During adolescence, we found that α5 nAChR disruption increases nicotine intake in mice of both sexes, but the effect on alcohol intake was only observed in females. The sex-specific increase in alcohol consumption in α5 SNP and KO was replicated in adulthood. The effect of adolescent alcohol or nicotine exposure on subsequent intake of the opposite drug in adulthood is modulated by sex and Chrna5 mutation. These observations suggest sex differences in the genetic architecture of alcohol dependence, and modulators of alcohol and nicotine interactions.
Collapse
Affiliation(s)
- Natalia A Quijano Cardé
- Pharmacology Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica Shaw
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina Carter
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Seung Kim
- Neuroscience Program, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Shyamala K Venkatesh
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Laboratory of Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Vijay A Ramchandani
- Laboratory of Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Mariella De Biasi
- Pharmacology Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Cortés-Tomás MT, Giménez-Costa JA, Motos-Sellés P, Sancerni-Beitia MD. Consequences, Motives, and Expectancies of Consumption as Predictors of Binge Drinking in University Women. Front Psychol 2022; 13:862334. [PMID: 35450336 PMCID: PMC9016132 DOI: 10.3389/fpsyg.2022.862334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/11/2022] [Indexed: 12/02/2022] Open
Abstract
The increasing presence of women, especially university women, in risky alcohol consumption such as Binge Drinking (BD), which is associated with gender-specific biopsychosocial problems, makes it necessary to analyze the variables underlying BD in order to adjust possible interventions more in line with their reality. The motives and expectancies of this pattern of consumption, as well as the consequences derived from it, are some of the variables that are shown to have the greatest weight in the prediction of BD. In the present study we analyze, on the one hand, the performance of these variables among college women with alcohol use, and on the other hand, which of these variables allow us to classify BD. A total of 501 female university consumers of alcohol (mean age 19.02 years) were assessed. Specifically, they completed a self-report of alcohol consumption (77.1% engage in BD), the Expectancy Questionnaire (EQ), the Drinking Motives Questionnaire (DMQ-R) and the Alcohol Consumption Consequences Evaluation (ACCE). BD female students scored significantly higher on these instruments, except for compliance motives. The logistic regression analysis carried out to estimate the probability of performing BD using the social and conformity motives, the ACCE and positive expectancies correctly estimated (χ2 8 = 9.149, p < 0.33) 88.6% of the cases and explained 26.2% of the BD. Thus, young women with a level of consequences classified as high risk (>25 in ACCE) have a 3.55-fold increase in the probability of having BD, compared to women classified as low risk by the ACCE. On the other hand, women classified as moderate risk by the ACCE have a 4.77-fold increase in the probability of having BD. In the case of social motives and positive expectancies, their increase multiplies by 1.165 and 1.024, respectively, the probability of having BD. The results of this study highlight the importance of adapting preventive measures to the consequences experienced by BD university students, especially in relation to the social motives and positive expectancies that modulate decision-making when engaging in this pattern of consumption.
Collapse
Affiliation(s)
| | | | - Patricia Motos-Sellés
- Department of Basic Psychology, Faculty of Psychology, University of Valencia, Valencia, Spain
| | - María-Dolores Sancerni-Beitia
- Department of Methodology of the Behavioral Sciences, Faculty of Psychology, University of Valencia, Valencia, Spain
| |
Collapse
|
6
|
Neuroadaptations and TGF-β signaling: emerging role in models of neuropsychiatric disorders. Mol Psychiatry 2022; 27:296-306. [PMID: 34131268 PMCID: PMC8671568 DOI: 10.1038/s41380-021-01186-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023]
Abstract
Neuropsychiatric diseases are manifested by maladaptive behavioral plasticity. Despite the greater understanding of the neuroplasticity underlying behavioral adaptations, pinpointing precise cellular mediators has remained elusive. This has stymied the development of pharmacological interventions to combat these disorders both at the level of progression and relapse. With increased knowledge on the putative role of the transforming growth factor (TGF- β) family of proteins in mediating diverse neuroadaptations, the influence of TGF-β signaling in regulating maladaptive cellular and behavioral plasticity underlying neuropsychiatric disorders is being increasingly elucidated. The current review is focused on what is currently known about the TGF-β signaling in the central nervous system in mediating cellular and behavioral plasticity related to neuropsychiatric manifestations.
Collapse
|
7
|
Biernacka JM, Coombes BJ, Batzler A, Ho AMC, Geske JR, Frank J, Hodgkinson C, Skime M, Colby C, Zillich L, Pozsonyiova S, Ho MF, Kiefer F, Rietschel M, Weinshilboum R, O’Malley SS, Mann K, Anton R, Goldman D, Karpyak VM. Genetic contributions to alcohol use disorder treatment outcomes: a genome-wide pharmacogenomics study. Neuropsychopharmacology 2021; 46:2132-2139. [PMID: 34302059 PMCID: PMC8505452 DOI: 10.1038/s41386-021-01097-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/23/2021] [Accepted: 07/08/2021] [Indexed: 01/09/2023]
Abstract
Naltrexone can aid in reducing alcohol consumption, while acamprosate supports abstinence; however, not all patients with alcohol use disorder (AUD) benefit from these treatments. Here we present the first genome-wide association study of AUD treatment outcomes based on data from the COMBINE and PREDICT studies of acamprosate and naltrexone, and the Mayo Clinic CITA study of acamprosate. Primary analyses focused on treatment outcomes regardless of pharmacological intervention and were followed by drug-stratified analyses to identify treatment-specific pharmacogenomic predictors of acamprosate and naltrexone response. Treatment outcomes were defined as: (1) time until relapse to any drinking (TR) and (2) time until relapse to heavy drinking (THR; ≥ 5 drinks for men, ≥4 drinks for women in a day), during the first 3 months of treatment. Analyses were performed within each dataset, followed by meta-analysis across the studies (N = 1083 European ancestry participants). Single nucleotide polymorphisms (SNPs) in the BRE gene were associated with THR (min p = 1.6E-8) in the entire sample, while two intergenic SNPs were associated with medication-specific outcomes (naltrexone THR: rs12749274, p = 3.9E-8; acamprosate TR: rs77583603, p = 3.1E-9). The top association signal for TR (p = 7.7E-8) and second strongest signal in the THR (p = 6.1E-8) analysis of naltrexone-treated patients maps to PTPRD, a gene previously implicated in addiction phenotypes in human and animal studies. Leave-one-out polygenic risk score analyses showed significant associations with TR (p = 3.7E-4) and THR (p = 2.6E-4). This study provides the first evidence of a polygenic effect on AUD treatment response, and identifies genetic variants associated with potentially medication-specific effects on AUD treatment response.
Collapse
Affiliation(s)
- Joanna M. Biernacka
- grid.66875.3a0000 0004 0459 167XDepartment of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA ,grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic, Rochester, MN USA
| | - Brandon J. Coombes
- grid.66875.3a0000 0004 0459 167XDepartment of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Anthony Batzler
- grid.66875.3a0000 0004 0459 167XDepartment of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Ada Man-Choi Ho
- grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic, Rochester, MN USA
| | - Jennifer R. Geske
- grid.66875.3a0000 0004 0459 167XDepartment of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Josef Frank
- grid.7700.00000 0001 2190 4373Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Colin Hodgkinson
- grid.420085.b0000 0004 0481 4802National Institute on Alcohol Abuse and Alcoholism, Rockville, MD USA
| | - Michelle Skime
- grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic, Rochester, MN USA
| | - Colin Colby
- grid.66875.3a0000 0004 0459 167XDepartment of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Lea Zillich
- grid.7700.00000 0001 2190 4373Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sofia Pozsonyiova
- grid.66875.3a0000 0004 0459 167XDepartment of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Ming-Fen Ho
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | - Falk Kiefer
- grid.7700.00000 0001 2190 4373Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marcella Rietschel
- grid.7700.00000 0001 2190 4373Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Richard Weinshilboum
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | | | - Karl Mann
- grid.7700.00000 0001 2190 4373Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ray Anton
- grid.259828.c0000 0001 2189 3475Medical University of South Carolina, Charleston, SC USA
| | - David Goldman
- grid.420085.b0000 0004 0481 4802National Institute on Alcohol Abuse and Alcoholism, Rockville, MD USA
| | - Victor M. Karpyak
- grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
8
|
Rabiant K, Antol J, Naassila M, Pierrefiche O. Sex difference in the vulnerability to hippocampus plasticity impairment after binge-like ethanol exposure in adolescent rat: Is estrogen the key? Addict Biol 2021; 26:e13002. [PMID: 33511744 DOI: 10.1111/adb.13002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 02/01/2023]
Abstract
Binge drinking during adolescence induces memory impairments, and evidences suggest that females are more vulnerable than males. However, the reason for such a difference is unclear, whereas preclinical studies addressing this question are lacking. Here we tested the hypothesis that endogenous estrogen level (E2) may explain sex differences in the effects of ethanol on hippocampus plasticity, the cellular mechanism of memory. Long-term depression (LTD) in hippocampus slice of pubertal female rats was recorded 24 h after two ethanol binges (3 g/kg, i.p., 9 h apart). Neither the estrous cycle nor ethanol altered LTD. However, if ethanol was administered during proestrus (i.e., at endogenous E2 peak), LTD was abolished 24 h later, whereas NMDA-fEPSPs response to a GluN2B antagonist increased. The abolition of LTD was not observed in adult female rats. Exogenous E2 combined with ethanol replicated LTD abolition in pubertal, prepubertal female, and in pubertal male rats without changes in ethanol metabolism. In male rats, a higher dose of ethanol was required to abolish LTD at 24-h delay. In pubertal female rats, tamoxifen, an antagonist of estrogen receptors, blocked the impairing effects of endogenous and exogenous E2 on LTD, suggesting estrogen interacts with ethanol through changes in gene expression. In addition, tamoxifen prevented LTD abolition at 24 h but not at 48-h delay. In conclusion, estrogen may explain the increased vulnerability to ethanol-induced plasticity impairment seen in females compared with males. This increased vulnerability of female rats is likely due to changes in the GluN2B subunit that represent a common target between ethanol and estrogen.
Collapse
Affiliation(s)
- Kevin Rabiant
- INSERM, UMR 1247 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances Univ Picardie Jules Verne, Centre Universitaire de Recherche en Santé (CURS) Amiens France
| | - Johan Antol
- INSERM, UMR 1247 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances Univ Picardie Jules Verne, Centre Universitaire de Recherche en Santé (CURS) Amiens France
| | - Mickael Naassila
- INSERM, UMR 1247 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances Univ Picardie Jules Verne, Centre Universitaire de Recherche en Santé (CURS) Amiens France
| | - Olivier Pierrefiche
- INSERM, UMR 1247 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances Univ Picardie Jules Verne, Centre Universitaire de Recherche en Santé (CURS) Amiens France
| |
Collapse
|
9
|
Sex Differences in the Brain Transcriptome Related to Alcohol Effects and Alcohol Use Disorder. Biol Psychiatry 2021; 91:43-52. [PMID: 34274109 PMCID: PMC8558111 DOI: 10.1016/j.biopsych.2021.04.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/05/2021] [Accepted: 04/26/2021] [Indexed: 01/03/2023]
Abstract
There is compelling evidence that sex and gender have crucial roles in excessive alcohol (ethanol) consumption. Here, we review some of the data from the perspective of brain transcriptional differences between males and females, focusing on rodent animal models. A key emerging transcriptional feature is the role of neuroimmune processes. Microglia are the resident neuroimmune cells in the brain and exhibit substantial functional differences between males and females. Selective breeding for binge ethanol consumption and the impacts of chronic ethanol consumption and withdrawal from chronic ethanol exposure all demonstrate sex-dependent neuroimmune signatures. A focus is on resolving sex-dependent differences in transcriptional responses to ethanol at the neurocircuitry level. Sex-dependent transcriptional differences are found in the extended amygdala and the nucleus accumbens. Telescoping of ethanol consumption is found in some, but not all, studies to be more prevalent in females. Recent transcriptional studies suggest that some sex differences may be due to female-dependent remodeling of the primary cilium. An interesting theme appears to be developing: at least from the animal model perspective, even when males and females are phenotypically similar, they differ significantly at the level of the transcriptome.
Collapse
|
10
|
Banu S, Srivastava S, Mohammed A, Kushawah G, Sowpati DT, Mishra RK. Tissue-specific transcriptome recovery on withdrawal from chronic alcohol exposure in zebrafish. Alcohol 2021; 91:29-38. [PMID: 33038458 DOI: 10.1016/j.alcohol.2020.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022]
Abstract
Alcohol consumption can lead to a wide range of systemic disorders brought about by transcriptional changes. Recent studies have documented altered behavior and physiology in zebrafish exposed to alcohol. In this work, we have identified the changes in the zebrafish transcriptome in response to chronic alcohol exposure. We have further followed the extent of transcriptional recovery upon withdrawal from alcohol and found evidence of tissue-specific responses. Our results indicate a greater extent of recovery of the brain transcriptome compared to the liver. We identify two distinct classes of genes in response to withdrawal from alcohol exposure - those that recover their pre-alcohol expression profile versus those that retain altered expression even after the fish are removed from the alcohol environment. Finally, we have examined gender-specific responses to alcohol exposure in zebrafish and find evidence for distinct alcohol tolerance levels. Upon chronic alcohol exposure, a higher percentage of genes show perturbation in expression profile in males compared to females. Female fish also recover better with more genes regaining the control expression level upon withdrawal from alcohol. Overall, our work identifies genes and pathways perturbed by exposure to alcohol, and demonstrates the extent of gender- and tissue-specific transcriptional changes associated with chronic alcoholism and withdrawal.
Collapse
|
11
|
Guinle MIB, Sinha R. The Role of Stress, Trauma, and Negative Affect in Alcohol Misuse and Alcohol Use Disorder in Women. Alcohol Res 2020; 40:05. [PMID: 32832310 PMCID: PMC7431322 DOI: 10.35946/arcr.v40.2.05] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent evidence indicates that the United States is facing a public health crisis of alcohol misuse and alcohol use disorder (AUD), which has been fueled in part by dramatic rises in binge and heavy drinking and prevalence of AUD in women. Historically, alcohol misuse and AUD have been more prevalent in men than in women. However, recent evidence on data from the past decade shows increases in AUD prevalence rates that are associated with substantially higher binge and heavy drinking and AUD prevalence in women compared to men. This paper first addresses the key roles of stress, trauma, childhood maltreatment, negative affect, and mood and anxiety disorders; sex differences in the presentation of these psychosocial and psychological factors; and their contributions to alcohol misuse, escalation to binge and heavy drinking, and transition to AUD in women. Also examined are potential central and peripheral biological mechanisms by which stressors and traumatic experiences, as well as chronic stress states-including depression and anxiety-may facilitate differential pathways to alcohol misuse, escalation, and transition to AUD in women. Finally, this paper discusses major gaps in the literature on sex differences in these areas as well as the need for greater research on sex-specific pathways to alcohol misuse and transition to AUD, so as to support a more comprehensive understanding of AUD etiology and for the development of new strategies for prevention and treatment of alcohol misuse and AUD in women.
Collapse
Affiliation(s)
| | - Rajita Sinha
- Yale Stress Center, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
12
|
Gano A, Mondello JE, Doremus-Fitzwater TL, Deak T. Rapid alterations in neuroimmune gene expression after acute ethanol: Timecourse, sex differences and sensitivity to cranial surgery. J Neuroimmunol 2019; 337:577083. [PMID: 31675629 PMCID: PMC6866658 DOI: 10.1016/j.jneuroim.2019.577083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 01/08/2023]
Abstract
Prior work has established that that an acute ethanol challenge mimicking high intensity alcohol consumption increased IL-6 and suppressed IL-1β and TNFα mRNA in intoxication, with the opposite pattern seen in withdrawal. These experiments utilized Sprague-Dawley rats to further extend these results across time course (from 45 min to 6 h after ethanol), sex, and central versus peripheral expression. Furthermore, these data show that cannulation surgery may selectively modify the central neuroimmune response to ethanol. These findings highlight a unique plasticity of IL-6 that is specific to central structures and responsive to alterations by environmental factors.
Collapse
Affiliation(s)
- Anny Gano
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States of America
| | - Jamie E Mondello
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States of America
| | - Tamara L Doremus-Fitzwater
- Department of Psychology, Williams Hall, Ithaca College, 953 Danby Road, Ithaca, NY 14850, United States of America
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States of America.
| |
Collapse
|
13
|
Greiner JFW, Merten M, Kaltschmidt C, Kaltschmidt B. Sexual dimorphisms in adult human neural, mesoderm-derived, and neural crest-derived stem cells. FEBS Lett 2019; 593:3338-3352. [PMID: 31529465 DOI: 10.1002/1873-3468.13606] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022]
Abstract
Sexual dimorphisms contribute, at least in part, to the severity and occurrence of a broad range of neurodegenerative, cardiovascular, and bone disorders. In addition to hormonal factors, increasing evidence suggests that stem cell-intrinsic mechanisms account for sex-specific differences in human physiology and pathology. Here, we discuss sex-related intrinsic mechanisms in adult stem cell populations, namely mesoderm-derived stem cells, neural stem cells (NSCs), and neural crest-derived stem cells (NCSCs), and their implications for stem cell differentiation and regeneration. We particularly focus on sex-specific differences in stem cell-mediated bone regeneration, in neuronal development, and in NSC-mediated neuroprotection. Moreover, we review our own recently published observations regarding the sex-dependent role of NF-κB-p65 in neuroprotection of human NCSC-derived neurons and sex differences in NCSC-related disorders, so-called neurocristopathies. These observations are in accordance with the increasing evidence pointing toward sex-specific differences in neurocristopathies and degenerative diseases like Parkinson's disease or osteoporosis. All findings discussed here indicate that sex-specific variability in stem cell biology may become a crucial parameter for the design of future treatment strategies.
Collapse
Affiliation(s)
| | - Madlen Merten
- Molecular Neurobiology, Bielefeld University, Germany
| | | | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, Germany.,Molecular Neurobiology, Bielefeld University, Germany
| |
Collapse
|
14
|
Gano A, Vore AS, Sammakia M, Deak T. Assessment of Extracellular Cytokines in the Hippocampus of the Awake Behaving Rat Using Large-Molecule Microdialysis Combined with Multiplex Arrays After Acute and Chronic Ethanol Exposure. Alcohol Clin Exp Res 2019; 43:640-654. [PMID: 30667526 PMCID: PMC6443416 DOI: 10.1111/acer.13963] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/14/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Studies have demonstrated persistent changes in central nervous system (CNS) cytokine gene expression following ethanol (EtOH) exposure. However, the low endogenous expression and short half-lives of cytokines in the CNS have made cytokine protein detection challenging. The goal of these studies was to establish parameters for use of large-molecule microdialysis and sensitive multiplexing technology for the simultaneous detection of brain cytokines, corticosterone (CORT), and EtOH concentrations in the awake behaving rat. METHODS Adult (P75+) male Sprague Dawley rats that were either naïve to EtOH (Experiment 1) or had a history of adolescent chronic intermittent EtOH (CIE; Experiment 2) were given an acute EtOH challenge during microdialysis. Experiment 1 examined brain EtOH concentrations, CORT and a panel of neuroimmune analytes, including cytokines associated with innate and adaptive immunity. The natural time course of changes in these cytokines was compared to the effects of an acute 1.5 or 3.0 g/kg intraperitoneal (i.p.) EtOH challenge. In Experiment 2, rats with a history of adolescent CIE or controls exposed to vehicle were challenged with 3.0 g/kg i.p. EtOH during microdialysis in adulthood, and a panel of cytokines was examined in parallel with brain EtOH concentrations and CORT. RESULTS The microdialysis procedure itself induced a cytokine-specific response that replicated across studies, specifically a sequential elevation of interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and IL-10. Surprisingly, acute EtOH did not significantly alter this course of cytokine fluctuations in the hippocampus. However, a history of adolescent CIE showed drastic effects on multiple neuroimmune analytes when rechallenged with EtOH as adults. Rats with a history of adolescent EtOH displayed a severely blunted neuroimmune response in adulthood, evinced by suppressed IL-1β, IL-10, and TNF-α. CONCLUSIONS Together, these findings provide a methodological framework for assessment of cytokine release patterns, their modulation by EtOH, and the long-lasting changes to neuroimmune reactivity evoked by a history of adolescent CIE.
Collapse
Affiliation(s)
- Anny Gano
- Medical University of South Carolina, Charleston Alcohol Research Center, Charleston, SC, USA
| | - Andrew S. Vore
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000
| | - Maryam Sammakia
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000
| |
Collapse
|
15
|
McCaul ME, Roach D, Hasin DS, Weisner C, Chang G, Sinha R. Alcohol and Women: A Brief Overview. Alcohol Clin Exp Res 2019; 43:774-779. [PMID: 30779446 DOI: 10.1111/acer.13985] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Mary E McCaul
- Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Deidra Roach
- National Institute on Alcohol Abuse and Alcoholism , Rockville, Maryland
| | - Deborah S Hasin
- New York State Psychiatric Institute (NYSPI) , Columbia University, New York, New York
| | | | - Grace Chang
- Department of Psychiatry , Harvard Medical School, Boston, Massachusetts
| | - Rajita Sinha
- Department of Psychiatry, Neuroscience and Child Study , Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
16
|
Ruszkiewicz JA, Miranda-Vizuete A, Tinkov AA, Skalnaya MG, Skalny AV, Tsatsakis A, Aschner M. Sex-Specific Differences in Redox Homeostasis in Brain Norm and Disease. J Mol Neurosci 2019; 67:312-342. [DOI: 10.1007/s12031-018-1241-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022]
|
17
|
Finn DA, Hashimoto JG, Cozzoli DK, Helms ML, Nipper MA, Kaufman MN, Wiren KM, Guizzetti M. Binge Ethanol Drinking Produces Sexually Divergent and Distinct Changes in Nucleus Accumbens Signaling Cascades and Pathways in Adult C57BL/6J Mice. Front Genet 2018; 9:325. [PMID: 30250478 PMCID: PMC6139464 DOI: 10.3389/fgene.2018.00325] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/30/2018] [Indexed: 12/20/2022] Open
Abstract
We previously determined that repeated binge ethanol drinking produced sex differences in the regulation of signaling downstream of Group 1 metabotropic glutamate receptors in the nucleus accumbens (NAc) of adult C57BL/6J mice. The purpose of the present study was to characterize RNA expression differences in the NAc of adult male and female C57BL/6J mice following 7 binge ethanol drinking sessions, when compared with controls consuming water. This binge drinking procedure produced high intakes (average >2.2 g/kg/30 min) and blood ethanol concentrations (average >1.3 mg/ml). Mice were euthanized at 24 h after the 7th binge session, and focused qPCR array analysis was employed on NAc tissue to quantify expression levels of 384 genes in a customized Mouse Mood Disorder array, with a focus on glutamatergic signaling (3 arrays/group). We identified significant regulation of 50 genes in male mice and 70 genes in female mice after 7 ethanol binges. Notably, 14 genes were regulated in both males and females, representing common targets to binge ethanol drinking. However, expression of 10 of these 14 genes was strongly dimorphic (e.g., opposite regulation for genes such as Crhr2, Fos, Nos1, and Star), and only 4 of the 14 genes were regulated in the same direction (Drd5, Grm4, Ranbp9, and Reln). Interestingly, the top 30 regulated genes by binge ethanol drinking for each sex differed markedly in the male and female mice, and this divergent neuroadaptive response in the NAc could result in dysregulation of distinct biological pathways between the sexes. Characterization of the expression differences with Ingenuity Pathway Analysis was used to identify Canonical Pathways, Upstream Regulators, and significant Biological Functions. Expression differences suggested that hormone signaling and immune function were altered by binge drinking in female mice, whereas neurotransmitter metabolism was a central target of binge ethanol drinking in male mice. Thus, these results indicate that the transcriptional response to repeated binge ethanol drinking was strongly influenced by sex, and they emphasize the importance of considering sex in the development of potential pharmacotherapeutic targets for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Deborah A Finn
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Research, VA Portland Health Care System, Portland, OR, United States
| | - Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Research, VA Portland Health Care System, Portland, OR, United States
| | - Debra K Cozzoli
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Melinda L Helms
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Research, VA Portland Health Care System, Portland, OR, United States
| | - Michelle A Nipper
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Research, VA Portland Health Care System, Portland, OR, United States
| | - Moriah N Kaufman
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Kristine M Wiren
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Research, VA Portland Health Care System, Portland, OR, United States
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Research, VA Portland Health Care System, Portland, OR, United States
| |
Collapse
|
18
|
Torres-Rojas C, Jones BC. Sex Differences in Neurotoxicogenetics. Front Genet 2018; 9:196. [PMID: 29922331 PMCID: PMC5996082 DOI: 10.3389/fgene.2018.00196] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/15/2018] [Indexed: 12/02/2022] Open
Abstract
A major development in biomedical research is the recognition that the sex of an individual plays a key role in susceptibility, treatment, and outcomes of most diseases. In this contribution, we present evidence that sex is also important in the toxicity of many environmental toxicants and contributes to the effect of genetics. Thus, individual differences in response to toxicants includes genetic makeup, the environment and sex; in fact, sex differences may be considered a part of genetic constitution. In this review, we present evidence for sex contribution to susceptibility for a number of toxicants.
Collapse
Affiliation(s)
- Carolina Torres-Rojas
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Byron C Jones
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
19
|
Wardzala C, Murchison C, Loftis JM, Schenning KJ, Mattek N, Woltjer R, Kaye J, Quinn JF, Wilhelm CJ. Sex differences in the association of alcohol with cognitive decline and brain pathology in a cohort of octogenarians. Psychopharmacology (Berl) 2018; 235:761-770. [PMID: 29185023 PMCID: PMC5839119 DOI: 10.1007/s00213-017-4791-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 11/14/2017] [Indexed: 10/18/2022]
Abstract
RATIONALE The beneficial effects of moderate alcohol may differ in aging men versus women. OBJECTIVES Cognitive and functional decline and neuropathology were investigated in a cohort of aging men and women with diverse alcohol histories. METHODS Non-demented (Clinical Dementia Rating (CDR) of ≤ 0.5 and a Mini-Mental State Examination (MMSE) score of > 24), autonomously living participants were tracked in longitudinal aging studies to examine self-report and objective tests of rates of decline in a cohort (n = 486) of octogenarians. Neurofibrillary tangles (NFTs; Braak stage) and neuritic plaques (NPs) were staged at autopsy in a subset of participants (n = 149) using current standard neuropathologic diagnostic criteria. RESULTS Moderate drinking men had an attenuated rate of decline compared to rare/never drinkers and women on the MMSE and CDR sum of boxes. In contrast, moderate drinking women had a reduced rate of decline only in the Logical Memory Delayed Recall Test (LMDR) compared to rare/never drinkers and men. Moderate alcohol consumption was associated with a reduction in the incidence of advanced (stages 5-6) Braak NFT stage in men (p < 0.05), with no effect in women. CONCLUSIONS In this cohort, men experienced a broader range of beneficial effects associated with alcohol. Alcohol's effects may differ in men and women in important ways that suggest a narrower beneficial window.
Collapse
|
20
|
Hilderbrand ER, Lasek AW. Estradiol enhances ethanol reward in female mice through activation of ERα and ERβ. Horm Behav 2018; 98:159-164. [PMID: 29305887 PMCID: PMC5829002 DOI: 10.1016/j.yhbeh.2018.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/27/2017] [Accepted: 01/01/2018] [Indexed: 11/16/2022]
Abstract
Alcohol use disorder (AUD) manifests differently in men and women, but little is known about sex differences in the brain's response to ethanol. It is known that the steroid hormone 17β-estradiol (E2) regulates voluntary ethanol consumption in female rodents. However, the role of E2 as a regulator of ethanol reward has not been investigated. In this study, we tested for the effects of E2 and agonists selective for the classical estrogen receptors, ERα and ERβ, on ethanol reward in ovariectomized (OVX) mice using the conditioned place preference (CPP) test. E2 enhanced ethanol CPP and, while specific activation of either receptor alone had no effect, co-activation of ERα and ERβ also enhanced ethanol CPP, suggesting that E2 enhances ethanol reward in female mice through actions at both ERα and ERβ. These results have implications for sex differences in the development of AUD, suggesting that women may find ethanol more rewarding than men because of higher circulating E2 levels.
Collapse
Affiliation(s)
- Elisa R Hilderbrand
- Graduate Program in Neuroscience, University of Illinois at Chicago, 1601 West Taylor Street, MC 912, Chicago, IL 60612, United States; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, 1601 West Taylor Street, MC 912, Chicago, IL 60612, United States
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, 1601 West Taylor Street, MC 912, Chicago, IL 60612, United States.
| |
Collapse
|
21
|
Satta R, Hilderbrand ER, Lasek AW. Ovarian Hormones Contribute to High Levels of Binge-Like Drinking by Female Mice. Alcohol Clin Exp Res 2018; 42:286-294. [PMID: 29205408 DOI: 10.1111/acer.13571] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recently, the incidence of binge drinking by women has increased. Binge drinking is detrimental to women's health, yet the biological mechanisms that promote excessive drinking by women are not well understood. One method of assessing binge-like ethanol (EtOH) consumption in mice is the drinking in the dark (DID) test, in which mice drink sufficient EtOH to achieve intoxication. In this study, we directly compared male, female, and ovariectomized (OVX) mice for DID and tested whether 17β-estradiol (E2) contributes to DID. We also measured whether DID varies throughout the estrous cycle and whether repeated intermittent DID impacts the estrous cycle. METHODS Male, female, and OVX C57BL/6J mice were tested for DID for 2 hours per day on days 1 to 3 and for 4 hours on day 4 using a single bottle containing 20% EtOH. To measure the effects of E2 on DID, OVX mice were treated with estradiol benzoate (EB) or vehicle daily starting 2 weeks prior to the drinking test and throughout the DID procedure. In a separate group of experiments, EtOH consumption and estrous cycle phase were measured in freely cycling mice that were drinking EtOH or water 5 days per week for 2 or 6 weeks. RESULTS Female mice consumed more EtOH than male and OVX mice. Treatment with EB increased EtOH consumption by OVX mice compared with vehicle-treated controls. However, EtOH intake did not vary across the estrous cycle, nor did long-term DID alter the estrous cycle. CONCLUSIONS These results demonstrate that ovarian hormones, specifically E2, contribute to increased EtOH consumption by female mice in the DID test. Although ovarian hormones contribute to this behavior, EtOH consumption is not affected by estrous cycle phase in freely cycling mice. This study provides a framework for understanding the factors that contribute to binge drinking in females.
Collapse
Affiliation(s)
- Rosalba Satta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| | - Elisa R Hilderbrand
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois.,Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, Illinois
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
22
|
Tapia-Rojas C, Mira RG, Torres AK, Jara C, Pérez MJ, Vergara EH, Cerpa W, Quintanilla RA. Alcohol consumption during adolescence: A link between mitochondrial damage and ethanol brain intoxication. Birth Defects Res 2017; 109:1623-1639. [DOI: 10.1002/bdr2.1172] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/31/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Cheril Tapia-Rojas
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Rodrigo G. Mira
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago 8331150 Chile
| | - Angie K. Torres
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Claudia Jara
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - María José Pérez
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Erick H. Vergara
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Waldo Cerpa
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago 8331150 Chile
| | - Rodrigo A. Quintanilla
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| |
Collapse
|
23
|
Lac A, Donaldson CD. Comparing the predictive validity of the four-factor and five-factor (bifactor) measurement structures of the drinking motives questionnaire. Drug Alcohol Depend 2017; 181:108-115. [PMID: 29040825 PMCID: PMC5683928 DOI: 10.1016/j.drugalcdep.2017.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The Drinking Motives Questionnaire (DMQ-R) is the most widely administered instrument to assess reasons for consuming alcohol and is conventionally premised on a four-factor structure. Recent research instead reveals that a bifactor measurement model of five motive factors (one general and four specific) represents a superior psychometric embodiment of the scale. The current study evaluated and compared the predictive validity of the four-factor and five-factor models of drinking motives in longitudinally explaining alcohol use and problems. METHODS Adult participants (N=413; age range=18-79 years) completed measures of drinking motives (Time 1) and alcohol use and problems one month later (Time 2). RESULTS Confirmatory factor analyses corroborated the four-factor (social, enhancement, conformity, and coping motives) and five-factor (each item double loading on general motives and a specific motives factor) measurement structures, but the latter rendered stronger fit indices. Structural equation models revealed that lower social motives, higher enhancement motives, and higher coping motives prospectively contributed to alcohol use. Furthermore, lower social motives, higher conformity motives, higher coping motives, and greater alcohol use contributed to alcohol problems. DISCUSSION The same set of paths emerged as significantly predictive in both models, but general motives additionally explained alcohol use and problems in the five-factor model. The incremental contribution of general motives (beyond the specific motives) on alcohol intake and detrimental consequences supports the predictive validity of the drinking reasons paradigm embodied by the inclusion of a global factor.
Collapse
Affiliation(s)
- Andrew Lac
- Department of Psychology, University of Colorado at Colorado Springs, 1420 Austin Bluffs Pkwy, Colorado Springs, CO 80918, USA.
| | - Candice D Donaldson
- Department of Psychology, Claremont Graduate University, 150 E. 10th St., Claremont, CA 91711, USA.
| |
Collapse
|
24
|
Hashimoto JG, Gavin DP, Wiren KM, Crabbe JC, Guizzetti M. Prefrontal cortex expression of chromatin modifier genes in male WSP and WSR mice changes across ethanol dependence, withdrawal, and abstinence. Alcohol 2017; 60:83-94. [PMID: 28433423 PMCID: PMC5497775 DOI: 10.1016/j.alcohol.2017.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 12/20/2022]
Abstract
Alcohol-use disorder (AUD) is a relapsing disorder associated with excessive ethanol consumption. Recent studies support the involvement of epigenetic mechanisms in the development of AUD. Studies carried out so far have focused on a few specific epigenetic modifications. The goal of this project was to investigate gene expression changes of epigenetic regulators that mediate a broad array of chromatin modifications after chronic alcohol exposure, chronic alcohol exposure followed by 8 h withdrawal, and chronic alcohol exposure followed by 21 days of abstinence in Withdrawal-Resistant (WSR) and Withdrawal Seizure-Prone (WSP) selected mouse lines. We found that chronic vapor exposure to highly intoxicating levels of ethanol alters the expression of several chromatin remodeling genes measured by quantitative PCR array analyses. The identified effects were independent of selected lines, which, however, displayed baseline differences in epigenetic gene expression. We reported dysregulation in the expression of genes involved in histone acetylation, deacetylation, lysine and arginine methylation and ubiquitinationhylation during chronic ethanol exposure and withdrawal, but not after 21 days of abstinence. Ethanol-induced changes are consistent with decreased histone acetylation and with decreased deposition of the permissive ubiquitination mark H2BK120ub, associated with reduced transcription. On the other hand, ethanol-induced changes in the expression of genes involved in histone lysine methylation are consistent with increased transcription. The net result of these modifications on gene expression is likely to depend on the combination of the specific histone tail modifications present at a given time on a given promoter. Since alcohol does not modulate gene expression unidirectionally, it is not surprising that alcohol does not unidirectionally alter chromatin structure toward a closed or open state, as suggested by the results of this study.
Collapse
Affiliation(s)
- Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, OR, 97239, United States; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, United States
| | - David P Gavin
- Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue (M/C 151), Chicago, IL, 60612, United States; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor St., Chicago, IL, 60612, United States
| | - Kristine M Wiren
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, OR, 97239, United States; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, United States
| | - John C Crabbe
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, OR, 97239, United States; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, United States
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, OR, 97239, United States; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, United States.
| |
Collapse
|
25
|
Shimizu J, Okazaki S, Nagoya S, Takahashi N, Kanaya K, Mizuo K, Hyodoh H, Watanabe S, Yamashita T. Susceptibility of Males, but Not Females to Developing Femoral Head Osteonecrosis in Response to Alcohol Consumption. PLoS One 2016; 11:e0165490. [PMID: 27788269 PMCID: PMC5082908 DOI: 10.1371/journal.pone.0165490] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/12/2016] [Indexed: 01/25/2023] Open
Abstract
Background We previously reported that ethanol-containing liquid diet feeding induces osteonecrosis of the femoral head in male rats. Also, it was reported that a large amount of consumed ethanol and a long-term history of drinking were risk factors for osteonecrosis of the femoral head, and that the frequency of alcohol-induced osteonecrosis of the femoral head in males was much greater than in females. The higher incidence of alcohol-induced osteonecrosis of the femoral head could be due to either higher prevalence of alcohol drinking in males or due to their potential higher sensitivity to alcohol. The aim of the study is to investigate the influence of alcohol consumption and drinking period on the development of osteonecrosis of the femoral head in rats of both sex. Methods All the experimental male rats were allocated to the male one-month ethanol drinking group (M1). Female rats were randomly divided into the female one- to five-months ethanol drinking groups (F1-5). All rats were fed a Lieber-DeCarli liquid diet containing 5% ethanol for one to five months. Results One-month feeding with the ethanol-containing liquid diet resulted in the development of osteonecrosis of the femoral head in seven of twenty in the M1 group, but none in the F1 group, although the mean intake of ethanol per body weight in the M1 group was significantly lower than that in the F1 group. Furthermore, long drinking periods with a large amount of ethanol intake in the F2-5 groups did not induce osteonecrosis of the femoral head. Conclusion The present study shows that lower alcohol consumption over short periods of time that were sufficient to induce osteonecrosis of the femoral head in males had no effect on females. Even with greater alcohol consumption and longer duration, females did not develop osteonecrosis of the femoral head. Therefore, unknown factors related to sex must be responsible for the development of this condition.
Collapse
Affiliation(s)
- Junya Shimizu
- Department of Orthopedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Legal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shunichiro Okazaki
- Department of Legal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Musculoskeletal Biomechanics and Surgical Development, Sapporo Medical University, Sapporo, Japan
- * E-mail:
| | - Satoshi Nagoya
- Department of Musculoskeletal Biomechanics and Surgical Development, Sapporo Medical University, Sapporo, Japan
| | - Nobuyuki Takahashi
- Department of Orthopedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Legal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kumiko Kanaya
- Department of Orthopedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keisuke Mizuo
- Department of Legal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hideki Hyodoh
- Department of Legal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Satoshi Watanabe
- Department of Legal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Yamashita
- Department of Orthopedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
26
|
Jung ME, Metzger DB. A sex difference in oxidative stress and behavioral suppression induced by ethanol withdrawal in rats. Behav Brain Res 2016; 314:199-214. [PMID: 27503149 DOI: 10.1016/j.bbr.2016.07.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/25/2016] [Accepted: 07/30/2016] [Indexed: 12/19/2022]
Abstract
Ethanol withdrawal (EW) is referred to the abrupt termination of long-term heavy drinking, and provokes oxidative brain damage. Here, we investigated whether the cerebellum and hippocampus of female rats are less affected by prooxidant EW than male rats due to the antioxidant effect of 17β-estradiol (E2). Female and male rats received a four-week ethanol diet and three-week withdrawal per cycle for two cycles. Some female rats were ovariectomized with E2 or antioxidant (Vitamin E+Co-Q10) treatment. Measurements were cerebellum (Rotarod) and hippocampus (water-maze)-related behaviors, oxidative markers (O2(-), malondialdehyde, protein carbonyls), mitochondrial membrane swelling, and a key mitochondrial enzyme, cytochrome c oxidase (CcO). Separately, HT22 (hippocampal) cells were subjected to ethanol-exposure and withdrawal for two cycles to assess the effect of a CcO inhibitor on E2's protection for mitochondrial respiration and cell viability. Ethanol-withdrawn female rats showed a smaller increase in oxidative markers in cerebellum and hippocampus than male rats, and E2 treatment decreased the oxidative markers. Compared to male counterparts, ethanol-withdrawn female rats showed better Rotarod but poorer water-maze performance, accompanied by more severe mitochondrial membrane swelling and CcO suppression in hippocampus. E2 or antioxidant treatment improved Rotarod but not water-maze performance. In the presence of a CcO inhibitor, E2 treatment failed to protect mitochondrial respiration and cell viability from EW. These data suggest that antioxidant E2 contributes to smaller oxidative stress in ethanol-withdrawn female than male rats. They also suggest that EW-induced severe mitochondrial damage in hippocampus may blunt E2's antioxidant protection for hippocampus-related behavior.
Collapse
Affiliation(s)
- Marianna E Jung
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd., Fort Worth, TX 76107-2699, USA.
| | - Daniel B Metzger
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd., Fort Worth, TX 76107-2699, USA
| |
Collapse
|
27
|
Hashimoto JG, Wiren KM, Wilhelm CJ. A neurotoxic alcohol exposure paradigm does not induce hepatic encephalopathy. Neurotoxicol Teratol 2016; 56:35-40. [PMID: 27268733 DOI: 10.1016/j.ntt.2016.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 02/07/2023]
Abstract
Alcohol abuse is associated with neurological dysfunction, brain morphological deficits and frank neurotoxicity. Although these disruptions may be a secondary effect due to hepatic encephalopathy, no clear evidence of causality is available. This study examined whether a 72h period of alcohol intoxication known to induce physical dependence, followed by a single withdrawal, was sufficient to induce signs of hepatic encephalopathy in male and female mice. Animals were continuously intoxicated via alcohol vapor inhalation, a procedure previously shown to induce significant neurotoxicity in female mice. At peak synchronized withdrawal (8h following the end of alcohol exposure), blood samples were taken and levels of several liver-regulated markers and brain swelling were characterized. Glutathione levels were also determined in the medial frontal cortex (mFC) and hippocampus. Results revealed elevated levels of cholesterol, albumin, alkaline phosphatase (ALP), alanine aminotransferase (ALT) and decreased levels of blood urea nitrogen and total bilirubin in alcohol-exposed male and female groups compared to controls. Brain water weight was not affected by alcohol exposure, though males tended to have slightly more water weight overall. Alcohol exposure led to reductions in tissue levels of glutathione in both the hippocampus and mFC which may indicate increased oxidative stress. Combined, these results suggest that hepatic encephalopathy does not appear to play a significant role in the neurotoxicity observed following alcohol exposure in this model.
Collapse
Affiliation(s)
- Joel G Hashimoto
- Research & Development Service, VA Portland Health Care System, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Kristine M Wiren
- Research & Development Service, VA Portland Health Care System, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Clare J Wilhelm
- Research & Development Service, VA Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
28
|
Carroll ME, Smethells JR. Sex Differences in Behavioral Dyscontrol: Role in Drug Addiction and Novel Treatments. Front Psychiatry 2016; 6:175. [PMID: 26903885 PMCID: PMC4745113 DOI: 10.3389/fpsyt.2015.00175] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 11/30/2015] [Indexed: 11/22/2022] Open
Abstract
The purpose of this review is to discuss recent findings related to sex differences in behavioral dyscontrol that lead to drug addiction, and clinical implications for humans are discussed. This review includes research conducted in animals and humans that reveals fundamental aspects of behavioral dyscontrol. The importance of sex differences in aspects of behavioral dyscontrol, such as impulsivity and compulsivity, is discussed as major determinants of drug addiction. Behavioral dyscontrol during adolescence is also an important consideration, as this is the time of onset for drug addiction. These vulnerability factors additively increase drug-abuse vulnerability, and they are integral aspects of addiction that covary and interact with sex differences. Sex differences in treatments for drug addiction are also reviewed in terms of their ability to modify the behavioral dyscontrol that underlies addictive behavior. Customized treatments to reduce behavioral dyscontrol are discussed, such as (1) using natural consequences such as non-drug rewards (e.g., exercise) to maintain abstinence, or using punishment as a consequence for drug use, (2) targeting factors that underlie behavioral dyscontrol, such as impulsivity or anxiety, by repurposing medications to relieve these underlying conditions, and (3) combining two or more novel behavioral or pharmacological treatments to produce additive reductions in drug seeking. Recent published work has indicated that factors contributing to behavioral dyscontrol are an important target for advancing our knowledge on the etiology of drug abuse, intervening with the drug addiction process and developing novel treatments.
Collapse
Affiliation(s)
| | - John R. Smethells
- Program in PharmacoNeuroImmunology, Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
29
|
Functional regulation of PI3K-associated signaling in the accumbens by binge alcohol drinking in male but not female mice. Neuropharmacology 2016; 105:164-174. [PMID: 26773198 DOI: 10.1016/j.neuropharm.2016.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/07/2015] [Accepted: 01/05/2016] [Indexed: 01/25/2023]
Abstract
It is well established that binge alcohol consumption produces alterations in Group 1 metabotropic glutamate receptors (mGlus) and related signaling cascades in the nucleus accumbens (NAC) of adult male mice, but female and adolescent mice have not been examined. Thus, the first set of studies determined whether repeated binge alcohol consumption produced similar alterations in protein and mRNA levels of Group 1 mGlu-associated signaling molecules in the NAC of male and female adult and adolescent mice. The adult (9 weeks) and adolescent (4 weeks) C57BL/6J mice were exposed to 7 binge alcohol sessions every 3rd day while controls drank water. Repeated binge alcohol consumption produced sexually divergent changes in protein levels and mRNA expression for Group 1 mGlus and downstream signaling molecules in the NAC, but there was no effect of age. Binge alcohol intake decreased mGlu5 levels in females, whereas it decreased indices of phosphoinositide 3-kinase (PI3K), mammalian target of rapamycin (mTOR), 4E-binding protein 1, and p70 ribosomal protein S6 kinase in males. Expression of genes encoding mGlu1, mGlu5, the NR2A subunit of the NMDA receptor, and Homer2 were all decreased by binge alcohol consumption in males, while females were relatively resistant (only phosphoinositide-dependent protein kinase 1 was decreased). The functional implication of these differences was investigated in a separate study by inhibiting mTOR in the NAC (via infusions of rapamycin) before binge drinking sessions. Rapamycin (50 and 100 ng/side) significantly decreased binge alcohol consumption in males, while consumption in females was unaffected. Altogether these results highlight that mTOR signaling in the NAC was necessary to maintain binge alcohol consumption only in male mice and that binge drinking recruits sexually divergent signaling cascades downstream of PI3K and presumably, Group 1 mGlus. Importantly, these findings emphasize that sex should be considered in the development of potential pharmacotherapeutic targets.
Collapse
|
30
|
Wilhelm CJ, Hashimoto JG, Roberts ML, Bloom SH, Andrew MR, Wiren KM. Astrocyte Dysfunction Induced by Alcohol in Females but Not Males. Brain Pathol 2015; 26:433-51. [PMID: 26088166 DOI: 10.1111/bpa.12276] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/10/2015] [Indexed: 01/08/2023] Open
Abstract
Chronic alcohol abuse is associated with brain damage in a sex-specific fashion, but the mechanisms involved are poorly described and remain controversial. Previous results have suggested that astrocyte gene expression is influenced by ethanol intoxication and during abstinence in vivo. Here, bioinformatic analysis of astrocyte-enriched ethanol-regulated genes in vivo revealed ubiquitin pathways as an ethanol target, but with sexually dimorphic cytokine signaling and changes associated with brain aging in females and not males. Consistent with this result, astrocyte activation was observed after exposure in female but not male animals, with reduced S100β levels in the anterior cingulate cortex and increased GFAP(+) cells in the hippocampus. In primary culture, the direct effects of chronic ethanol exposure followed by recovery on sex-specific astrocyte function were examined. Male astrocyte responses were consistent with astrocyte deactivation with reduced GFAP expression during ethanol exposure. In contrast, female astrocytes exhibited increased expression of Tnf, reduced expression of the neuroprotective cytokine Tgfb1, disrupted bioenergetics and reduced excitatory amino acid uptake following exposure or recovery. These results indicate widespread astrocyte dysfunction in ethanol-exposed females and suggest a mechanism that may underlie increased vulnerability to ethanol-induced neurotoxicity in females.
Collapse
Affiliation(s)
- Clare J Wilhelm
- VA Portland Health Care System, Portland, OR.,Department of Psychiatry, Oregon Health & Science University, Portland, OR
| | - Joel G Hashimoto
- VA Portland Health Care System, Portland, OR.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | | | | | - Melissa R Andrew
- Cincinnati Center for Growth Disorders, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kristine M Wiren
- VA Portland Health Care System, Portland, OR.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| |
Collapse
|
31
|
Wilhelm CJ, Hashimoto JG, Roberts ML, Bloom SH, Beard DK, Wiren KM. Females uniquely vulnerable to alcohol-induced neurotoxicity show altered glucocorticoid signaling. Brain Res 2015; 1601:102-16. [PMID: 25601008 DOI: 10.1016/j.brainres.2015.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/28/2014] [Accepted: 01/01/2015] [Indexed: 12/29/2022]
Abstract
Women are more sensitive to the harmful effects of alcohol (EtOH) abuse than men, yet the underlying mechanisms remain poorly understood. Previous gene expression analysis of the medial prefrontal cortex (mPFC) following a chronic intoxication paradigm using continuous 72 h vapor inhalation found that females, but not males, exhibit an inflammatory response at peak withdrawal that is associated with cell damage. Given that glucocorticoids can function as anti-inflammatories, are known to increase with EtOH exposure, and influence neurotoxicity, we hypothesized that males and females may exhibit an altered corticosterone (CORT) response following chronic intoxication. Analysis of serum CORT levels revealed the expected increase during withdrawal with no difference between males and females, while control males but not females exhibited higher CORT concentrations than naive animals. Glucocorticoid signaling characterized using focused qPCR arrays identified a sexually dimorphic response in the mPFC during withdrawal, particularly among astrocyte-enriched genes. These genes include aquaporin-1 (Aqp1), sphingosine kinase 1 (Sphk1) and connective tissue growth factor (Ctgf); genes associated with inflammatory signaling, and tissue damage and repair. Bioinformatic analysis also revealed activation of inflammatory signaling and cell death pathways in females. Confirmation studies showed that female mice exhibited significant neuronal degeneration within the anterior cingulate cortex (ACC). By contrast, EtOH exposure lead to a significant reduction in cell death in males. Thus, distinct glucocorticoid signaling pathways are associated with sexually dimorphic neurotoxicity, suggesting one mechanism by which EtOH-exposed females are particularly vulnerable to the damaging effects of alcohol in the CNS.
Collapse
Affiliation(s)
- Clare J Wilhelm
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Joel G Hashimoto
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | - Kristine M Wiren
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
32
|
Wilhelm CJ, Hashimoto JG, Roberts ML, Sonmez MK, Wiren KM. Understanding the addiction cycle: a complex biology with distinct contributions of genotype vs. sex at each stage. Neuroscience 2014; 279:168-86. [PMID: 25194791 PMCID: PMC4224688 DOI: 10.1016/j.neuroscience.2014.08.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 02/07/2023]
Abstract
Ethanol abuse can lead to addiction, brain damage and premature death. The cycle of alcohol addiction has been described as a composite consisting of three stages: intoxication, withdrawal and craving/abstinence. There is evidence for contributions of both genotype and sex to alcoholism, but an understanding of the biological underpinnings is limited. Utilizing both sexes of genetic animal models with highly divergent alcohol withdrawal severity, Withdrawal Seizure-Resistant (WSR) and Withdrawal Seizure-Prone (WSP) mice, the distinct contributions of genotype/phenotype and of sex during addiction stages on neuroadaptation were characterized. Transcriptional profiling was performed to identify expression changes as a consequence of chronic intoxication in the medial prefrontal cortex. Significant expression differences were identified on a single platform and tracked over a behaviorally relevant time course that covered each stage of alcohol addiction; i.e., after chronic intoxication, during peak withdrawal, and after a defined period of abstinence. Females were more sensitive to ethanol with higher fold expression differences. Bioinformatics showed a strong effect of sex on the data structure of expression profiles during chronic intoxication and at peak withdrawal irrespective of genetic background. However, during abstinence, differences were observed instead between the lines/phenotypes irrespective of sex. Confirmation of identified pathways showed distinct inflammatory signaling following intoxication at peak withdrawal, with a pro-inflammatory phenotype in females but overall suppression of immune signaling in males. Combined, these results suggest that each stage of the addiction cycle is influenced differentially by sex vs. genetic background and support the development of stage- and sex-specific therapies for alcohol withdrawal and the maintenance of sobriety.
Collapse
Affiliation(s)
- C J Wilhelm
- Department of Veterans Affairs Medical Center, Portland, OR 97239, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR 97239, USA.
| | - J G Hashimoto
- Department of Veterans Affairs Medical Center, Portland, OR 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - M L Roberts
- Department of Veterans Affairs Medical Center, Portland, OR 97239, USA
| | - M K Sonmez
- Center For Spoken Language Understanding, Oregon Health & Science University, Portland, OR 97239, USA
| | - K M Wiren
- Department of Veterans Affairs Medical Center, Portland, OR 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
33
|
Fattore L, Melis M, Fadda P, Fratta W. Sex differences in addictive disorders. Front Neuroendocrinol 2014; 35:272-84. [PMID: 24769267 DOI: 10.1016/j.yfrne.2014.04.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 04/04/2014] [Accepted: 04/04/2014] [Indexed: 12/16/2022]
Abstract
Gender-dependent differences in the rate of initiation and frequency of misuse of addicting drugs have been widely described. Yet, men and women also differ in their propensity to become addicted to other rewarding stimuli (e.g., sex, food) or activities (e.g., gambling, exercising). The goal of the present review is to summarize current evidence for gender differences not only in drug addiction, but also in other forms of addictive behaviours. Thus, we first reviewed studies showing gender-dependent differences in drug addiction, food addiction, compulsive sexual activity, pathological gambling, Internet addiction and physical exercise addiction. Potential risk factors and underlying brain mechanisms are also examined, with particular emphasis given to the role of sex hormones in modulating addictive behaviours. Investigations on factors allowing the pursuit of non-drug rewards to become pathological in men and women are crucial for designing gender-appropriate treatments of both substance and non-substance addictions.
Collapse
Affiliation(s)
- Liana Fattore
- Institute of Neuroscience - Cagliari National Research Council of Italy, Cittadella Universitaria di Monserrato, Italy; Centre of Excellence "Neurobiology of Dependence", University of Cagliari, Monserrato, Italy.
| | - Miriam Melis
- Centre of Excellence "Neurobiology of Dependence", University of Cagliari, Monserrato, Italy; Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, Cittadella Universitaria di Monserrato, University of Cagliari, Monserrato, Italy
| | - Paola Fadda
- Centre of Excellence "Neurobiology of Dependence", University of Cagliari, Monserrato, Italy; Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, Cittadella Universitaria di Monserrato, University of Cagliari, Monserrato, Italy; National Institute of Neuroscience (INN), University of Cagliari, Italy
| | - Walter Fratta
- Centre of Excellence "Neurobiology of Dependence", University of Cagliari, Monserrato, Italy; Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, Cittadella Universitaria di Monserrato, University of Cagliari, Monserrato, Italy; National Institute of Neuroscience (INN), University of Cagliari, Italy
| |
Collapse
|
34
|
Chitty KM, Kaur M, Lagopoulos J, Hickie IB, Hermens DF. Risky alcohol use predicts temporal mismatch negativity impairments in young people with bipolar disorder. Biol Psychol 2014; 99:60-8. [PMID: 24594113 DOI: 10.1016/j.biopsycho.2014.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 01/22/2014] [Accepted: 02/23/2014] [Indexed: 01/08/2023]
Abstract
Alcohol misuse in bipolar disorder (BD) has a negative impact on illness progression. The NMDA/glutamatergic system is implicated in BD pathophysiology and is critically involved in the effects of alcohol on the brain. Mismatch negativity (MMN) is purported to reflect NMDA receptor output, providing a measure for investigating this association. Forty-two patients and 34 controls (16-30 years) were split into low and high-risk drinkers (based on the Alcohol Use Disorders Identification Test) and underwent a two-tone passive auditory oddball, duration deviant MMN paradigm. Multiple regression models revealed risky drinking and BD diagnosis were predictors of impaired temporal MMN. Potentially reflecting an additive effect of alcohol on a perturbed NMDA/glutamatergic system in BD, these findings highlight alcohol as both a modifiable risk factor of neurobiological impairments and as a potential confounder in MMN studies. Given the increasing use of glutamatergic agents for BD treatment, this finding is important clinically.
Collapse
Affiliation(s)
- Kate M Chitty
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia.
| | - Manreena Kaur
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Jim Lagopoulos
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Ian B Hickie
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Daniel F Hermens
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| |
Collapse
|
35
|
Houston RJ, Derrick JL, Leonard KE, Testa M, Quigley BM, Kubiak A. Effects of heavy drinking on executive cognitive functioning in a community sample. Addict Behav 2014; 39:345-9. [PMID: 24459697 DOI: 10.1016/j.addbeh.2013.09.032] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Deficits in several aspects of executive cognitive functioning (ECF) have been consistently associated with alcohol use disorders. Most of this research, however, has been conducted in alcohol dependent patient samples. A handful of recent studies, primarily in college students, have also reported similar deficits, but little is known about the effects of heavy drinking in adult, non-patient men and women. METHODS A community sample (N = 560) of men and women completed a brief battery of ECF measures including measures of attentional control, cognitive flexibility, working memory and response inhibition.Quantity/frequency of alcohol and illicit drug use in the past year were also assessed. RESULTS Regression analyses indicated that men and women with higher levels of alcohol consumption exhibited greater impairment on several ECF measures, primarily those pertaining to cognitive flexibility and response inhibition. These results remained after controlling for demographic factors such as age, gender, education, and illicit drug use. CONCLUSIONS These findings support and extend prior work documenting the deleterious effects of heavy alcohol consumption on ECF in a community sample and specifically indicate robust effects on cognitive flexibility,psychomotor speed, and response inhibition.
Collapse
|
36
|
An L, Zhang T. Spatial cognition and sexually dimorphic synaptic plasticity balance impairment in rats with chronic prenatal ethanol exposure. Behav Brain Res 2013; 256:564-74. [PMID: 24050890 DOI: 10.1016/j.bbr.2013.09.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 09/03/2013] [Accepted: 09/07/2013] [Indexed: 01/12/2023]
Abstract
Prenatal ethanol exposure can lead to long-lasting impairments in the ability of rats to process spatial information, as well as produce long-lasting deficits in long-term potentiation (LTP), a biological model of learning and memory processing. The present study aimed to examine the sexually dimorphic effects of chronic prenatal ethanol exposure (CPEE) on behavior cognition and synaptic plasticity balance (SPB), and tried to understand a possible mechanism by evaluating the alternation of SPB. The animal model was produced by ethanol exposure throughout gestational period with 4 g/kg bodyweight. Offspring of both male and female were selected and studied on postnatal days 36. Subsequently, the data showed that chronic ethanol exposure resulted in birth weight reduction, losing bodyweight gain, microcephaly and hippocampus weight retardation. In Morris water maze (MWM) test, escape latencies were significantly higher in CPEE-treated rats than that in control ones. They also spent much less time in the target quadrant compared to that of control animals in the probe phase. In addition, it was found that there was a more severe impairment in females than that in males after CPEE treatment. Electrophysiological studies showed that CPEE considerably inhibited hippocampal LTP and facilitated depotentiation in males, while significantly enhanced LTP and suppressed depotentiation in females. A novel index, developed by us, showed that the action of CPEE on SPB was more sensitive in females than that in males, suggesting that it might be an effective index to distinguish the difference of SPB impairment between males and females.
Collapse
Affiliation(s)
- Lei An
- College of Life Sciences, Nankai University, 300071 Tianjin, PR China
| | | |
Collapse
|
37
|
Wiren KM. Males and females are just different: Sexually dimorphic responses to chronic ethanol exposure in hippocampal slice cultures. Neurosci Lett 2013; 550:1-5. [DOI: 10.1016/j.neulet.2013.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 01/07/2023]
|
38
|
Walls SA, Rosenwasser AM, Devaud LL. Sex and regional differences in effects of chronic intermittent ethanol exposure on subsequent excitotoxic challenges in hippocampal slice cultures. Neurosci Lett 2013; 550:6-11. [PMID: 23680460 DOI: 10.1016/j.neulet.2013.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/02/2013] [Accepted: 05/03/2013] [Indexed: 01/07/2023]
Abstract
The organotypic hippocampal slice culture technique was used to study how the effects of repeated ethanol withdrawal might differ between males and females at the cellular level, including potential modulation of subsequent insults. A chronic intermittent ethanol (CIE) exposure paradigm was employed, with 3 days of exposure followed by 24 h withdrawal for 3 cycles. Slices were next exposed to corticosterone (CORT) or pentylenetetrazol (PTZ) for 24 h then imaged for propidium iodide (PI) signal intensities. There were sex-selective responses in the CA1 region and dentate gyrus of the hippocampal slice cultures to treatment with CIE and/or CORT or PTZ. The 50 mM CIE alone generally did not increase the PI signal, but enhanced sensitivity to the toxic effects of CORT (particularly for females) and PTZ (particularly for males). In contrast, 100 mM CIE elicited a toxic response that was greater in females than males, and was exacerbated by exposure to PTZ. These data showed that hippocampal sexual dimorphism influences sensitivity to ethanol and other toxic chemicals even in an immature state. Low-dose CIE may attenuate harm from additional challenges in a hippocampal sex- and region-selective manner. These findings add to the growing evidence of important neurobiological sex differences in responses to chronic ethanol exposure and withdrawal.
Collapse
Affiliation(s)
- Shawn A Walls
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, United States.
| | | | | |
Collapse
|
39
|
Crabbe JC, Kendler KS, Hitzemann RJ. Modeling the diagnostic criteria for alcohol dependence with genetic animal models. Curr Top Behav Neurosci 2013; 13:187-221. [PMID: 21910077 PMCID: PMC3371181 DOI: 10.1007/7854_2011_162] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
A diagnosis of alcohol dependence (AD) using the DSM-IV-R is categorical, based on an individual's manifestation of three or more symptoms from a list of seven. AD risk can be traced to both genetic and environmental sources. Most genetic studies of AD risk implicitly assume that an AD diagnosis represents a single underlying genetic factor. We recently found that the criteria for an AD diagnosis represent three somewhat distinct genetic paths to individual risk. Specifically, heavy use and tolerance versus withdrawal and continued use despite problems reflected separate genetic factors. However, some data suggest that genetic risk for AD is adequately described with a single underlying genetic risk factor. Rodent animal models for alcohol-related phenotypes typically target discrete aspects of the complex human AD diagnosis. Here, we review the literature derived from genetic animal models in an attempt to determine whether they support a single-factor or multiple-factor genetic structure. We conclude that there is modest support in the animal literature that alcohol tolerance and withdrawal reflect distinct genetic risk factors, in agreement with our human data. We suggest areas where more research could clarify this attempt to align the rodent and human data.
Collapse
Affiliation(s)
- John C Crabbe
- Portland Alcohol Research Center, Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | |
Collapse
|
40
|
Contet C. Gene Expression Under the Influence: Transcriptional Profiling of Ethanol in the Brain. CURRENT PSYCHOPHARMACOLOGY 2012; 1:301-314. [PMID: 24078902 PMCID: PMC3783024 DOI: 10.2174/2211556011201040301] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sensitivity to ethanol intoxication, propensity to drink ethanol and vulnerability to develop alcoholism are all influenced by genetic factors. Conversely, exposure to ethanol or subsequent withdrawal produce gene expression changes, which, in combination with environmental variables, may participate in the emergence of compulsive drinking and relapse. The present review offers an integrated perspective on brain gene expression profiling in rodent models of predisposition to differential ethanol sensitivity or consumption, in rats and mice subjected to acute or chronic ethanol exposure, as well as in human alcoholics. The functional categories over-represented among differentially expressed genes suggest that the transcriptional effects of chronic ethanol consumption contribute to the neuroplasticity and neurotoxicity characteristic of alcoholism. Importantly, ethanol produces distinct transcriptional changes within the different brain regions involved in intoxication, reinforcement and addiction. Special emphasis is put on recent profiling studies that have provided some insights into the molecular mechanisms potentially mediating genome-wide regulation of gene expression by ethanol. In particular, current evidence for a role of transcription factors, chromatin remodeling and microRNAs in coordinating the expression of large sets of genes in animals predisposed to excessive ethanol drinking or exposed to protracted abstinence, as well as in human alcoholics, is presented. Finally, studies that have compared ethanol with other drugs of abuse have highlighted common gene expression patterns that may play a central role in drug addiction. The availability of novel technologies and a focus on mechanistic approaches are shaping the future of ethanol transcriptomics.
Collapse
Affiliation(s)
- Candice Contet
- The Scripps Research Institute, Committee on the Neurobiology of Addictive Disorders, La Jolla, CA, USA
| |
Collapse
|
41
|
Melendez RI, McGinty JF, Kalivas PW, Becker HC. Brain region-specific gene expression changes after chronic intermittent ethanol exposure and early withdrawal in C57BL/6J mice. Addict Biol 2012; 17:351-64. [PMID: 21812870 DOI: 10.1111/j.1369-1600.2011.00357.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neuroadaptations that participate in the ontogeny of alcohol dependence are likely a result of altered gene expression in various brain regions. The present study investigated brain region-specific changes in the pattern and magnitude of gene expression immediately following chronic intermittent ethanol (CIE) exposure and 8 hours following final ethanol exposure [i.e. early withdrawal (EWD)]. High-density oligonucleotide microarrays (Affymetrix 430A 2.0, Affymetrix, Santa Clara, CA, USA) and bioinformatics analysis were used to characterize gene expression and function in the prefrontal cortex (PFC), hippocampus (HPC) and nucleus accumbens (NAc) of C57BL/6J mice (Jackson Laboratories, Bar Harbor, ME, USA). Gene expression levels were determined using gene chip robust multi-array average followed by statistical analysis of microarrays and validated by quantitative real-time reverse transcription polymerase chain reaction and Western blot analysis. Results indicated that immediately following CIE exposure, changes in gene expression were strikingly greater in the PFC (284 genes) compared with the HPC (16 genes) and NAc (32 genes). Bioinformatics analysis revealed that most of the transcriptionally responsive genes in the PFC were involved in Ras/MAPK signaling, notch signaling or ubiquitination. In contrast, during EWD, changes in gene expression were greatest in the HPC (139 genes) compared with the PFC (four genes) and NAc (eight genes). The most transcriptionally responsive genes in the HPC were involved in mRNA processing or actin dynamics. Of the few genes detected in the NAc, the most representatives were involved in circadian rhythms. Overall, these findings indicate that brain region-specific and time-dependent neuroadaptive alterations in gene expression play an integral role in the development of alcohol dependence and withdrawal.
Collapse
Affiliation(s)
- Roberto I Melendez
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
42
|
|
43
|
Alfonso-Loeches S, Guerri C. Molecular and behavioral aspects of the actions of alcohol on the adult and developing brain. Crit Rev Clin Lab Sci 2011; 48:19-47. [PMID: 21657944 DOI: 10.3109/10408363.2011.580567] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The brain is one of the major target organs of alcohol actions. Alcohol abuse can lead to alterations in brain structure and functions and, in some cases, to neurodegeneration. Cognitive deficits and alcohol dependence are highly damaging consequences of alcohol abuse. Clinical and experimental studies have demonstrated that the developing brain is particularly vulnerable to alcohol, and that drinking during gestation can lead to a range of physical, learning and behavioral defects (fetal alcohol spectrum disorders), with the most dramatic presentation corresponding to fetal alcohol syndrome. Recent findings also indicate that adolescence is a stage of brain maturation and that heavy drinking at this stage can have a negative impact on brain structure and functions causing important short- and long-term cognitive and behavioral consequences. The effects of alcohol on the brain are not uniform; some brain areas or cell populations are more vulnerable than others. The prefrontal cortex, the hippocampus, the cerebellum, the white matter and glial cells are particularly susceptible to the effects of ethanol. The molecular actions of alcohol on the brain are complex and involve numerous mechanisms and signaling pathways. Some of the mechanisms involved are common for the adult brain and for the developing brain, while others depend on the developmental stage. During brain ontogeny, alcohol causes irreversible alterations to the brain structure. It also impairs several molecular, neurochemical and cellular events taking place during normal brain development, including alterations in both gene expression regulation and the molecules involved in cell-cell interactions, interference with the mitogenic and growth factor response, enhancement of free radical formation and derangements of glial cell functions. However, in both adult and adolescent brains, alcohol damages specific brain areas through mechanisms involving excitotoxicity, free radical formation and neuroinflammatory damage resulting from activation of the innate immune system mediated by TLR4 receptors. Alcohol also acts on specific membrane proteins, such as neurotransmitter receptors (e.g. NMDA, GABA-A), ion channels (e.g. L-type Ca²⁺ channels, GIRKs), and signaling pathways (e.g. PKA and PKC signaling). These effects might underlie the wide variety of behavioral effects induced by ethanol drinking. The neuroadaptive changes affecting neurotransmission systems which are more sensitive to the acute effects of alcohol occur after long-term alcohol consumption. Alcohol-induced maladaptations in the dopaminergic mesolimbic system, abnormal plastic changes in the reward-related brain areas and genetic and epigenetic factors may all contribute to alcohol reinforcement and alcohol addiction. This manuscript reviews the mechanisms by which ethanol impacts the adult and the developing brain, and causes both neural impairments and cognitive and behavioral dysfunctions. The identification and the understanding of the cellular and molecular mechanisms involved in ethanol toxicity might contribute to the development of treatments and/or therapeutic agents that could reduce or eliminate the deleterious effects of alcohol on the brain.
Collapse
|
44
|
Forquer MR, Hashimoto JG, Roberts ML, Wiren KM. Elevated testosterone in females reveals a robust sex difference in altered androgen levels during chronic alcohol withdrawal. Alcohol 2011; 45:161-71. [PMID: 20843636 DOI: 10.1016/j.alcohol.2010.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 07/19/2010] [Accepted: 08/14/2010] [Indexed: 01/11/2023]
Abstract
The endocrine disruption associated with alcohol (ethanol) abuse in both males and females is widely recognized. Ethanol intoxication and withdrawal in males results in significant reductions in androgen levels. Less is known about female alcoholics, and because the changes in testosterone concentrations remain controversial, we systematically characterized changes in sex steroids after chronic ethanol exposure and withdrawal in both sexes. Testosterone and 17β-estradiol concentrations were determined during chronic high intoxication, over a withdrawal time course, and following a period of abstinence using a genetic model of withdrawal vulnerability, the Withdrawal Seizure-Resistant (WSR) and -Prone (WSP) selected lines. In males, testosterone concentrations were significantly lower in intoxicated WSP mice after chronic ethanol exposure, and were dramatically and transiently reduced during the withdrawal period in both WSR and WSP lines. In contrast, testosterone levels were increased in intoxicated WSP females and in both WSR and WSP mice during withdrawal. Chronic ethanol exposure disrupted normal estrous cycling in WSP mice, associated with hyperandrogenemia while intoxicated. In abstinence, elevated testosterone was observed in both sexes but only in WSR mice. Estrogen levels were modestly reduced during withdrawal in both WSR and WSP lines, predominantly in males. These findings identify a mechanism based on altered androgen signaling that likely contributes to sex-specific responses during withdrawal. However, only WSR mice showed similar elevations in androgen long after withdrawal in both sexes, suggesting that genotype is an important determinant of steroid responses after abstinence. Increased androgen signaling in females as a consequence of chronic ethanol exposure may play an important and relatively uncharacterized role in sexually dimorphic responses to alcohol abuse.
Collapse
|
45
|
Molecular targets of alcohol action: Translational research for pharmacotherapy development and screening. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:293-347. [PMID: 21199775 DOI: 10.1016/b978-0-12-385506-0.00007-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alcohol abuse and dependence are multifaceted disorders with neurobiological, psychological, and environmental components. Research on other complex neuropsychiatric diseases suggests that genetically influenced intermediate characteristics affect the risk for heavy alcohol consumption and its consequences. Diverse therapeutic interventions can be developed through identification of reliable biomarkers for this disorder and new pharmacological targets for its treatment. Advances in the fields of genomics and proteomics offer a number of possible targets for the development of new therapeutic approaches. This brain-focused review highlights studies identifying neurobiological systems associated with these targets and possible pharmacotherapies, summarizing evidence from clinically relevant animal and human studies, as well as sketching improvements and challenges facing the fields of proteomics and genomics. Concluding thoughts on using results from these profiling technologies for medication development are also presented.
Collapse
|
46
|
Hashimoto JG, Forquer MR, Tanchuck MA, Finn DA, Wiren KM. Importance of genetic background for risk of relapse shown in altered prefrontal cortex gene expression during abstinence following chronic alcohol intoxication. Neuroscience 2010; 173:57-75. [PMID: 21081154 DOI: 10.1016/j.neuroscience.2010.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 10/21/2010] [Accepted: 11/05/2010] [Indexed: 11/19/2022]
Abstract
Alcoholism is a relapsing disorder associated with excessive consumption after periods of abstinence. Neuroadaptations in brain structure, plasticity and gene expression occur with chronic intoxication but are poorly characterized. Here we report identification of pathways altered during abstinence in prefrontal cortex, a brain region associated with cognitive dysfunction and damage in alcoholics. To determine the influence of genetic differences, an animal model was employed with widely divergent responses to alcohol withdrawal, the Withdrawal Seizure-Resistant (WSR) and Withdrawal Seizure-Prone (WSP) lines. Mice were chronically exposed to highly intoxicating concentrations of ethanol and withdrawn, then left abstinent for 21 days. Transcriptional profiling by microarray analyses identified a total of 562 genes as significantly altered during abstinence. Hierarchical cluster analysis revealed that the transcriptional response correlated with genotype/withdrawal phenotype rather than sex. Gene Ontology category overrepresentation analysis identified thyroid hormone metabolism, glutathione metabolism, axon guidance and DNA damage response as targeted classes of genes in low response WSR mice, with acetylation and histone deacetylase complex as highly dimorphic between WSR and WSP mice. Confirmation studies in WSR mice revealed both increased neurotoxicity by histopathologic examination and elevated triidothyronine (T3) levels. Most importantly, relapse drinking was reduced by inhibition of thyroid hormone synthesis in dependent WSR mice compared to controls. These findings provide in vivo physiological and behavioral validation of the pathways identified. Combined, these results indicate a fundamentally distinct neuroadaptive response during abstinence in mice genetically selected for divergent withdrawal severity. Identification of pathways altered in abstinence may aid development of novel therapeutics for targeted treatment of relapse in abstinent alcoholics.
Collapse
Affiliation(s)
- J G Hashimoto
- Research Service, Portland Veterans Affairs Medical Center, Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
47
|
Repunte-Canonigo V, Berton F, Cottone P, Reifel-Miller A, Roberts AJ, Morales M, Francesconi W, Sanna PP. A potential role for adiponectin receptor 2 (AdipoR2) in the regulation of alcohol intake. Brain Res 2010; 1339:11-7. [PMID: 20380822 PMCID: PMC2906226 DOI: 10.1016/j.brainres.2010.03.060] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 03/17/2010] [Indexed: 01/03/2023]
Abstract
The anterior cingulate cortex (ACC) has been implicated in alcohol and drug addiction. We recently identified the small G protein K-ras as an alcohol-regulated gene in the ACC by gene expression analysis. We show here that the adiponectin receptor 2 (AdipoR2) was differentially regulated by alcohol in the ACC in a K-ras-dependent manner. Additionally, withdrawal-associated increased drinking was attenuated in AdipoR2 null mice. Intracellular recordings revealed that adiponectin increased the excitability of ACC neurons and that this effect was more pronounced during alcohol withdrawal, suggesting that AdipoR2 signaling may contribute to increased ACC activity. Altogether, the data implicate K-ras-regulated pathways involving AdipoR2 in the cellular and behavioral actions of alcohol that may contribute to overactivity of the ACC during withdrawal and excessive alcohol drinking.
Collapse
Affiliation(s)
- Vez Repunte-Canonigo
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, 10550 N Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Fulvia Berton
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, 10550 N Torrey Pines Rd., La Jolla, CA, 92037, USA
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Pietro Cottone
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Anne Reifel-Miller
- Diabetes Research Division, Lilly Research Laboratories, Indianapolis, IN 46285
| | - Amanda J. Roberts
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, 10550 N Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Marisela Morales
- Behavioral Neuroscience Branch, IRP/NIDA/NIH/DHHS, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Walter Francesconi
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, 10550 N Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Pietro Paolo Sanna
- Molecular and Integrative Neuroscience Department, The Scripps Research Institute, 10550 N Torrey Pines Rd., La Jolla, CA, 92037, USA
| |
Collapse
|
48
|
Genome-wide gene expression analysis identifies K-ras as a regulator of alcohol intake. Brain Res 2010; 1339:1-10. [PMID: 20388501 DOI: 10.1016/j.brainres.2010.03.063] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 03/17/2010] [Indexed: 12/30/2022]
Abstract
Adaptations in the anterior cingulate cortex (ACC) have been implicated in alcohol and drug addiction. To identify genes that may contribute to excessive drinking, here we performed microarray analyses in laser microdissected rat ACC after a single or repeated administration of an intoxicating dose of alcohol (3 g/kg). Expression of the small G protein K-ras was differentially regulated following both single and repeated alcohol administration. We also observed that voluntary alcohol intake in K-ras heterozygous null mice (K-ras(+/-)) did not increase after withdrawal from repeated cycles of intermittent ethanol vapor exposure, unlike in their wild-type littermates. To identify K-ras regulated pathways, we then profiled gene expression in the ACC of K-ras(+/-), heterozygous null mice for the K-ras negative regulator Nf1 (Nf1(+/-)) and wild-type mice following repeated administration of an intoxicating dose of alcohol. Pathway analysis showed that alcohol differentially affected various pathways in a K-ras dependent manner - some of which previously shown to be regulated by alcohol - including the insulin/PI3K pathway, the NF-kappaB, the phosphodiesterases (PDEs) pathway, the Jak/Stat and the adipokine signaling pathways. Altogether, the data implicate K-ras-regulated pathways in the regulation of excessive alcohol drinking after a history of dependence.
Collapse
|
49
|
Abstract
Regulator of G protein-signaling (RGS) proteins are a family of more than 30 intracellular proteins that negatively modulate intracellular signaling of receptors in the G protein-coupled receptor family. This family includes receptors for opioids, cannabinoids, and dopamine that mediate the acute effects of addictive drugs or behaviors and chronic effects leading to the development of addictive disease. Members of the RGS protein family, by negatively modulating receptor signaling, influence the intracellular processes that lead to addiction. In turn, addictive drugs control the expression levels of several RGS proteins. This review will consider the distribution and mechanisms of action of RGS proteins, particularly the R4 and R7 families that have been implicated in the actions of addictive drugs, how knowledge of these proteins is contributing to an understanding of addictive processes, and whether specific RGS proteins could provide targets for the development of medications to manage and/or treat addiction.
Collapse
Affiliation(s)
- John Traynor
- Department of Pharmacology and Substance Abuse Research Center, University of Michigan, Ann Arbor, Michigan 48109-5632, USA.
| |
Collapse
|
50
|
Abstract
Alcohol intake at levels posing an acute heath risk is common amongst teenagers. Alcohol abuse is the second most common mental disorder worldwide. The incidence of smoking is decreasing in the Western world but increasing in developing countries and is the leading cause of preventable death worldwide. Considering the longstanding history of alcohol and tobacco consumption in human societies, it might be surprising that the molecular mechanisms underlying alcohol and smoking dependence are still incompletely understood. Effective treatments against the risk of relapse are lacking. Drugs of abuse exert their effect manipulating the dopaminergic mesocorticolimbic system. In this brain region, alcohol has many potential targets including membranes and several ion channels, while other drugs, for example nicotine, act via specific receptors or binding proteins. Repeated consumption of drugs of abuse mediates adaptive changes within this region, resulting in addiction. The high incidence of alcohol and nicotine co-abuse complicates analysis of the molecular basis of the disease. Gene expression profiling is a useful approach to explore novel drug targets in the brain. Several groups have utilised this technology to reveal drug-sensitive pathways in the mesocorticolimbic system of animal models and in human subjects. These studies are the focus of the present review.
Collapse
|