1
|
Huang D, Li M, Qiao Z, Zhou H, Cai Y, Li X, Zhang Z, Zhou J. Effects of adolescent alcohol exposure on oligodendrocyte lineage cells and myelination in mice: Age and subregion differences. IBRO Neurosci Rep 2024; 17:220-234. [PMID: 39282551 PMCID: PMC11401168 DOI: 10.1016/j.ibneur.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 09/19/2024] Open
Abstract
Adolescence is an important phase for the structural and functional development of the brain. The immaturity of adolescent brain development is associated with high susceptibility to exogenous disturbances, including alcohol. In this study, the acquisition of conditioned place preference (CPP) in adolescent mice by alcohol (2 g/kg) and the parvalbumin-positive interneurons (PV+ interneurons), oligodendrocyte lineage cells (OPCs), and myelination in the medial prefrontal cortex (mPFC) were assessed. We aim to determine the age- and subregional-specificity of the effects of alcohol. Alcohol (2 g/kg) was injected intraperitoneally on even days, and saline was injected intraperitoneally on odd days. The control group received a continuous intraperitoneal injection with saline. Differences in alcohol-induced CPP acquisition were assessed, followed by immunohistochemical staining. The results showed a pronounced CPP acquisition in 4- and 5-week-old mice. In the mPFC, there were reduced PV+ interneurons and OPCs in 3-week-old mice and reduced oligodendrocyte numbers in 4-week-old mice. The 5-week-old mice showed impaired myelination and a decrease in the number of PV+ interneurons, mature oligodendrocytes, and OPCs in the mPFC. Since the alterations in 5-week-old mice are more pronounced, we further explored the mPFC-associated subregional-specificity. In the alcohol-exposed mice, the oligodendrocyte numbers were decreased in the anterior cingulate cortex (ACC), PV+ interneuron numbers were declined in the prelimbic cortex (PL), and the number of oligodendrocytes, PV+ interneurons, and OPCs was also decreased with impaired myelination in the infralimbic cortex (IL). Our data suggest that adolescent alcohol exposure notably affected the acquisition of CPP, myelin formation, and the counts of PV+ interneurons, mature oligodendrocytes, and OPCs in the mPFC in 5-week-old mice. Also, the IL subregion was the worst-affected subregion of the mPFC in alcohol-exposed 5-week-old mice. It reveals that the effects of alcohol on adolescence and its mPFC myelination show obvious age- and subregional-specificity.
Collapse
Affiliation(s)
- Dong Huang
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Maolin Li
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhifei Qiao
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hongli Zhou
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Cai
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaolong Li
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zuo Zhang
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiyin Zhou
- Clinical Research Center, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
2
|
Brocato ER, Easter R, Morgan A, Kakani M, Lee G, Wolstenholme JT. Adolescent binge ethanol impacts H3K9me3-occupancy at synaptic genes and the regulation of oligodendrocyte development. Front Mol Neurosci 2024; 17:1389100. [PMID: 38840776 PMCID: PMC11150558 DOI: 10.3389/fnmol.2024.1389100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Binge drinking in adolescence can disrupt myelination and cause brain structural changes that persist into adulthood. Alcohol consumption at a younger age increases the susceptibility of these changes. Animal models to understand ethanol's actions on myelin and white matter show that adolescent binge ethanol can alter the developmental trajectory of oligodendrocytes, myelin structure, and myelin fiber density. Oligodendrocyte differentiation is epigenetically regulated by H3K9 trimethylation (H3K9me3). Prior studies have shown that adolescent binge ethanol dysregulates H3K9 methylation and decreases H3K9-related gene expression in the PFC. Methods Here, we assessed ethanol-induced changes to H3K9me3 occupancy at genomic loci in the developing adolescent PFC. We further assessed ethanol-induced changes at the transcription level with qPCR time course approaches in oligodendrocyte-enriched cells to assess changes in oligodendrocyte progenitor and oligodendrocytes specifically. Results Adolescent binge ethanol altered H3K9me3 regulation of synaptic-related genes and genes specific for glutamate and potassium channels in a sex-specific manner. In PFC tissue, we found an early change in gene expression in transcription factors associated with oligodendrocyte differentiation that may lead to the later significant decrease in myelin-related gene expression. This effect appeared stronger in males. Conclusion Further exploration in oligodendrocyte cell enrichment time course and dose response studies could suggest lasting dysregulation of oligodendrocyte maturation at the transcriptional level. Overall, these studies suggest that binge ethanol may impede oligodendrocyte differentiation required for ongoing myelin development in the PFC by altering H3K9me3 occupancy at synaptic-related genes. We identify potential genes that may be contributing to adolescent binge ethanol-related myelin loss.
Collapse
Affiliation(s)
- Emily R. Brocato
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Rachel Easter
- Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Alanna Morgan
- Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Meenakshi Kakani
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Grace Lee
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T. Wolstenholme
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
- Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
3
|
Crews FT, Macht V, Vetreno RP. Epigenetic regulation of microglia and neurons by proinflammatory signaling following adolescent intermittent ethanol (AIE) exposure and in human AUD. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2024; 4:12094. [PMID: 38524847 PMCID: PMC10957664 DOI: 10.3389/adar.2024.12094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/05/2024] [Indexed: 03/26/2024]
Abstract
Adolescent alcohol drinking is linked to high rates of adult alcohol problems and alcohol use disorder (AUD). The Neurobiology of Alcohol Drinking in Adulthood (NADIA) consortium adolescent intermittent ethanol (AIE) models adolescent binge drinking, followed by abstinent maturation to adulthood to determine the persistent AIE changes in neurobiology and behavior. AIE increases adult alcohol drinking and preference, increases anxiety and reward seeking, and disrupts sleep and cognition, all risks for AUD. In addition, AIE induces changes in neuroimmune gene expression in neurons and glia that alter neurocircuitry and behavior. HMGB1 is a unique neuroimmune signal released from neurons and glia by ethanol that activates multiple proinflammatory receptors, including Toll-like receptors (TLRs), that spread proinflammatory gene induction. HMGB1 expression is increased by AIE in rat brain and in post-mortem human AUD brain, where it correlates with lifetime alcohol consumption. HMGB1 activation of TLR increase TLR expression. Human AUD brain and rat brain following AIE show increases in multiple TLRs. Brain regional differences in neurotransmitters and cell types impact ethanol responses and neuroimmune gene induction. Microglia are monocyte-like cells that provide trophic and synaptic functions, that ethanol proinflammatory signals sensitize or "prime" during repeated drinking cycles, impacting neurocircuitry. Neurocircuits are differently impacted dependent upon neuronal-glial signaling. Acetylcholine is an anti-inflammatory neurotransmitter. AIE increases HMGB1-TLR4 signaling in forebrain, reducing cholinergic neurons by silencing multiple cholinergic defining genes through upregulation of RE-1 silencing factor (REST), a transcription inhibitor known to regulate neuronal differentiation. HMGB1 REST induction reduces cholinergic neurons in basal forebrain and cholinergic innervation of hippocampus. Adult brain hippocampal neurogenesis is regulated by a neurogenic niche formed from multiple cells. In vivo AIE and in vitro studies find ethanol increases HMGB1-TLR4 signaling and other proinflammatory signaling as well as reducing trophic factors, NGF, and BDNF, coincident with loss of the cholinergic synapse marker vChAT. These changes in gene expression-transcriptomes result in reduced adult neurogenesis. Excitingly, HMGB1 antagonists, anti-inflammatories, and epigenetic modifiers like histone deacetylase inhibitors restore trophic the neurogenesis. These findings suggest anti-inflammatory and epigenetic drugs should be considered for AUD therapy and may provide long-lasting reversal of psychopathology.
Collapse
Affiliation(s)
- Fulton T. Crews
- Departments of Pharmacology and Psychiatry, Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | |
Collapse
|
4
|
De Clerck M, Manguin M, Henkous N, d’Almeida MN, Beracochea D, Mons N. Chronic alcohol-induced long-lasting working memory deficits are associated with altered histone H3K9 dimethylation in the prefrontal cortex. Front Behav Neurosci 2024; 18:1354390. [PMID: 38495426 PMCID: PMC10941761 DOI: 10.3389/fnbeh.2024.1354390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction Epigenetic modifications have emerged as key contributors to the enduring behavioral, molecular and epigenetic neuroadaptations during withdrawal from chronic alcohol exposure. The present study investigated the long-term consequences of chronic alcohol exposure on spatial working memory (WM) and associated changes of transcriptionally repressive histone H3 lysine 9 dimethylation (H3K9me2) in the prefrontal cortex (PFC). Methods Male C57BL/6 mice were allowed free access to either 12% (v/v) ethanol for 5 months followed by a 3-week abstinence period or water. Spatial WM was assessed through the spontaneous alternation T-maze test. Alcoholic and water mice received daily injections of GABAB agonist baclofen or saline during alcohol fading and early withdrawal. Global levels of histone modifications were determined by immunohistochemistry. Results Withdrawal mice displayed WM impairments along with reduced prefrontal H3K9me2 levels, compared to water-drinking mice. The withdrawal-induced decrease of H3K9me2 occurred concomitantly with increased level of permissive H3K9 acetylation (H3K9ac) in the PFC. Baclofen treatment rescued withdrawal-related WM deficits and fully restored prefrontal H3K9me2 and H3K9ac. Alcohol withdrawal induced brain region-specific changes of H3K9me2 and H3K9ac after testing, with significant decreases of both histone marks in the dorsal hippocampus and no changes in the amygdala and dorsal striatum. Furthermore, the magnitude of H3K9me2 in the PFC, but not the hippocampus, significantly and positively correlated with individual WM performances. No correlation was observed between H3K9ac and behavioral performance. Results also indicate that pre-testing intraperitoneal injection of UNC0642, a selective inhibitor of histone methyltransferase G9a responsible for H3K9me2, led to WM impairments in water-drinking and withdrawal-baclofen mice. Collectively, our results demonstrate that alcohol withdrawal induced brain-region specific alterations of H3K9me2 and H3K9ac, an effect that persisted for at least three weeks after cessation of chronic alcohol intake. Conclusion The findings suggest a role for long-lasting decreased H3K9me2 specifically in the PFC in the persistent WM impairments related to alcohol withdrawal.
Collapse
|
5
|
Lodha J, Brocato ER, Nash M, Marcus MM, Pais AC, Pais AB, Miles MF, Wolstenholme JT. Adolescent social housing protects against adult emotional and cognitive deficits and alters the PFC and NAc transcriptome in male and female C57BL/6J mice. Front Neurosci 2023; 17:1287584. [PMID: 38130694 PMCID: PMC10733512 DOI: 10.3389/fnins.2023.1287584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Adolescence is a critical period in cognitive and emotional development, characterized by high levels of social interaction and increases in risk-taking behavior including binge drinking. Adolescent exposure to social stress and binge ethanol have individually been associated with the development of social, emotional, and cognitive deficits, as well as increased risk for alcohol use disorder. Disruption of cortical development by early life social stress and/or binge drinking may partly underlie these enduring emotional, cognitive, and behavioral effects. The study goal is to implement a novel neighbor housing environment to identify the effects of adolescent neighbor housing and/or binge ethanol drinking on (1) a battery of emotional and cognitive tasks (2) adult ethanol drinking behavior, and (3) the nucleus accumbens and prefrontal cortex transcriptome. Methods Adolescent male and female C57BL/6J mice were single or neighbor housed with or without access to intermittent ethanol. One cohort underwent behavioral testing during adulthood to determine social preference, expression of anxiety-like behavior, cognitive performance, and patterns of ethanol intake. The second cohort was sacrificed in late adolescence and brain tissue was used for transcriptomics analysis. Results As adults, single housed mice displayed decreased social interaction, deficits in the novel object recognition task, and increased anxiety-like behavior, relative to neighbor-housed mice. There was no effect of housing condition on adolescent or adult ethanol consumption. Adolescent ethanol exposure did not alter adult ethanol intake. Transcriptomics analysis revealed that adolescent housing condition and ethanol exposure resulted in differential expression of genes related to synaptic plasticity in the nucleus accumbens and genes related to methylation, the extracellular matrix and inflammation in the prefrontal cortex. Discussion The behavioral results indicate that social interaction during adolescence via the neighbor housing model may protect against emotional, social, and cognitive deficits. In addition, the transcriptomics results suggest that these behavioral alterations may be mediated in part by dysregulation of transcription in the frontal cortex or the nucleus accumbens.
Collapse
Affiliation(s)
- Jyoti Lodha
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Emily R. Brocato
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - McKenzie Nash
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Madison M. Marcus
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - A. Chris Pais
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Alex B. Pais
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Michael F. Miles
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer Theresa Wolstenholme
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
6
|
Wooden JI, Peacoe LE, Anasooya Shaji C, Melbourne JK, Chandler CM, Bardo MT, Nixon K. Adolescent Intermittent Ethanol Drives Modest Neuroinflammation but Does Not Escalate Drinking in Male Rats. Cells 2023; 12:2572. [PMID: 37947650 PMCID: PMC10649200 DOI: 10.3390/cells12212572] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
During adolescence, the brain is highly susceptible to alcohol-induced damage and subsequent neuroimmune responses, effects which may enhance development of an alcohol use disorder (AUD). Neuroimmune reactions are implicated in adolescent alcohol exposure escalating adulthood drinking. Therefore, we investigated whether intermittent alcohol exposure in male, adolescent rats (AIE) escalated adult drinking via two-bottle choice (2BC). We also examined the influence of housing environment across three groups: standard (group-housed with enrichment during 2BC), impoverished (group-housed without enrichment during 2BC), or isolation (single-housed without bedding or enrichment throughout). In the standard group immediately after AIE/saline and after 2BC, we also examined the expression of microglial marker, Iba1, reactive astrocyte marker, vimentin, and neuronal cell death dye, FluoroJade B (FJB). We did not observe an escalation of adulthood drinking following AIE, regardless of housing condition. Further, only a modest neuroimmune response occurred after AIE in the standard group: no significant microglial reactivity or neuronal cell death was apparent using this model, although some astrocyte reactivity was detected in adolescence following AIE that resolved by adulthood. These data suggest that the lack of neuroimmune response in adolescence in this model may underlie the lack of escalation of alcohol drinking, which could not be modified through isolation stress.
Collapse
Affiliation(s)
- Jessica I. Wooden
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lauren E. Peacoe
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Chinchusha Anasooya Shaji
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer K. Melbourne
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Cassie M. Chandler
- Department of Psychology, University of Kentucky, Lexington, KY 40506, USA (M.T.B.)
| | - Michael T. Bardo
- Department of Psychology, University of Kentucky, Lexington, KY 40506, USA (M.T.B.)
| | - Kimberly Nixon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
7
|
Smith ML, Sergi Z, Mignogna KM, Rodriguez NE, Tatom Z, MacLeod L, Choi KB, Philip V, Miles MF. Identification of Genetic and Genomic Influences on Progressive Ethanol Consumption in Diversity Outbred Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.15.554349. [PMID: 37745421 PMCID: PMC10515943 DOI: 10.1101/2023.09.15.554349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Genetic factors play a significant role in the risk for development of alcohol use disorder (AUD). Using 3-bottle choice intermittent access ethanol (IEA), we have employed the Diversity Outbred (DO) mouse panel as a model of alcohol use disorder in a genetically diverse population. Through use of gene expression network analysis techniques, in combination with expression quantitative trait loci (eQTL) mapping, we have completed an extensive analysis of the influence of genetic background on gene expression changes in the prefrontal cortex (PFC). This approach revealed that, in DO mice, genes whose expression was significantly disrupted by intermittent ethanol in the PFC also tended to be those whose expression correlated to intake. This finding is in contrast to previous studies of both mice and nonhuman primates. Importantly, these analyses identified genes involved in myelination in the PFC as significantly disrupted by IEA, correlated to ethanol intake, and having significant eQTLs. Genes that code for canonical components of the myelin sheath, such as Mbp, also emerged as key drivers of the gene expression response to intermittent ethanol drinking. Several regulators of myelination were also key drivers of gene expression, and had significant QTLs, indicating that genetic background may play an important role in regulation of brain myelination. These findings underscore the importance of disruption of normal myelination in the PFC in response to prolonged ethanol exposure, that genetic variation plays an important role in this response, and that this interaction between genetics and myelin disruption in the presence of ethanol may underlie previously observed behavioral changes under intermittent access ethanol drinking such as escalation of consumption.
Collapse
Affiliation(s)
- M L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Z Sergi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - K M Mignogna
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - N E Rodriguez
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Z Tatom
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - L MacLeod
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - K B Choi
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - V Philip
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - M F Miles
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
8
|
Jacobson MM, Jenkins LM, Feldman DA, Crane NA, Langenecker SA. Reduced connectivity of the cognitive control neural network at rest in young adults who had their first drink of alcohol prior to age 18. Psychiatry Res Neuroimaging 2023; 332:111642. [PMID: 37086604 PMCID: PMC10247408 DOI: 10.1016/j.pscychresns.2023.111642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/21/2023] [Accepted: 04/05/2023] [Indexed: 04/24/2023]
Abstract
The cognitive control network (CCN) is an important network responsible for performing and modulating executive functions. In adolescents, alcohol use has been associated with weaker cognitive control, higher reward sensitivity, and later-in-life alcohol problems. Given that the CCN continues to develop into young adulthood, it is important to understand relations between early alcohol use, the CCN, and reward networks. Participants included individuals 18-23 years without alcohol use disorder. Based upon self-reported age of first alcoholic drink, participants were split into two groups: Early (onset) Drinkers (first drink < age 18, N = 52) and Late (onset) Drinkers (first drink > age 18, N = 44). All participants underwent an 8-minute resting-state fMRI scan. Seed regions of interest included the anterior dorsolateral prefrontal cortex (DLPFC), amygdala, and ventral striatum. Early Drinkers demonstrated significant reduced connectivity of CCN regions, including bilateral anterior DLPFC, compared to Late Drinkers. There were no significant differences between Early and Late Drinkers in connectivity between reward and CCN regions. These results suggest that individuals who begin drinking alcohol earlier in life may have alterations in the development of the CCN; however, longitudinal research is necessary to determine whether lower connectivity precedes or follows early alcohol use, and any other relevant factors.
Collapse
Affiliation(s)
- Maci M Jacobson
- Department of Psychiatry, The University of Utah, United States; Interdisciplinary Neuroscience Program, The University of Utah, United States.
| | - Lisanne M Jenkins
- Department of Psychiatry and Behavioral Sciences, Northwestern University, United States; Department of Psychiatry, The University of Illinois at Chicago, United States
| | | | - Natania A Crane
- Department of Psychiatry, The University of Illinois at Chicago, United States
| | - Scott A Langenecker
- Department of Psychiatry, The University of Utah, United States; Interdisciplinary Neuroscience Program, The University of Utah, United States; Department of Psychiatry, The University of Illinois at Chicago, United States
| |
Collapse
|
9
|
Brocato ER, Wolstenholme JT. Adolescent binge ethanol impacts H3K36me3 regulation of synaptic genes. Front Mol Neurosci 2023; 16:1082104. [PMID: 36937047 PMCID: PMC10020663 DOI: 10.3389/fnmol.2023.1082104] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Adolescence is marked in part by the ongoing development of the prefrontal cortex (PFC). Binge ethanol use during this critical stage in neurodevelopment induces significant structural changes to the PFC, as well as cognitive and behavioral deficits that can last into adulthood. Previous studies showed that adolescent binge ethanol causes lasting deficits in working memory, decreases in the expression of chromatin remodeling genes responsible for the methylation of histone 3 lysine 36 (H3K36), and global decreases in H3K36 in the PFC. H3K36me3 is present within the coding region of actively-transcribed genes, and safeguards against aberrant, cryptic transcription by RNA Polymerase II. We hypothesize that altered methylation of H3K36 could play a role in adolescent binge ethanol-induced memory deficits. To investigate this at the molecular level, ethanol (4 g/kg, i.g.) or water was administered intermittently to adolescent mice. RNA-and ChIP-sequencing were then performed within the same tissue to determine gene expression changes and identify genes and loci where H3K36me3 was disrupted by ethanol. We further assessed ethanol-induced changes at the transcription level with differential exon-use and cryptic transcription analysis - a hallmark of decreased H3K36me3. Here, we found ethanol-induced changes to the gene expression and H3K36me3-regulation of synaptic-related genes in all our analyses. Notably, H3K36me3 was differentially trimethylated between ethanol and control conditions at synaptic-related genes, and Snap25 and Cplx1 showed evidence of cryptic transcription in males and females treated with ethanol during adolescence. Our results provide preliminary evidence that ethanol-induced changes to H3K36me3 during adolescent neurodevelopment may be linked to synaptic dysregulation at the transcriptional level, which may explain the reported ethanol-induced changes to PFC synaptic function.
Collapse
Affiliation(s)
- Emily R. Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T. Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
10
|
Crews FT, Coleman LG, Macht VA, Vetreno RP. Targeting Persistent Changes in Neuroimmune and Epigenetic Signaling in Adolescent Drinking to Treat Alcohol Use Disorder in Adulthood. Pharmacol Rev 2023; 75:380-396. [PMID: 36781218 PMCID: PMC9969522 DOI: 10.1124/pharmrev.122.000710] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
Studies universally find early age of drinking onset is linked to lifelong risks of alcohol problems and alcohol use disorder (AUD). Assessment of the lasting effect of drinking during adolescent development in humans is confounded by the diversity of environmental and genetic factors that affect adolescent development, including emerging personality disorders and progressive increases in drinking trajectories into adulthood. Preclinical studies using an adolescent intermittent ethanol (AIE) exposure rat model of underage binge drinking avoid the human confounds and support lifelong changes that increase risks. AIE increases adult alcohol drinking, risky decision-making, reward-seeking, and anxiety as well as reductions in executive function that all increase risks for the development of an AUD. AIE causes persistent increases in brain neuroimmune signaling high-mobility group box 1 (HMGB1), Toll-like receptor, receptor for advanced glycation end products, and innate immune genes that are also found to be increased in human AUD brain. HMGB1 is released from cells by ethanol, both free and within extracellular vesicles, that act on neurons and glia, shifting transcription and cellular phenotype. AIE-induced decreases in adult hippocampal neurogenesis and loss of basal forebrain cholinergic neurons are reviewed as examples of persistent AIE-induced pathology. Both are prevented and reversed by anti-inflammatory and epigenetic drugs. Findings suggest AIE-increased HMGB1 signaling induces the RE-1 silencing transcript blunting cholinergic gene expression, shifting neuronal phenotype. Inhibition of HMGB1 neuroimmune signaling, histone methylation enzymes, and galantamine, the cholinesterase inhibitor, both prevent and reverse AIE pathology. These findings provide new targets that may reverse AUD neuropathology as well as other brain diseases linked to neuroimmune signaling. SIGNIFICANCE STATEMENT: Adolescent underage binge drinking studies find that earlier adolescent drinking is associated with lifelong alcohol problems including high levels of lifetime alcohol use disorder (AUD). Preclinical studies find the underage binge drinking adolescent intermittent ethanol (AIE) model causes lasting changes in adults that increase risks of developing adult alcohol problems. Loss of hippocampal neurogenesis and loss of basal forebrain cholinergic neurons provide examples of how AIE-induced epigenetic and neuroimmune signaling provide novel therapeutic targets for adult AUD.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies and Departments of Pharmacology and Psychiatry, School of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Leon G Coleman
- Bowles Center for Alcohol Studies and Departments of Pharmacology and Psychiatry, School of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Victoria A Macht
- Bowles Center for Alcohol Studies and Departments of Pharmacology and Psychiatry, School of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies and Departments of Pharmacology and Psychiatry, School of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
11
|
Holloway KN, Pinson MR, Douglas JC, Rafferty TM, Kane CJM, Miranda RC, Drew PD. Cerebellar Transcriptomic Analysis in a Chronic plus Binge Mouse Model of Alcohol Use Disorder Demonstrates Ethanol-Induced Neuroinflammation and Altered Glial Gene Expression. Cells 2023; 12:745. [PMID: 36899881 PMCID: PMC10000476 DOI: 10.3390/cells12050745] [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: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Alcohol use disorder (AUD) is one of the most common preventable mental health disorders and can result in pathology within the CNS, including the cerebellum. Cerebellar alcohol exposure during adulthood has been associated with disruptions in proper cerebellar function. However, the mechanisms regulating ethanol-induced cerebellar neuropathology are not well understood. High-throughput next generation sequencing was performed to compare control versus ethanol-treated adult C57BL/6J mice in a chronic plus binge model of AUD. Mice were euthanized, cerebella were microdissected, and RNA was isolated and submitted for RNA-sequencing. Down-stream transcriptomic analyses revealed significant changes in gene expression and global biological pathways in control versus ethanol-treated mice that included pathogen-influenced signaling pathways and cellular immune response pathways. Microglial-associated genes showed a decrease in homeostasis-associated transcripts and an increase in transcripts associated with chronic neurodegenerative diseases, while astrocyte-associated genes showed an increase in transcripts associated with acute injury. Oligodendrocyte lineage cell genes showed a decrease in transcripts associated with both immature progenitors as well as myelinating oligodendrocytes. These data provide new insight into the mechanisms by which ethanol induces cerebellar neuropathology and alterations to the immune response in AUD.
Collapse
Affiliation(s)
- Kalee N. Holloway
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Marisa R. Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (M.R.P.); (R.C.M.)
| | - James C. Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Tonya M. Rafferty
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Cynthia J. M. Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Rajesh C. Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (M.R.P.); (R.C.M.)
| | - Paul D. Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
12
|
CB2R activation ameliorates late adolescent chronic alcohol exposure-induced anxiety-like behaviors during withdrawal by preventing morphological changes and suppressing NLRP3 inflammasome activation in prefrontal cortex microglia in mice. Brain Behav Immun 2023; 110:60-79. [PMID: 36754245 DOI: 10.1016/j.bbi.2023.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/08/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chronic alcohol exposure (CAE) during late adolescence increases the risk of anxiety development. Alcohol-induced prefrontal cortex (PFC) microglial activation, characterized by morphological changes and increased associations with neurons, plays a critical role in the pathogenesis of anxiety. Alcohol exposure increases NLRP3 inflammasome expression, increasing cytokine secretion by activated microglia. Cannabinoid type 2 receptor (CB2R), an essential receptor of the endocannabinoid system, regulates microglial activation and neuroinflammatory reactions. We aimed to investigate the role of CB2R activation in ameliorating late adolescent CAE-induced anxiety-like behaviors and microglial activation in C57BL/6J mice. METHODS Six-week-old C57BL/6J mice were acclimated for 7 days and then were administered alcohol by gavage (4 g/kg, 25 % w/v) for 28 days. The mice were intraperitoneally injected with the specific CB2R agonist AM1241 1 h before alcohol treatment. Anxiety-like behaviors during withdrawal were assessed by open field test and elevated plus maze test 24 h after the last alcohol administration. Microglial activation, microglia-neuron interactions, and CB2R and NLRP3 inflammasome-related molecule expression in the PFC were measured using immunofluorescence, immunohistochemical, qPCR, and Western blotting assays. Microglial morphology was evaluated by Sholl analysis and the cell body-to-total cell size index. Additionally, N9 microglia were activated by LPS in vitro, and the effects of AM1241 on NLRP3 and N9 microglial activation were investigated. RESULTS After CAE, mice exhibited severe anxiety-like behaviors during withdrawal. CAE induced obvious microglia-neuron associations, and increased expression of microglial activation markers, CB2R, and NLRP3 inflammasome-related molecules in the PFC. Microglia also showed marked filament retraction and reduction and cell body enlargement after CAE. AM1241 treatment ameliorated anxiety-like behaviors in CAE model mice, and it prevented microglial morphological changes, reduced microglial activation marker expression, and suppressed the microglial NLRP3 inflammasome activation and proinflammatory cytokine secretion induced by CAE. AM1241 suppressed the LPS-induced increase in NLRP3 inflammasome-related molecules, IL-1β release, and M1 phenotype markers (iNOS and CD86) in N9 cell, which was reversed by CB2R antagonist treatment. CONCLUSIONS CAE caused anxiety-like behaviors in late adolescent mice at least partly by inducing microglial activation and increasing microglia-neuron associations in the PFC. CB2R activation ameliorated these effects by preventing morphological changes and suppressing NLRP3 inflammasome activation in PFC microglia.
Collapse
|
13
|
Tetteh-Quarshie S, Risher ML. Adolescent brain maturation and the neuropathological effects of binge drinking: A critical review. Front Neurosci 2023; 16:1040049. [PMID: 36733924 PMCID: PMC9887052 DOI: 10.3389/fnins.2022.1040049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
Adolescence is a transitional stage marked by continued brain development. This period is accompanied by physical and neurochemical modifications in the shape and function of the hippocampus, prefrontal cortex, and other limbic system structures. Brain maturation during adolescence, which is typically governed by intrinsic factors, can be dramatically altered by environmental influences such as drugs and alcohol. Unlike many other addictive substances, binge drinking is very common and normative among teenagers and young adults. This repeated pattern of excessive alcohol consumption in adolescents has been shown to cause behavioral changes and neurocognitive impairments that include increased anxiety, risky decision-making, and learning deficits, which could lead to the development of alcohol use disorder (AUD). This manuscript highlights factors that lead to adolescent binge drinking, discusses maturational changes that occur in an adolescent's brain, and then evaluates the effect of adolescent alcohol consumption on brain structure, function, and neurocognitive abilities in both human studies and animal models. The impact of gender/sex and COVID-19 are briefly discussed. Understanding the factors that promote the onset of adolescent binge drinking and its undesirable consequences could serve as a catalyst for developing therapeutic agents that would decrease or eradicate the damaging effects of alcohol on an adolescent brain.
Collapse
Affiliation(s)
- Samuel Tetteh-Quarshie
- Department of Biomedical Science and Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Mary-Louise Risher
- Department of Biomedical Science and Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States,Neurobiology Research Laboratory, Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV, United States,*Correspondence: Mary-Louise Risher,
| |
Collapse
|
14
|
Narendra S, Klengel C, Hamzeh B, Patel D, Otten J, Lardenoije R, Newman EL, Miczek KA, Klengel T, Ressler KJ, Suh J. Genome-wide transcriptomics of the amygdala reveals similar oligodendrocyte-related responses to acute and chronic alcohol drinking in female mice. Transl Psychiatry 2022; 12:476. [PMID: 36371333 PMCID: PMC9653459 DOI: 10.1038/s41398-022-02231-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/13/2022] Open
Abstract
Repeated excessive alcohol consumption is a risk factor for alcohol use disorder (AUD). Although AUD has been more common in men than women, women develop more severe behavioral and physical impairments. However, relatively few new therapeutics targeting development of AUD, particularly in women, have been validated. To gain a better understanding of molecular mechanisms underlying alcohol intake, we conducted a genome-wide RNA-sequencing analysis in female mice exposed to different modes (acute vs chronic) of ethanol drinking. We focused on transcriptional profiles in the amygdala including the central and basolateral subnuclei, brain areas previously implicated in alcohol drinking and seeking. Surprisingly, we found that both drinking modes triggered similar changes in gene expression and canonical pathways, including upregulation of ribosome-related/translational pathways and myelination pathways, and downregulation of chromatin binding and histone modification. In addition, analyses of hub genes and upstream regulatory pathways revealed that voluntary ethanol consumption affects epigenetic changes via histone deacetylation pathways, oligodendrocyte and myelin function, and the oligodendrocyte-related transcription factor, Sox17. Furthermore, a viral vector-assisted knockdown of Sox17 gene expression in the amygdala prevented a gradual increase in alcohol consumption during repeated accesses. Overall, these results suggest that the expression of oligodendrocyte-related genes in the amygdala is sensitive to voluntary alcohol drinking in female mice. These findings suggest potential molecular targets for future therapeutic approaches to prevent the development of AUD, due to repeated excessive alcohol consumption, particularly in women.
Collapse
Affiliation(s)
- Sharvari Narendra
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Bioinformatics, Northeastern University, Boston, MA, 02115, USA
| | - Claudia Klengel
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Bilal Hamzeh
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Drasti Patel
- Department of Bioinformatics, Northeastern University, Boston, MA, 02115, USA
| | - Joy Otten
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Roy Lardenoije
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Emily L Newman
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Klaus A Miczek
- Psychology and Neuroscience Departments, Tufts University, Medford, MA, 02155, USA
| | - Torsten Klengel
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Kerry J Ressler
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA.
| | - Junghyup Suh
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
15
|
de Abreu MS, Parker MO, Kalueff AV. The critical impact of sex on preclinical alcohol research - Insights from zebrafish. Front Neuroendocrinol 2022; 67:101014. [PMID: 35810841 DOI: 10.1016/j.yfrne.2022.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/27/2022]
Abstract
Sex is an important biological variable that is widely recognized in studies of alcohol-related effects. Complementing clinical and preclinical rodent research, the zebrafish (Danio rerio) is the second most used laboratory species, and a powerful model organism in biomedicine. Like clinical and rodent models, zebrafish demonstrate overt sex differences in alcohol-related responses. Collectively, this evidence shows that the zebrafish becomes a sensitive model species to further probe in-depth sex differences commonly reported in alcohol research.
Collapse
Affiliation(s)
- Murilo S de Abreu
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia.
| | - Matthew O Parker
- School of Pharmacy and Biomedical Science, University of Portsmouth, UK
| | - Allan V Kalueff
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Ural Federal University, Ekaterinburg, Russia; Neuroscience Program, Sirius University of Science and Technology, Sochi, Russia; Granov Scientific Research Center of Radiology and Surgical Technologies, St. Petersburg, Russia; Almazov National Medical Research Center, St. Petersburg, Russia; COBRAIN Center - Brain Research Excellence Center, M Heratsi Yerevan State Medical University, Yerevan, Armenia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia.
| |
Collapse
|
16
|
Ramos A, Joshi RS, Szabo G. Innate immune activation: Parallels in alcohol use disorder and Alzheimer’s disease. Front Mol Neurosci 2022; 15:910298. [PMID: 36157070 PMCID: PMC9505690 DOI: 10.3389/fnmol.2022.910298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Alcohol use disorder is associated with systemic inflammation and organ dysfunction especially in the liver and the brain. For more than a decade, studies have highlighted alcohol abuse-mediated impairment of brain function and acceleration of neurodegeneration through inflammatory mechanisms that directly involve innate immune cells. Furthermore, recent studies indicate overlapping genetic risk factors between alcohol use and neurodegenerative disorders, specifically regarding the role of innate immunity in the pathomechanisms of both areas. Considering the pressing need for a better understanding of the relevance of alcohol abuse in dementia progression, here we summarize the molecular mechanisms of neuroinflammation observed in alcohol abuse and Alzheimer’s disease, the most common cause of dementia. In addition, we highlight mechanisms that are already established in the field of Alzheimer’s disease that may be relevant to explore in alcoholism to better understand alcohol mediated neurodegeneration and dementia, including the relevance of the liver-brain axis.
Collapse
Affiliation(s)
- Adriana Ramos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Radhika S. Joshi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- *Correspondence: Gyongyi Szabo,
| |
Collapse
|
17
|
Lodha J, Brocato E, Wolstenholme JT. Areas of Convergence and Divergence in Adolescent Social Isolation and Binge Drinking: A Review. Front Behav Neurosci 2022; 16:859239. [PMID: 35431830 PMCID: PMC9009335 DOI: 10.3389/fnbeh.2022.859239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Adolescence is a critical developmental period characterized by enhanced social interactions, ongoing development of the frontal cortex and maturation of synaptic connections throughout the brain. Adolescents spend more time interacting with peers than any other age group and display heightened reward sensitivity, impulsivity and diminished inhibitory self-control, which contribute to increased risky behaviors, including the initiation and progression of alcohol use. Compared to adults, adolescents are less susceptible to the negative effects of ethanol, but are more susceptible to the negative effects of stress, particularly social stress. Juvenile exposure to social isolation or binge ethanol disrupts synaptic connections, dendritic spine morphology, and myelin remodeling in the frontal cortex. These structural effects may underlie the behavioral and cognitive deficits seen later in life, including social and memory deficits, increased anxiety-like behavior and risk for alcohol use disorders (AUD). Although the alcohol and social stress fields are actively investigating the mechanisms through which these effects occur, significant gaps in our understanding exist, particularly in the intersection of the two fields. This review will highlight the areas of convergence and divergence in the fields of adolescent social stress and ethanol exposure. We will focus on how ethanol exposure or social isolation stress can impact the development of the frontal cortex and lead to lasting behavioral changes in adulthood. We call attention to the need for more mechanistic studies and the inclusion of the evaluation of sex differences in these molecular, structural, and behavioral responses.
Collapse
Affiliation(s)
- Jyoti Lodha
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Emily Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T. Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
18
|
Miguel-Hidalgo JJ. Astroglia in the Vulnerability and Maintenance of Alcohol Use Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:255-279. [PMID: 34888838 DOI: 10.1007/978-3-030-77375-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes induced in the morphology and the multiplicity of functional roles played by astrocytes in brain regions critical to the establishment and maintenance of alcohol abuse suggest that they make an important contribution to the vulnerability to alcohol use disorders. The understanding of the relevant mechanisms accounting for that contribution is complicated by the fact that alcohol itself acts directly on astrocytes altering their metabolism, gene expression, and plasticity, so that the ultimate result is a complex interaction of various cellular pathways, including intracellular calcium regulation, neuroimmune responses, and regulation of neurotransmitter and gliotransmitter release and uptake. The recent years have seen a steady increase in the characterization of several of the relevant mechanisms, but much remains to be done for a full understanding of the astrocytes' contribution to the vulnerability to alcohol dependence and abuse and for using that knowledge in designing effective therapies for AUDs.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
19
|
Vilpoux C, Fouquet G, Deschamps C, Lefebvre E, Gosset P, Antol J, Zabijak L, Marcq I, Naassila M, Pierrefiche O. Astrogliosis and compensatory neurogenesis after the first ethanol binge drinking-like exposure in the adolescent rat. Alcohol Clin Exp Res 2021; 46:207-220. [PMID: 34862633 DOI: 10.1111/acer.14757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Multiple ethanol binge drinking-like exposures during adolescence in the rat induce neuroinflammation, loss of neurogenesis, and cognitive deficits in adulthood. Interestingly, the first ethanol binge drinking-like exposure during adolescence also induces short- term impairments in cognition and synaptic plasticity in the hippocampus though the cellular mechanisms of these effects are unclear. Here, we sought to determine which of the cellular effects of ethanol might play a role in the disturbances in cognition and synaptic plasticity observed in the adolescent male rat after two binge-like ethanol exposures. METHODS Using immunochemistry, we measured neurogenesis, neuronal loss, astrogliosis, neuroinflammation, and synaptogenesis in the hippocampus of adolescent rats 48 h after two binge-like ethanol exposures (3 g/kg, i.p., 9 h apart). We used flow cytometry to analyze activated microglia and identify the TLR4-expressing cell types. RESULTS We detected increased hippocampal doublecortin immunoreactivity in the subgranular zone (SGZ) of the dentate gyrus (DG), astrogliosis in the SGZ, and a reduced number of mature neurons in the DG and in CA3, suggesting compensatory neurogenesis. Synaptic density decreased in the stratum oriens of CA1 revealing structural plasticity. There was no change in microglial TLR4 expression or in the number of activated microglia, suggesting a lack of neuroinflammatory processes, although neuronal TLR4 was decreased in CA1 and DG. CONCLUSIONS Our findings demonstrate that the cognitive deficits associated with hippocampal synaptic plasticity alterations that we previously characterized 48 h after the first binge-like ethanol exposures are associated with hippocampal structural plasticity, astrogliosis, and decreased neuronal TLR4 expression, but not with microglia reactivity.
Collapse
Affiliation(s)
- Catherine Vilpoux
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Gregory Fouquet
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Chloe Deschamps
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Elise Lefebvre
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Philippe Gosset
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Johann Antol
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Luciane Zabijak
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France.,Plateforme d'Ingénierie Cellulaire & Analyses des Protéines (ICAP), Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Ingrid Marcq
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Mickael Naassila
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Olivier Pierrefiche
- UMR1247 INSERM, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| |
Collapse
|
20
|
Wooden JI, Thompson KR, Guerin SP, Nawarawong NN, Nixon K. Consequences of adolescent alcohol use on adult hippocampal neurogenesis and hippocampal integrity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:281-304. [PMID: 34696876 DOI: 10.1016/bs.irn.2021.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alcohol is the most commonly used drug among adolescents. Their decreased sensitivity to self-regulating cues to stop drinking coincides with an enhanced vulnerability to negative outcomes of excessive drinking. In adolescents, the hippocampus is one brain region that is particularly susceptible to alcohol-induced neurodegeneration. While cell death is causal, alcohol effects on adult neurogenesis also impact hippocampal structure and function. This review describes what little is known about adolescent-specific effects of alcohol on adult neurogenesis and its relationship to hippocampal integrity. For example, alcohol intoxication inhibits neurogenesis persistently in adolescents but produces aberrant neurogenesis after alcohol dependence. Little is known, however, about the role of adolescent-born neurons in hippocampal integrity or the mechanisms of these effects. Understanding the role of neurogenesis in adolescent alcohol use and misuse is critical to our understanding of adolescent susceptibility to alcohol pathology and increased likelihood of developing alcohol problems in adulthood.
Collapse
Affiliation(s)
- J I Wooden
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - K R Thompson
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - S P Guerin
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - N N Nawarawong
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - K Nixon
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
21
|
Kipp BT, Nunes PT, Galaj E, Hitchcock B, Nasra T, Poynor KR, Heide SK, Reitz NL, Savage LM. Adolescent Ethanol Exposure Alters Cholinergic Function and Apical Dendritic Branching Within the Orbital Frontal Cortex. Neuroscience 2021; 473:52-65. [PMID: 34450212 DOI: 10.1016/j.neuroscience.2021.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
During adolescence, heavy binge-like ethanol consumption can lead to frontocortical structural and functional impairments. These impairments are likely driven by adolescence being a critical time point for maturation of brain regions associated with higher-order cognitive functioning. Rodent models of heavy binge-like ethanol exposure show consistent disruptions to the typical development of the prefrontal cortex (PFC). All deep cortical layers receive cholinergic projections that originate from the Nucleus basalis of Meynert (NbM) complex. These cholinergic projections are highly involved in learning, memory, and attention. Adolescent intermittent ethanol exposure (AIE) induces cholinergic dysfunction as a result of an epigenetic suppression of the genes that drive the cholinergic phenotype. The current study used a model of AIE to assess structural and functional changes to the frontal cortex and NbM following binge-like ethanol exposure in adolescence. Western blot analysis revealed long-term disruptions of the cholinergic circuit following AIE: choline acetyltransferase (ChAT) was suppressed in the NbM and vesicular acetylcholine transporter (VAChT) was suppressed in the orbitofrontal cortex (OFC). In vivo microdialysis for acetylcholine efflux during a spatial memory task determined changes in cholinergic modulation within the PFC following AIE. However, AIE spared performance on the spatial memory task and on an operant reversal task. In a second study, Golgi-Cox staining determined that AIE increased apical dendritic complexity in the OFC, with sex influencing whether the increase in branching occurred near or away from the soma. Spine density or maturity was not affected, likely compensating for a disruption in neurotransmitter function following AIE.
Collapse
Affiliation(s)
- B T Kipp
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - P T Nunes
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - E Galaj
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - B Hitchcock
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - T Nasra
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - K R Poynor
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - S K Heide
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - N L Reitz
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - L M Savage
- Department of Psychology, Binghamton University of the State University of New York, New York, USA.
| |
Collapse
|
22
|
Silva-Gotay A, Davis J, Tavares ER, Richardson HN. Alcohol drinking during early adolescence activates microglial cells and increases frontolimbic Interleukin-1 beta and Toll-like receptor 4 gene expression, with heightened sensitivity in male rats compared to females. Neuropharmacology 2021; 197:108698. [PMID: 34252404 PMCID: PMC8552486 DOI: 10.1016/j.neuropharm.2021.108698] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/13/2021] [Accepted: 06/15/2021] [Indexed: 12/31/2022]
Abstract
Adolescent drinking is risky because neural circuits in the frontal lobes are undergoing maturational processes important for cognitive function and behavioral control in adulthood. Previous studies have shown that myelinated axons in the medial prefrontal cortex (mPFC) are particularly sensitive to alcohol drinking, especially in males. Pro-inflammatory mediators like toll-like receptor 4 (TLR4) and interleukin-1 beta (IL1b) have been implicated in alcohol induced-inflammation and demyelination; thus, herein we test the hypothesis that voluntary alcohol drinking early in adolescence elicits a pro-inflammatory state that is more pronounced in the brain of males compared to females. Adolescent male and female Wistar rats self-administered sweetened alcohol or sweetened water from postnatal days 28-42 and separate sets of brains were processed for 1) immunolabeling for ionized calcium-binding adapter molecule 1 to analyze microglial cell morphology, or 2) qPCR analysis of gene expression of pro-inflammatory mediators. Binge drinking alcohol activated microglia in the mPFC and hippocampus of both males and females, suggesting that voluntary alcohol exposure initiates an inflammatory response. Il1b mRNA was upregulated in the mPFC of both sexes. Conversely, Tlr4 mRNA levels were elevated after drinking only in males, which could explain more robust effects of alcohol on myelin in this region in developing males compared to females. Il1b mRNA changes were not observed in the hippocampus, but alcohol elevated Tlr4 mRNA in both sexes, highlighting regional specificity in inflammatory responses to alcohol. Overall, these findings give insight into potential mechanisms by which low-to-moderate voluntary alcohol intake impacts the developing brain. This article is part of the special Issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- Andrea Silva-Gotay
- Neuroscience and Behavior Graduate Program, University of
Massachusetts, Amherst, MA 01003
| | - Jillian Davis
- Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| | - Elizabeth R. Tavares
- Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| | - Heather N. Richardson
- Neuroscience and Behavior Graduate Program, University of
Massachusetts, Amherst, MA 01003,Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| |
Collapse
|
23
|
Flores-Bonilla A, De Oliveira B, Silva-Gotay A, Lucier KW, Richardson HN. Shortening time for access to alcohol drives up front-loading behavior, bringing consumption in male rats to the level of females. Biol Sex Differ 2021; 12:51. [PMID: 34526108 PMCID: PMC8444481 DOI: 10.1186/s13293-021-00395-y] [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: 06/10/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022] Open
Abstract
Background Incentives to promote drinking (“happy hour”) can encourage faster rates of alcohol consumption, especially in women. Sex differences in drinking dynamics may underlie differential health vulnerabilities relating to alcohol in women versus men. Herein, we used operant procedures to model the happy hour effect and gain insight into the alcohol drinking dynamics of male and female rats. Methods Adult male and female Wistar rats underwent operant training to promote voluntary drinking of 10% (w/v) alcohol (8 rats/sex). We tested how drinking patterns changed after manipulating the effort required for alcohol (fixed ratio, FR), as well as the length of time in which rats had access to alcohol (self-administration session length). Rats were tested twice within the 12 h of the dark cycle, first at 2 h (early phase of the dark cycle, “early sessions”) and then again at 10 h into the dark cycle (late phase of the dark cycle, “late sessions”) with an 8-h break between the two sessions in the home cage. Results Adult females consumed significantly more alcohol (g/kg) than males in the 30-min sessions with the FR1 schedule of reinforcement when tested late in the dark cycle. Front-loading of alcohol was the primary factor driving higher consumption in females. Changing the schedule of reinforcement from FR1 to FR3 reduced total consumption. Notably, this manipulation had minimal effect on front-loading behavior in females, whereas front-loading behavior was significantly reduced in males when more effort was required to access alcohol. Compressing drinking access to 15 min to model a happy hour drove up front-loading behavior, generating alcohol drinking patterns in males that were similar to patterns in females (faster drinking and higher intake). Conclusions This strategy could be useful for exploring sex differences in the neural mechanisms underlying alcohol drinking and related health vulnerabilities. Our findings also highlight the importance of the time of testing for detecting sex differences in drinking behavior. Voluntary alcohol drinking is higher in adult female rats compared to adult male rats. This sex difference is most pronounced in the later phase of the dark cycle, and when the operant effort is minimal (when 1 lever press gives 1 reward: fixed ratio 1, FR1). Higher alcohol intake in females is primarily due to “front-loading”, or the rapid consumption of alcohol within the first 5 min of access. Increasing the effort required to obtain alcohol from FR1 to FR3 dampens front-loading drinking behavior, resulting in similar levels of total intake in males and females. Compressing the time of access to 15 min drives up front-loading to such a degree that rats end up consuming more alcohol in total than they do in 30-min sessions. In males, this increase in drinking is large enough that it eliminates the sex difference in total alcohol intake.
Collapse
Affiliation(s)
- Annabelle Flores-Bonilla
- Neuroscience and Behavior Program, The University of Massachusetts Amherst, Amherst, MA, 01003, USA.,Department of Psychological and Brain Sciences, The University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Barbara De Oliveira
- Department of Psychological and Brain Sciences, The University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Andrea Silva-Gotay
- Neuroscience and Behavior Program, The University of Massachusetts Amherst, Amherst, MA, 01003, USA.,Department of Psychological and Brain Sciences, The University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Kyle W Lucier
- Department of Psychological and Brain Sciences, The University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Heather N Richardson
- Neuroscience and Behavior Program, The University of Massachusetts Amherst, Amherst, MA, 01003, USA. .,Department of Psychological and Brain Sciences, The University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
24
|
Guo F, Zhang YF, Liu K, Huang X, Li RX, Wang SY, Wang F, Xiao L, Mei F, Li T. Chronic Exposure to Alcohol Inhibits New Myelin Generation in Adult Mouse Brain. Front Cell Neurosci 2021; 15:732602. [PMID: 34512271 PMCID: PMC8429601 DOI: 10.3389/fncel.2021.732602] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/10/2021] [Indexed: 12/01/2022] Open
Abstract
Chronic alcohol consumption causes cognitive impairments accompanying with white matter atrophy. Recent evidence has shown that myelin dynamics remain active and are important for brain functions in adulthood. For example, new myelin generation is required for learning and memory functions. However, it remains undetermined whether alcohol exposure can alter myelin dynamics in adulthood. In this study, we examine the effect of chronic alcohol exposure on myelin dynamics by using genetic approaches to label newly generated myelin (NG2-CreERt; mT/mG). Our results indicated that alcohol exposure (either 5% or 10% in drinking water) for 3 weeks remarkably reduced mGFP + /NG2- new myelin and mGFP + /CC1 + new oligodendrocytes in the prefrontal cortex and corpus callosum of 6-month-old NG2-CreERt; mT/mG mice as compared to controls without changing the mGFP + /NG2 + oligodendrocyte precursor cells (OPCs) density, suggesting that alcohol exposure may inhibit oligodendrocyte differentiation. In support with these findings, the alcohol exposure did not significantly alter apoptotic cell number or overall MBP expression in the brains. Further, the alcohol exposure decreased the histone deacetylase1 (HDAC1) expression in mGFP + /NG2 + OPCs, implying epigenetic mechanisms were involved in the arrested OPC differentiation. Together, our results indicate that chronic exposure to alcohol can inhibit myelinogenesis in the adult mouse brain and that may contribute to alcohol-related cognitive impairments.
Collapse
Affiliation(s)
- Feng Guo
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China.,The First Camp of Cadet Brigade, School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi-Fan Zhang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China.,The First Camp of Cadet Brigade, School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Kun Liu
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xu Huang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rui-Xue Li
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shu-Yue Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tao Li
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
25
|
Brocato E, Wolstenholme JT. Neuroepigenetic consequences of adolescent ethanol exposure. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:45-84. [PMID: 34696879 DOI: 10.1016/bs.irn.2021.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Adolescence is a critical developmental period characterized by ongoing brain maturation processes including myelination and synaptic pruning. Adolescents experience heightened reward sensitivity, sensation seeking, impulsivity, and diminished inhibitory self-control, which contribute to increased participation in risky behaviors, including the initiation of alcohol use. Ethanol exposure in adolescence alters memory and cognition, anxiety-like behavior, and ethanol sensitivity as well as brain myelination and dendritic spine morphology, with effects lasting into adulthood. Emerging evidence suggests that epigenetic modifications may explain these lasting effects. Focusing on the amygdala, prefrontal cortex and hippocampus, we review studies investigating the epigenetic consequences of adolescent ethanol exposure. Ethanol metabolism globally increases donor substrates for histone acetylation and histone and DNA methylation, and this chapter discusses how this can further impact epigenetic programming of the adolescent brain. Elucidation of the mechanisms through which ethanol can alter the epigenetic code at specific transcripts may provide therapeutic targets for intervention.
Collapse
Affiliation(s)
- Emily Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
26
|
Ghanbarirad M, Hashemi M, Saberi SM, Majd A. Dysregulation of Myt1 expression acts as a potential peripheral biomarker for major depressive disorder and bipolar disorder. J Neurogenet 2021; 35:381-386. [PMID: 34011236 DOI: 10.1080/01677063.2021.1928663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Major depressive disorder (MDD) and bipolar disorder (BPD) are among the most debilitating mental conditions. Diagnostic criteria for MDD include psychological and physical symptoms, such as low mood and changes in appetite or sleep, respectively. BPD in addition to periods of depression represents episodes of mania or hypomania, and elevation in mood and energy levels are associated with this condition. Dysregulation in adult neurogenesis and myelination have been reported in psychiatric disorders. As a key factor in neurogenesis, it was hypothesized that Myt1 gene expression may be altered in these conditions. Using Real-time PCR, Myt1 expression level in 100 MDD patients and 100 BPD patients, compared with healthy control (HC) individuals was evaluated. Results demonstrate significant downregulation of Myt1 in MDD and BPD. Logistic regression analysis and binary classification evaluation reveal potential risk factor and biomarker characteristics of Myt1, respectively. Moreover, forward and backward digit span results denote a significant reduction in the function of working memory (WM) of MDD and BPD subjects. Correlation analysis revealed a significant association between Myt1 downregulation and WM disruption in the affected individuals. In conclusion, due to its altered role in neurogenesis, downregulation of Myt1 can be associated with the pathology of MDD and BPD.
Collapse
Affiliation(s)
- Maryam Ghanbarirad
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Farhikhtegan Medical Convergence Science Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyed Mehdi Saberi
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Ahmad Majd
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
27
|
Pairing Binge Drinking and a High-Fat Diet in Adolescence Modulates the Inflammatory Effects of Subsequent Alcohol Consumption in Mice. Int J Mol Sci 2021; 22:ijms22105279. [PMID: 34067897 PMCID: PMC8157004 DOI: 10.3390/ijms22105279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/15/2021] [Indexed: 12/12/2022] Open
Abstract
Alcohol binge drinking (BD) and poor nutritional habits are two frequent behaviors among many adolescents that alter gut microbiota in a pro-inflammatory direction. Dysbiotic changes in the gut microbiome are observed after alcohol and high-fat diet (HFD) consumption, even before obesity onset. In this study, we investigate the neuroinflammatory response of adolescent BD when combined with a continuous or intermittent HFD and its effects on adult ethanol consumption by using a self-administration (SA) paradigm in mice. The inflammatory biomarkers IL-6 and CX3CL1 were measured in the striatum 24 h after BD, 3 weeks later and after the ethanol (EtOH) SA. Adolescent BD increased alcohol consumption in the oral SA and caused a greater motivation to seek the substance. Likewise, mice with intermittent access to HFD exhibited higher EtOH consumption, while the opposite effect was found in mice with continuous HFD access. Biochemical analyses showed that after BD and three weeks later, striatal levels of IL-6 and CX3CL1 were increased. In addition, in saline-treated mice, CX3CL1 was increased after continuous access to HFD. After oral SA procedure, striatal IL-6 was increased only in animals exposed to BD and HFD. In addition, striatal CX3CL1 levels were increased in all BD- and HFD-exposed groups. Overall, our findings show that adolescent BD and intermittent HFD increase adult alcohol intake and point to neuroinflammation as an important mechanism modulating this interaction.
Collapse
|
28
|
Liu W, Rohlman AR, Vetreno R, Crews FT. Expression of Oligodendrocyte and Oligoprogenitor Cell Proteins in Frontal Cortical White and Gray Matter: Impact of Adolescent Development and Ethanol Exposure. Front Pharmacol 2021; 12:651418. [PMID: 34025418 PMCID: PMC8134748 DOI: 10.3389/fphar.2021.651418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Adolescent development of prefrontal cortex (PFC) parallels maturation of executive functions as well as increasing white matter and myelination. Studies using MRI and other methods find that PFC white matter increases across adolescence into adulthood in both humans and rodents. Adolescent binge drinking is common and has been found to alter adult behaviors and PFC functions. This study examines development of oligoprogenitor (OPC) and oligodendrocytes (OLs) in Wistar rats from adolescence to adulthood within PFC white matter, corpus callosum forceps minor (fmi), PFC gray matter, and the neurogenic subventricular zone (SVZ) using immunohistochemistry for marker proteins. In addition, the effects of adolescent intermittent ethanol exposure [AIE; 5.0 g/kg/day, intragastric, 2 days on/2 days off on postnatal day (P)25-54], which is a weekend binge drinking model, were determined. OPC markers NG2+, PDGFRα+ and Olig2+IHC were differentially impacted by both age and PFC region. In both fmi and SVZ, NG2+IHC cells declined from adolescence to adulthood with AIE increasing adult NG2+IHC cells and their association with microglial marker Iba1. PFC gray matter decline in NG2+IHC in adulthood was not altered by AIE. Both adult maturation and AIE impacted OL expression of PLP+, MBP+, MAG+, MOG+, CNPase+, Olig1+, and Olig2+IHC in all three PFC regions, but in region- and marker-specific patterns. These findings are consistent with PFC region-specific changes in OPC and OL markers from adolescence to adulthood as well as following AIE that could contribute to lasting changes in PFC function.
Collapse
Affiliation(s)
| | | | | | - Fulton T. Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
29
|
Jones SA, Kliamovich D, Nagel BJ. Sex hormones partially explain the sex-dependent effect of lifetime alcohol use on adolescent white matter microstructure. Psychiatry Res Neuroimaging 2021; 307:111230. [PMID: 33271433 PMCID: PMC7775887 DOI: 10.1016/j.pscychresns.2020.111230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 11/17/2022]
Abstract
Previous studies demonstrate profound sex-specific patterns of white matter microstructural neurodevelopment (i.e. fractional anisotropy; FA, and mean diffusivity; MD) during adolescence. While alcohol use has been associated with alterations in FA and MD, no studies have addressed the potential for sex-specific, alcohol-dose-dependent effects, during development. This prospective longitudinal study (2-4 visits, 310 total scans) used voxel-wise multilevel modeling, in 132 (68 female) adolescents (ages 12-21), to assess the sex-specific effects of lifetime alcohol use on FA and MD, during development. Follow-up analyses tested the role of sex hormones, testosterone and estradiol, in explaining the effects of alcohol use on FA and MD. In the splenium of the corpus callosum and posterior thalamic radiation, male adolescents demonstrated lower FA and greater MD as a function of more lifetime alcohol use, while female adolescents demonstrated the opposite. Further, significant associations between sex hormones and FA/MD partially explained the effect of alcohol use on FA and MD in male adolescents. These results provide evidence for sex-specific and dose-related effects of alcohol use on white matter microstructure, which are partially explained by sex hormones, and highlight the importance of studying sex and hormones when investigating the effects of alcohol use on the adolescent brain.
Collapse
Affiliation(s)
- Scott A Jones
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
| | - Dakota Kliamovich
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Bonnie J Nagel
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
30
|
Salmanzadeh H, Ahmadi-Soleimani SM, Azadi M, Halliwell RF, Azizi H. Adolescent Substance Abuse, Transgenerational Consequences and Epigenetics. Curr Neuropharmacol 2021; 19:1560-1569. [PMID: 33655865 PMCID: PMC8762180 DOI: 10.2174/1570159x19666210303121519] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/13/2021] [Accepted: 02/12/2021] [Indexed: 11/22/2022] Open
Abstract
Adolescence is the transitional period between childhood and adulthood and a critical period in brain development. Adolescence in humans is also associated with increased expression of risk-taking behaviors. Epidemiological and clinical studies, for example, show a surge of drug abuse and raise the hypothesis that the adolescent brain undergoes critical changes resulting in diminished control. Determining how substance abuse during this critical period might cause longterm neurobiological changes in cognition and behavior is therefore critically important. The present work aims to provide an evaluation of the transgenerational and multi-generational phenotypes derived from parent animals exposed to drugs of abuse only during their adolescence. Specifically, we will consider changes found following the administration of cannabinoids, nicotine, alcohol and opiates. In addition, epigenetic modifications of the genome following drug exposure will be discussed as emerging evidence of the underlying adverse transgenerational effects. Notwithstanding, much of the new data discussed here is from animal models, indicating that future clinical studies are much needed to better understand the neurobiological consequences and mechanisms of drug actions on the human brains' development and maturation.
Collapse
Affiliation(s)
| | | | | | - Robert F. Halliwell
- Address correspondence to this author at the TJ Long School of Pharmacy, University of the Pacific, Stockton, California, USA; Tel: +1 (209) 946 2074; E-mail: and Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Tel: +98-21-82884587; Fax: +98-21-82884528; E-mail:
| | - Hossein Azizi
- Address correspondence to this author at the TJ Long School of Pharmacy, University of the Pacific, Stockton, California, USA; Tel: +1 (209) 946 2074; E-mail: and Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Tel: +98-21-82884587; Fax: +98-21-82884528; E-mail:
| |
Collapse
|
31
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
32
|
Cortez I, Rodgers SP, Kosten TA, Leasure JL. Sex and Age Effects on Neurobehavioral Toxicity Induced by Binge Alcohol. Brain Plast 2020; 6:5-25. [PMID: 33680843 PMCID: PMC7902983 DOI: 10.3233/bpl-190094] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Historically, most alcohol neurotoxicity studies were conducted in young adult males and focused on chronic intake. There has been a shift towards studying the effects of alcohol on the adolescent brain, due to alcohol consumption during this formative period disrupting the brain's developmental trajectory. Because the most typical pattern of adolescent alcohol intake is heavy episodic (binge) drinking, there has also been a shift towards the study of binge alcohol-induced neurobehavioral toxicity. It has thus become apparent that binge alcohol damages the adolescent brain and there is increasing attention to sex-dependent effects. Significant knowledge gaps remain in our understanding of the effects of binge alcohol on the female brain, however. Moreover, it is unsettling that population-level studies indicate that the prevalence of binge drinking is increasing among American women, particularly those in older age groups. Although study of adolescents has made it apparent that binge alcohol disrupts ongoing brain maturational processes, we know almost nothing about how it impacts the aging brain, as studies of its effects on the aged brain are relatively scarce, and the study of sex-dependent effects is just beginning. Given the rapidly increasing population of older Americans, it is crucial that studies address age-dependent effects of binge alcohol, and given the increase in binge drinking in older women who are at higher risk for cognitive decline relative to men, studies must encompass both sexes. Because adolescence and older age are both characterized by age-typical brain changes, and because binge drinking is the most common pattern of alcohol intake in both age groups, the knowledge that we have amassed on binge alcohol effects on the adolescent brain can inform our study of its effects on the aging brain. In this review, we therefore cover the current state of knowledge of sex and age-dependent effects of binge alcohol, as well as statistical and methodological considerations for studies aimed at addressing them.
Collapse
Affiliation(s)
- Ibdanelo Cortez
- Department of Psychology, University of Houston, Houston, TX, USA
| | | | | | - J. Leigh Leasure
- Department of Psychology, University of Houston, Houston, TX, USA
- Department of Biology & Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
33
|
Drzewiecki CM, Juraska JM. The structural reorganization of the prefrontal cortex during adolescence as a framework for vulnerability to the environment. Pharmacol Biochem Behav 2020; 199:173044. [DOI: 10.1016/j.pbb.2020.173044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/04/2020] [Accepted: 09/30/2020] [Indexed: 11/26/2022]
|
34
|
Wukitsch TJ, Moser TJ, Brase EC, Kiefer SW, Cain ME. Adolescent ethanol exposure and differential rearing environment affect taste reactivity to ethanol in rats. Alcohol 2020; 89:113-122. [PMID: 32937167 DOI: 10.1016/j.alcohol.2020.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/20/2022]
Abstract
The identification and characterization of variables that influence "liking" and enhance vulnerability to repeated alcohol use are vital to understanding and treating alcohol use disorders. In the current study, we explore the influence of rearing environment and experimenter-administered adolescent ethanol on the hedonic value of ethanol, sucrose, and quinine. Male and female rats were reared for 30 days starting at postnatal day (PND) 21 in either an enriched, isolated, or standard condition and received 1.5 g/kg (intraperitoneally [i.p.]) 20% (w/v) ethanol or saline every other day for 12 days starting at PND 28. Thereafter, all rats had indwelling intraoral fistulae implanted and their taste reactivities to water, ethanol (5, 10, 20, 30, 40% v/v), sucrose (0.1, 0.25, 0.5 M), and quinine (0.1, 0.5 mM) were recorded and analyzed. Results indicated that enrichment elevated hedonic responding to sucrose compared to isolation, and induced a stronger negative relationship between hedonic responding and ethanol concentration compared to standard conditions. Enrichment also elevated aversive responding to ethanol and quinine compared to both isolated and standard condition rats. Adolescent ethanol injections marginally reduced aversive responding to quinine. These results replicate previous findings that environmental enrichment enhances both "liking" and aversion. In addition, the current findings suggest that, while adolescent ethanol injections may blunt aversive responses to quinine, they have no effect on aversive or hedonic responding to ethanol or sucrose. Together with existing literature, our results may suggest that experience with the taste of ethanol is necessary for alterations to ethanol "liking" and aversion.
Collapse
|
35
|
Hua JPY, Sher KJ, Boness CL, Trela CJ, McDowell YE, Merrill AM, Piasecki TM, Kerns JG. Prospective Study Examining the Effects of Extreme Drinking on Brain Structure in Emerging Adults. Alcohol Clin Exp Res 2020; 44:2200-2211. [PMID: 32970324 PMCID: PMC7680366 DOI: 10.1111/acer.14446] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Emerging adulthood is a critical neurodevelopment period in which extreme drinking has a potentially pronounced neurotoxic effect. Therefore, extreme drinking, even a single episode, could be particularly harmful to the developing brain's structure. Relatedly, heavy alcohol use in emerging adults has been associated with structural brain damage, especially in the corpus callosum. However, it is unclear whether and how much a single extreme drinking episode would affect brain morphometry. METHODS For the first time in the literature, the current study prospectively examined the impact of an extreme drinking episode (i.e., twenty-first birthday celebration) on the brain morphometry of emerging adults immediately following their birthday celebration (n = 50) and approximately 5 weeks post-birthday celebration (n = 29). RESULTS We found evidence that a single extreme drinking episode was associated with structural changes immediately post-birthday celebration. Specifically, higher twenty-first birthday estimated blood-alcohol concentration was associated with decreased volume of the posterior and central corpus callosum immediately post-birthday celebration. This extreme drinking episode was not associated with further structural changes, or recovery, 5 weeks post-twenty-first birthday celebration. CONCLUSIONS Overall, results suggest that a single episode of heavy drinking in emerging adulthood may be associated with immediate structural changes of the corpus callosum. Thus, emerging adulthood, which is characterized by high rates of extreme drinking, could be a critical period for targeted prevention and intervention.
Collapse
Affiliation(s)
- Jessica P. Y. Hua
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211,San Francisco VA Medical Center, San Francisco, CA 94121
| | - Kenneth J. Sher
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211
| | - Cassandra L. Boness
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211
| | - Constantine J. Trela
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211
| | - Yoanna E. McDowell
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211
| | - Anne M. Merrill
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211
| | - Thomas M. Piasecki
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211
| | - John G. Kerns
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211,To whom correspondence should be addressed: John G. Kerns, tel: 573-882-6860, fax: 573-882-7710,
| |
Collapse
|
36
|
Abstract
Sex differences may play a critical role in modulating how chronic or heavy alcohol use impacts the brain to cause the development of alcohol use disorder (AUD). AUD is a multifaceted and complex disorder driven by changes in key neurobiological structures that regulate executive function, memory, and stress. A three-stage framework of addiction (binge/intoxication; withdrawal/negative affect; preoccupation/anticipation) has been useful for conceptualizing the complexities of AUD and other addictions. Initially, alcohol drinking causes short-term effects that involve signaling mediated by several neurotransmitter systems such as dopamine, corticotropin releasing factor, and glutamate. With continued intoxication, alcohol leads to dysfunctional behaviors that are thought to be due in part to alterations of these and other neurotransmitter systems, along with alterations in neural pathways connecting prefrontal and limbic structures. Using the three-stage framework, this review highlights examples of research examining sex differences in drinking and differential modulation of neural systems contributing to the development of AUD. New insights addressing the role of sex differences in AUD are advancing the field forward by uncovering the complex interactions that mediate vulnerability.
Collapse
Affiliation(s)
| | - Heather N Richardson
- Department of Psychological and Brain Sciences at the University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
37
|
Joffe ME, Winder DG, Conn PJ. Contrasting sex-dependent adaptations to synaptic physiology and membrane properties of prefrontal cortex interneuron subtypes in a mouse model of binge drinking. Neuropharmacology 2020; 178:108126. [PMID: 32781000 DOI: 10.1016/j.neuropharm.2020.108126] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/18/2020] [Accepted: 04/29/2020] [Indexed: 12/24/2022]
Abstract
Alcohol use disorder (AUD) affects all sexes, however women who develop AUD may be particularly susceptible to cravings and other components of the disease. While many brain regions are involved in AUD etiology, proper prefrontal cortex (PFC) function is particularly important for top-down craving management and the moderation of drinking behaviors. Essential regulation of PFC output is provided by local inhibitory interneurons, yet how drinking affects interneuron physiology remains poorly understood, particularly in female individuals. To address this gap, we generated fluorescent reporter transgenic mice to label the two major classes of interneuron in deep layer prelimbic PFC, based on expression of parvalbumin (PV-IN) or somatostatin (SST-IN). We then interrogated PV-IN and SST-IN membrane and synaptic physiology in a rodent model of binge drinking. Beginning in late adolescence, mice received 3-4 weeks of intermittent access (IA) ethanol. We prepared acute brain slices one day after the last drinking session. PV-INs but not SST-INs from IA ethanol mice displayed increased excitability relative to controls, regardless of sex. On the contrary, synaptic adaptations to PV-INs differed based on sex. While drinking decreased excitatory synaptic strength onto PV-INs from female mice, PV-INs from IA ethanol male mice exhibited potentiated excitatory transmission relative to controls. In contrast, decreased synaptic strength onto SST-INs was observed following IA ethanol in all groups of mice. Together, these findings illustrate novel sex differences in drinking-related PFC pathophysiology. Discovering means to restore PV-IN and SST-IN dysfunction following extended drinking provides opportunities for developing new treatments for all AUD patients.
Collapse
Affiliation(s)
- Max E Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA.
| | - Danny G Winder
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA
| |
Collapse
|
38
|
Adrienne McGinn M, Edwards KN, Edwards S. Chronic inflammatory pain alters alcohol-regulated frontocortical signaling and associations between alcohol drinking and thermal sensitivity. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100052. [PMID: 33005820 PMCID: PMC7509777 DOI: 10.1016/j.ynpai.2020.100052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022]
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing psychiatric disorder that is characterized by the emergence of negative affective states. The transition from recreational, limited intake to uncontrolled, escalated intake is proposed to involve a transition from positive to negative reinforcement mechanisms for seeking alcohol. Past work has identified the emergence of significant hyperalgesia/allodynia in alcohol-dependent animals, which may serve as a key negative reinforcement mechanism. Chronic pain has been associated with enhanced extracellular signal-regulated kinase (ERK) activity in cortical and subcortical nociceptive areas. Additionally, both pain and AUD have been associated with increased activity of the glucocorticoid receptor (GR), a key mediator of stress responsiveness. The objectives of the current study were to first determine relationships between thermal nociceptive sensitivity and alcohol drinking in male Wistar rats. While inflammatory pain induced by complete Freund's adjuvant (CFA) administration did not modify escalation of home cage drinking in animals over four weeks, the relationship between drinking levels and hyperalgesia symptoms reversed between acute (1 week) and chronic (3-4 week) periods post-CFA administration, suggesting that either the motivational or analgesic effects of alcohol may be altered over the time course of chronic pain. We next examined ERK and GR phosphorylation in pain-related brain areas (including the central amygdala and prefrontal cortex subregions) in animals experiencing acute withdrawal from binge alcohol administration (2 g/kg, 6 h withdrawal) and CFA administration (four weeks) to model the neurobiological consequences of binge alcohol exposure in the context of pain. We observed a significant interaction between alcohol and pain state, whereby alcohol withdrawal increased ERK phosphorylation across all four frontocortical areas examined, although this effect was absent in animals experiencing chronic inflammatory pain. Alcohol withdrawal also increased GR phosphorylation across all four frontocortical areas, but these changes were not altered by CFA. Interestingly, we observed significant inter-brain regional correlations in GR phosphorylation between the insula and other regions investigated only in animals exposed to both alcohol and CFA, suggesting coordinated activity in insula circuitry and glucocorticoid signaling in this context. The results of these studies provide a greater understanding of the neurobiology of AUD and will contribute to the development of effective treatment strategies for comorbid AUD and pain.
Collapse
Affiliation(s)
- M. Adrienne McGinn
- Neurobiology of Addiction Section, National Institute on Drug Abuse IRP, United States
| | - Kimberly N. Edwards
- Department of Physiology, LSU Health-New Orleans, United States
- Alcohol & Drug Abuse Center of Excellence, LSU Health-New Orleans, United States
| | - Scott Edwards
- Department of Physiology, LSU Health-New Orleans, United States
- Alcohol & Drug Abuse Center of Excellence, LSU Health-New Orleans, United States
- Neuroscience Center of Excellence, LSU Health-New Orleans, United States
- Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health-New Orleans, United States
| |
Collapse
|
39
|
Buján GE, Serra HA, Molina SJ, Guelman LR. Oxidative Stress-Induced Brain Damage Triggered by Voluntary Ethanol Consumption during Adolescence: A Potential Target for Neuroprotection? Curr Pharm Des 2020; 25:4782-4790. [PMID: 31814553 DOI: 10.2174/1381612825666191209121735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/23/2019] [Indexed: 12/28/2022]
Abstract
Alcohol consumption, in particular ethanol (EtOH), typically begins in human adolescence, often in a "binge like" manner. However, although EtOH abuse has a high prevalence at this stage, the effects of exposure during adolescence have been less explored than prenatal or adult age exposure. Several authors have reported that EtOH intake during specific periods of development might induce brain damage. Although the mechanisms are poorly understood, it has been postulated that oxidative stress may play a role. In fact, some of these studies revealed a decrease in brain antioxidant enzymes' level and/or an increase in reactive oxygen species (ROS) production. Nevertheless, although existing literature shows a number of studies in which ROS were measured in developing animals, fewer reported the measurement of ROS levels after EtOH exposure in adolescence. Importantly, neuroprotective agents aimed to these potential targets may be relevant tools useful to reduce EtOH-induced neurodegeneration, restore cognitive function and improve treatment outcomes for alcohol use disorders (AUDs). The present paper reviews significant evidences about the mechanisms involved in EtOH-induced brain damage, as well as the effect of different potential neuroprotectants that have shown to be able to prevent EtOH-induced oxidative stress. A selective inhibitor of the endocannabinoid anandamide metabolism, a flavonol present in different fruits (quercetin), an antibiotic with known neuroprotective properties (minocycline), a SOD/catalase mimetic, a potent antioxidant and anti-inflammatory molecule (resveratrol), a powerful ROS scavenger (melatonin), an isoquinoline alkaloid (berberine), are some of the therapeutic strategies that could have some clinical relevance in the treatment of AUDs. As most of these works were performed in adult animal models and using EtOH-forced paradigms, the finding of neuroprotective tools that could be effective in adolescent animal models of voluntary EtOH intake should be encouraged.
Collapse
Affiliation(s)
- Gustavo E. Buján
- Universidad de Buenos Aires, Facultad de Medicina, 1 Cátedra de Farmacología, Buenos Aires, Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBACONICET), Facultad de Medicina, Buenos Aires, Argentina
| | - Hector A. Serra
- Universidad de Buenos Aires, Facultad de Medicina, 1 Cátedra de Farmacología, Buenos Aires, Argentina
| | - Sonia J. Molina
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBACONICET), Facultad de Medicina, Buenos Aires, Argentina
| | - Laura R. Guelman
- Universidad de Buenos Aires, Facultad de Medicina, 1 Cátedra de Farmacología, Buenos Aires, Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBACONICET), Facultad de Medicina, Buenos Aires, Argentina
| |
Collapse
|
40
|
Galaj E, Barrera E, Morris D, Ma YY, Ranaldi R. Aberrations in Incentive Learning and Responding to Heroin in Male Rats After Adolescent or Adult Chronic Binge-Like Alcohol Exposure. Alcohol Clin Exp Res 2020; 44:1214-1223. [PMID: 32311102 PMCID: PMC7313436 DOI: 10.1111/acer.14341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/08/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE Binge drinking is a serious problem among adolescents and young adults despite its adverse consequences on the brain and behavior. One area that remains poorly understood concerns the impact of chronic intermittent ethanol (CIE) exposure on incentive learning. METHODS Here, we examined the effects of CIE exposure during different developmental stages on conditioned approach and conditioned reward learning in rats experiencing acute or protracted withdrawal from alcohol. Two or 21 days after adolescent or adult CIE exposure, male rats were exposed to pairings of a light stimulus (CS) and food pellets for 3 consecutive daily sessions (30 CS-food pellet pairings per session). This was followed by conditioned approach testing measuring responses (food trough head entries) to the CS-only presentations and by conditioned reward testing measuring responses on a lever producing the CS and on another producing a tone. We then measured behavioral sensitization to repeated injections of heroin (2 mg/kg/d for 9 days). RESULTS Adolescent and adult alcohol-treated rats showed significantly impaired conditioned reward learning regardless of withdrawal period (acute or prolonged). We found no evidence of changes to conditioned approach learning after adolescent or adult exposure to CIE. Finally, in addition to producing long-term impairments in incentive learning, CIE exposure enhanced locomotor activity in response to heroin and had no effect on behavioral sensitization to heroin regardless of age and withdrawal period. CONCLUSIONS Our work sets a framework for identifying CIE-induced alterations in incentive learning and inducing susceptibility to subsequent opioid effects.
Collapse
Affiliation(s)
- Ewa Galaj
- National Institute on Drug Abuse Intramural Research Program, Molecular Targets and Medication Discovery Branch, 251 Bayview Blvd, Baltimore, MD, 21224, US
| | - Eddy Barrera
- Queens College, City University of New York, Department of Psychology, 65-30 Kissena Blvd., Flushing, NY, 11367, US
| | - Debra Morris
- Queens College, City University of New York, Department of Psychology, 65-30 Kissena Blvd., Flushing, NY, 11367, US
| | - Yao-Ying Ma
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, 635 Barnhill Drive, Indianapolis, IN, 46202, US
| | - Robert Ranaldi
- Queens College, City University of New York, Department of Psychology, 65-30 Kissena Blvd., Flushing, NY, 11367, US
| |
Collapse
|
41
|
Smith ML, Lopez MF, Wolen AR, Becker HC, Miles MF. Brain regional gene expression network analysis identifies unique interactions between chronic ethanol exposure and consumption. PLoS One 2020; 15:e0233319. [PMID: 32469986 PMCID: PMC7259766 DOI: 10.1371/journal.pone.0233319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/01/2020] [Indexed: 11/28/2022] Open
Abstract
Progressive increases in ethanol consumption is a hallmark of alcohol use disorder (AUD). Persistent changes in brain gene expression are hypothesized to underlie the altered neural signaling producing abusive consumption in AUD. To identify brain regional gene expression networks contributing to progressive ethanol consumption, we performed microarray and scale-free network analysis of expression responses in a C57BL/6J mouse model utilizing chronic intermittent ethanol by vapor chamber (CIE) in combination with limited access oral ethanol consumption. This model has previously been shown to produce long-lasting increased ethanol consumption, particularly when combining oral ethanol access with repeated cycles of intermittent vapor exposure. The interaction of CIE and oral consumption was studied by expression profiling and network analysis in medial prefrontal cortex, nucleus accumbens, hippocampus, bed nucleus of the stria terminalis, and central nucleus of the amygdala. Brain region expression networks were analyzed for ethanol-responsive gene expression, correlation with ethanol consumption and functional content using extensive bioinformatics studies. In all brain-regions studied the largest number of changes in gene expression were seen when comparing ethanol naïve mice to those exposed to CIE and drinking. In the prefrontal cortex, however, unique patterns of gene expression were seen compared to other brain-regions. Network analysis identified modules of co-expressed genes in all brain regions. The prefrontal cortex and nucleus accumbens showed the greatest number of modules with significant correlation to drinking behavior. Across brain-regions, however, many modules with strong correlations to drinking, both baseline intake and amount consumed after CIE, showed functional enrichment for synaptic transmission and synaptic plasticity.
Collapse
Affiliation(s)
- Maren L. Smith
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Marcelo F. Lopez
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Aaron R. Wolen
- Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Howard C. Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, United States of America
- RHJ Department of Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
| | - Michael F. Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
42
|
Lees B, Meredith LR, Kirkland AE, Bryant BE, Squeglia LM. Effect of alcohol use on the adolescent brain and behavior. Pharmacol Biochem Behav 2020; 192:172906. [PMID: 32179028 PMCID: PMC7183385 DOI: 10.1016/j.pbb.2020.172906] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/31/2020] [Accepted: 03/12/2020] [Indexed: 12/18/2022]
Abstract
Adolescence is a particularly vulnerable neurodevelopmental period marked by high rates of engagement with risky alcohol use. This review summarizes the cognitive and neural consequences following alcohol use during adolescence from longitudinal design studies in humans and animals. Findings from human adolescent studies suggest that binge drinking and heavy alcohol use is associated with poorer cognitive functioning on a broad range of neuropsychological assessments, including learning, memory, visuospatial functioning, psychomotor speed, attention, executive functioning, and impulsivity. Alcohol use during adolescence is associated with accelerated decreases in gray matter and attenuated increases in white matter volume, and aberrant neural activity during executive functioning, attentional control, and reward sensitivity tasks, when compared to non-drinking adolescents. Animal studies in rodents and non-human primates have replicated human findings, and suggest cognitive and neural consequences of adolescent alcohol use may persist into adulthood. Novel rodent studies demonstrate that adolescent alcohol use may increase reward responsiveness of the dopamine system to alcohol later in life, as well as disrupt adolescent neurogenesis, potentially through neuroinflammation, with long-lasting neural and behavioral effects into adulthood. Larger longitudinal human cognitive and neuroimaging studies with more diverse samples are currently underway which will improve understanding of the impact of polysubstance use, as well as the interactive effects of substance use, physical and mental health, and demographic factors on cognition and neurodevelopment.
Collapse
Affiliation(s)
- Briana Lees
- The Matilda Centre for Research in Mental Health and Substance Use, University of Sydney, Australia.
| | - Lindsay R Meredith
- University of California, Los Angeles, Department of Psychology, United States of America
| | - Anna E Kirkland
- American University, Department of Psychology, United States of America
| | - Brittany E Bryant
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, United States of America
| | - Lindsay M Squeglia
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, United States of America
| |
Collapse
|
43
|
Brain-wide functional architecture remodeling by alcohol dependence and abstinence. Proc Natl Acad Sci U S A 2020; 117:2149-2159. [PMID: 31937658 DOI: 10.1073/pnas.1909915117] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Alcohol abuse and alcohol dependence are key factors in the development of alcohol use disorder, which is a pervasive societal problem with substantial economic, medical, and psychiatric consequences. Although our understanding of the neurocircuitry that underlies alcohol use has improved, novel brain regions that are involved in alcohol use and novel biomarkers of alcohol use need to be identified. The present study used a single-cell whole-brain imaging approach to 1) assess whether abstinence from alcohol in an animal model of alcohol dependence alters the functional architecture of brain activity and modularity, 2) validate our current knowledge of the neurocircuitry of alcohol abstinence, and 3) discover brain regions that may be involved in alcohol use. Alcohol abstinence resulted in the whole-brain reorganization of functional architecture in mice and a pronounced decrease in modularity that was not observed in nondependent moderate drinkers. Structuring of the alcohol abstinence network revealed three major brain modules: 1) extended amygdala module, 2) midbrain striatal module, and 3) cortico-hippocampo-thalamic module, reminiscent of the three-stage theory. Many hub brain regions that control this network were identified, including several that have been previously overlooked in alcohol research. These results identify brain targets for future research and demonstrate that alcohol use and dependence remodel brain-wide functional architecture to decrease modularity. Further studies are needed to determine whether the changes in coactivation and modularity that are associated with alcohol abstinence are causal features of alcohol dependence or a consequence of excessive drinking and alcohol exposure.
Collapse
|
44
|
Boda E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress‐related disorders. Eur J Neurosci 2019; 53:281-297. [DOI: 10.1111/ejn.14621] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/06/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi‐Montalcini University of Turin Turin Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO) University of Turin Turin Italy
| |
Collapse
|
45
|
Rice J, Coutellier L, Weiner JL, Gu C. Region-specific interneuron demyelination and heightened anxiety-like behavior induced by adolescent binge alcohol treatment. Acta Neuropathol Commun 2019; 7:173. [PMID: 31703603 DOI: 10.1186/s40478-019-0829-9] [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: 08/23/2019] [Accepted: 10/13/2019] [Indexed: 12/12/2022] Open
Abstract
Adolescent binge drinking represents a major public health challenge and can lead to persistent neurological and mental conditions, but the underlying pathogenic mechanisms remain poorly understood. Using a mouse model of adolescent binge ethanol treatment (ABET), we found that this treatment induced behavioral changes associated with demyelination in different brain regions. After ABET, adolescent mice exhibited anxiogenic behaviors with no change in locomotion on the elevated plus maze, and impaired spatial memory indicated by a significant reduction in spontaneous alternation in the Y maze test. Both effects persisted into adulthood. Anatomical studies further showed that ABET induced a significant reduction of parvalbumin-positive (PV+) GABAergic interneurons and myelin density in the hippocampus and medial prefrontal cortex (mPFC). While these deficits in PV+ interneurons and myelin persisted into early adulthood in the hippocampus, the myelin density recovered in the mPFC. Moreover, whereas ABET mainly damaged myelin of PV+ axons in the hippocampus, it primarily damaged myelin of PV-negative axons in the mPFC. Thus, our findings reveal that an adolescent binge alcohol treatment regimen disrupts spatial working memory, increases anxiety-like behaviors, and exerts unique temporal and spatial patterns of gray matter demyelination in the hippocampus and mPFC.
Collapse
|
46
|
Abstract
There are vast literatures on the neural effects of alcohol and the neural effects of exercise. Simply put, exercise is associated with brain health, alcohol is not, and the mechanisms by which exercise benefits the brain directly counteract the mechanisms by which alcohol damages it. Although a degree of brain recovery naturally occurs upon cessation of alcohol consumption, effective treatments for alcohol-induced brain damage are badly needed, and exercise is an excellent candidate from a mechanistic standpoint. In this chapter, we cover the small but growing literature on the interactive neural effects of alcohol and exercise, and the capacity of exercise to repair alcohol-induced brain damage. Increasingly, exercise is being used as a component of treatment for alcohol use disorders (AUD), not because it reverses alcohol-induced brain damage, but because it represents a rewarding, alcohol-free activity that could reduce alcohol cravings and improve comorbid conditions such as anxiety and depression. It is important to bear in mind, however, that multiple studies attest to a counterintuitive positive relationship between alcohol intake and exercise. We therefore conclude with cautionary notes regarding the use of exercise to repair the brain after alcohol damage.
Collapse
|
47
|
Internet Addiction Among Lebanese Adolescents: The Role of Self-Esteem, Anger, Depression, Anxiety, Social Anxiety and Fear, Impulsivity, and Aggression-A Cross-Sectional Study. J Nerv Ment Dis 2019; 207:838-846. [PMID: 31503174 DOI: 10.1097/nmd.0000000000001034] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The study objective was to evaluate the association between depression, anxiety, social anxiety and fear, impulsivity, and aggression and Internet addiction (IA) among Lebanese adolescents. This cross-sectional study, conducted between October 2017 and April 2018, enrolled 1103 young adolescents aged between 13 and 17 years. The Internet Addiction Test (IAT) was used to screen for IA. The results also showed that 56.4% of the participants were average Internet users (IAT score ≤49), 40.0% had occasional/frequent problems (IAT scores between 50 and 79), and 3.6% had significant problems (IAT scores ≥80) because of Internet use. The results of a stepwise regression showed that higher levels of aggression (β = 0.185), depression (Multiscore Depression Inventory for Children) (β = 0.219), impulsivity (β = 0.344), and social fear (β = 0.084) were associated with higher IA, whereas an increased number of siblings (β = -0.779) and a higher socioeconomic status (β = -1.707) were associated with lower IA. Uncontrolled use of the Internet can be associated with addiction and other psychological comorbidities.
Collapse
|
48
|
Bogenpohl JW, Smith ML, Farris SP, Dumur CI, Lopez MF, Becker HC, Grant KA, Miles MF. Cross-Species Co-analysis of Prefrontal Cortex Chronic Ethanol Transcriptome Responses in Mice and Monkeys. Front Mol Neurosci 2019; 12:197. [PMID: 31456662 PMCID: PMC6701453 DOI: 10.3389/fnmol.2019.00197] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Despite recent extensive genomic and genetic studies on behavioral responses to ethanol, relatively few new therapeutic targets for the treatment of alcohol use disorder have been validated. Here, we describe a cross-species genomic approach focused on identifying gene networks associated with chronic ethanol consumption. To identify brain mechanisms underlying a chronic ethanol consumption phenotype highly relevant to human alcohol use disorder, and to elucidate potential future therapeutic targets, we conducted a genomic study in a non-human primate model of chronic open-access ethanol consumption. Microarray analysis of RNA expression in anterior cingulate and subgenual cortices from rhesus macaques was performed across multiple cohorts of animals. Gene networks correlating with ethanol consumption or showing enrichment for ethanol-regulated genes were identified, as were major ethanol-related hub genes within these networks. A subsequent consensus module analysis was used to co-analyze monkey data with expression data from a chronic intermittent ethanol vapor-exposure and consumption model in C57BL/6J mice. Ethanol-related gene networks conserved between primates and rodents were enriched for genes involved in discrete biological functions, including; myelination, synaptic transmission, chromatin modification, Golgi apparatus function, translation, cellular respiration, and RNA processing. The myelin-related network, in particular, showed strong correlations with ethanol consumption behavior and displayed marked network reorganization between control and ethanol-drinking animals. Further bioinformatics analysis revealed that these networks also showed highly significant overlap with other ethanol-regulated gene sets. Altogether, these studies provide robust primate and rodent cross-species validation of gene networks associated with chronic ethanol consumption. Our results also suggest potential novel focal points for future therapeutic interventions in alcohol use disorder.
Collapse
Affiliation(s)
- James W Bogenpohl
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, United States
| | - Maren L Smith
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Sean P Farris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, United States
| | - Catherine I Dumur
- Aurora Diagnostics-Sonic Healthcare, Bernhardt Laboratories, Jacksonville, FL, United States
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Howard C Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Kathleen A Grant
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States.,Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Michael F Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States.,Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Department of Neurology, Virginia Commonwealth University, Richmond, VA, United States.,VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
49
|
Sex Differences in the Effect of Alcohol Drinking on Myelinated Axons in the Anterior Cingulate Cortex of Adolescent Rats. Brain Sci 2019; 9:brainsci9070167. [PMID: 31315270 PMCID: PMC6680938 DOI: 10.3390/brainsci9070167] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 12/29/2022] Open
Abstract
Cognitive deficits associated with teenage drinking may be due to disrupted myelination of prefrontal circuits. To better understand how alcohol affects myelination, male and female Wistar rats (n = 7-9/sex/treatment) underwent two weeks of intermittent operant self-administration of sweetened alcohol or sweetened water early in adolescence (postnatal days 28-42) and we tested for macro- and microstructural changes to myelin. We previously reported data from the males of this study showing that alcohol drinking reduced myelinated fiber density in layers II-V of the anterior cingulate division of the medial prefrontal cortex (Cg1); herein, we show that myelinated fiber density was not significantly altered by alcohol in females. Alcohol drinking patterns were similar in both sexes, but males were in a pre-pubertal state for a larger proportion of the alcohol exposure period, which may have contributed to the differential effects on myelinated fiber density. To gain more insight into how alcohol impacts myelinated axons, brain sections from a subset of these animals (n = 6/sex/treatment) were used for microstructural analyses of the nodes of Ranvier. Confocal analysis of nodal domains, flanked by immunofluorescent-labeled contactin-associated protein (Caspr) clusters, indicated that alcohol drinking reduced nodal length-to-width ratios in layers II/III of the Cg1 in both sexes. Despite sex differences in the underlying cause (larger diameter axons after alcohol in males vs. shorter nodal lengths after alcohol in females), reduced nodal ratios could have important implications for the speed and integrity of neural transmission along these axons in both males and females. Alcohol-induced changes to myelinated axonal populations in the Cg1 may contribute to long-lasting changes in prefrontal function associated with early onset drinking.
Collapse
|
50
|
Rice J, Gu C. Function and Mechanism of Myelin Regulation in Alcohol Abuse and Alcoholism. Bioessays 2019; 41:e1800255. [PMID: 31094014 DOI: 10.1002/bies.201800255] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/31/2019] [Indexed: 12/26/2022]
Abstract
Excessive alcohol use has adverse effects on the central nervous system (CNS) and can lead to alcohol use disorders (AUDs). Recent studies have suggested that myelin reductions may directly contribute to CNS dysfunctions associated with AUDs. Myelin consists of compact lipid membranes wrapped around axons to provide electrical insulation and trophic support. Regulation of myelin is considered as a new form of neural plasticity due to its profound impacts on the computation of neural networks. In this review, the authors first discuss experimental evidence showing how alcohol exposure causes demyelination in different brain regions, often accompanied by deficits in cognition and emotion. Next, they discuss postulated molecular and cellular mechanisms underlying alcohol's impact on myelin. It is clear that more extensive investigations are needed in this important but underexplored research field in order to gain a better understanding of the myelin-behavior relationship and to develop new treatment strategies for AUDs.
Collapse
Affiliation(s)
- James Rice
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 1060 Carmack Road, Columbus, OH, 43210, USA
| | - Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 1060 Carmack Road, Columbus, OH, 43210, USA
| |
Collapse
|