1
|
Nwachukwu KN, Nelson JC, Hill KM, Clark KA, Healey K, Swartzwelder HS, Marshall SA. Donepezil Reverses Alcohol-Induced Changes in Hippocampal Neurogenic and Glial Responses Following Adolescent Intermittent Ethanol Exposure Into Adulthood in Female Rats. Hippocampus 2025; 35:e70001. [PMID: 39967057 PMCID: PMC11836526 DOI: 10.1002/hipo.70001] [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: 05/07/2024] [Revised: 12/03/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025]
Abstract
Adolescent intermittent ethanol (AIE) exposure leads to persisting increases in glial markers and significantly decreases the neurogenic niche in the dentate gyrus of the hippocampus. Our previous study indicated that donepezil (DZ), a cholinesterase inhibitor, can reverse the AIE effect of decreased doublecortin (DCX), a neurogenic marker, and increased cleaved caspase 3, a marker of apoptosis, in the dentate gyrus of male rats. However, to date, no studies have assessed the effects of DZ on AIE effects in females. The purpose of this study was to determine whether DZ can reverse neuroimmune, neurogenic, and neuronal death effects in adulthood after AIE in female rats. Adolescent female rats were given 14 doses of ethanol (5 g/kg) over 24 days by intragastric gavage. Seventeen days later, DZ (2.5 mg/kg, 1.88 mL/kg, i.g., in water) was then administered daily for 4 days prior to sacrifice. Immunohistochemical techniques were utilized to determine the effects of DZ on AIE-induced changes in neurogenesis, cell death, glial, and neuroimmune markers. As expected, AIE decreased the neurogenic markers DCX, SOX2, and Ki-67 in the dentate gyrus and also caused an increase in the glial markers GFAP and Iba-1 in the hippocampus. The effects of AIE on neurogenic and glial markers were reversed by DZ treatment, but the reversal of AIE effects on glial markers was regionally specific within the hippocampus. Overall, these findings indicate that systemic DZ in adult female rats ameliorates the effects of AIE on neurogenesis, neuronal cell death, neuroimmune markers, and glial activation markers. Future studies will determine if DZ alters hippocampally driven behaviors, as well as the mechanisms underlying donepezil's effects.
Collapse
Affiliation(s)
- Kala N. Nwachukwu
- Department of Biological & Biomedical SciencesNorth Carolina Central UniversityDurhamNorth CarolinaUSA
- Integrated Biosciences PhD ProgramNorth Carolina Central UniversityDurhamNorth CarolinaUSA
| | - James C. Nelson
- Department of Biological & Biomedical SciencesNorth Carolina Central UniversityDurhamNorth CarolinaUSA
| | - Kennedy M. Hill
- Department of Biological & Biomedical SciencesNorth Carolina Central UniversityDurhamNorth CarolinaUSA
| | - Kennedy A. Clark
- Department of Biological & Biomedical SciencesNorth Carolina Central UniversityDurhamNorth CarolinaUSA
| | - Kati Healey
- Durham Veterans Affairs Medical CenterDurhamNorth CarolinaUSA
| | - H. Scott Swartzwelder
- Department of Psychiatry and Behavioral SciencesDuke University Medical CenterDurhamNorth CarolinaUSA
| | - S. Alex Marshall
- Department of Biological & Biomedical SciencesNorth Carolina Central UniversityDurhamNorth CarolinaUSA
- Julius L. Chambers Biomedical & Biotechnology Research InstituteNorth Carolina Central UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
2
|
Yoon G, Sofuoglu M, Petrakis IL, Pittman B, Bell MD. The combination of donepezil and cognitive training for improving treatment outcomes for alcohol use disorder: Design of a randomized controlled trial. Contemp Clin Trials 2024; 145:107657. [PMID: 39111388 PMCID: PMC11423257 DOI: 10.1016/j.cct.2024.107657] [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: 11/10/2023] [Revised: 07/13/2024] [Accepted: 08/03/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND The development of alcohol use disorder (AUD) is a major concern in public health, and cognitive impairments caused by alcohol are involved in this process. Emerging neurobiological evidence suggests that donepezil, an anticholinesterase agent, may improve AUD treatment outcomes by enhancing neurocognitive functioning. Previous research has also suggested that cognitive remediation therapy (CRT) could potentially improve cognitive function and AUD treatment outcomes. We present the rationale and design of a trial to evaluate the combination of donepezil and cognitive remediation therapy (donepezil + CRT) as an intervention for AUD. METHODS We propose a 13-week, randomized, double-blind, placebo-controlled, between-subjects trial comparing 4 groups (donepezil + CRT vs. donepezil alone vs. CRT alone vs. placebos) as an intervention for AUD. The main goal of the study is to evaluate if donepezil + CRT is superior to placebo in reducing heavy drinking days and improving neurocognitive functioning. A total of 160 patients (4 groups, 40 per each group) with AUD between the ages of 18-80 years will be recruited at Yale University and the VA Connecticut Healthcare System. Primary outcome measures include 1) heavy drinking by Timeline Follow Back (TLFB) over 13 weeks and 2) global neurocognitive functioning by a global index of neurocognitive function score at 7 and 13 weeks. DISCUSSION This protocol paper describes the rationale and proposed methods for the randomized controlled trial for improving AUD treatment outcomes. This project has significant clinical potential to help patients suffering from AUD by improving their cognition and reducing alcohol consumption. TRIAL REGISTRATION NCT05042102.
Collapse
Affiliation(s)
- Gihyun Yoon
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
| | - Mehmet Sofuoglu
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Ismene L Petrakis
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Brian Pittman
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Morris D Bell
- VA Connecticut Healthcare System, West Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Cai Y, Wang LW, Wu J, Chen ZW, Yu XF, Liu FH, Gao DP. Fasudil alleviates alcohol-induced cognitive deficits and hippocampal morphology injury partly by altering the assembly of the actin cytoskeleton and microtubules. Behav Brain Res 2024; 471:115068. [PMID: 38830386 DOI: 10.1016/j.bbr.2024.115068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
Alcohol-Related Brain Damage (ARBD) manifests predominantly as cognitive impairment and brain atrophy with the hippocampus showing particular vulnerability. Fasudil, a Rho kinase (ROCK) inhibitor, has established neuroprotective properties; however, its impact on alcohol-induced cognitive dysfunction and hippocampal structural damage remains unelucidated. This study probes Fasudil's neuroprotective potential and identifies its mechanism of action in an in vivo context. Male C57BL/6 J mice were exposed to 30% (v/v, 6.0 g/kg) ethanol by intragastric administration for four weeks. Concurrently, these mice received a co-treatment with Fasudil through intraperitoneal injections at a dosage of 10 mg/kg/day. Fasudil was found to mitigate alcohol-induced spatial and recognition memory deficits, which were quantified using Y maze, Morris water maze, and novel object recognition tests. Concurrently, Fasudil attenuated hippocampal structural damage prompted by chronic alcohol exposure. Notably, Fasudil moderated alcohol-induced disassembly of the actin cytoskeleton and microtubules-mechanisms central to the maintenance of hippocampal synaptic integrity. Collectively, our findings indicate that Fasudil partially reverses alcohol-induced cognitive and morphological detriments by modulating cytoskeletal dynamics, offering insights into potential therapeutic strategies for ARBD.
Collapse
Affiliation(s)
- Yu Cai
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Lu-Wan Wang
- School of Medical, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, PR China
| | - Jing Wu
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Zi-Wei Chen
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Xue-Feng Yu
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Fu-He Liu
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Da-Peng Gao
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China.
| |
Collapse
|
4
|
Healey K, Waters RC, Knight SG, Wandling GM, Hall NI, Jones BN, Shobande MJ, Melton JG, Pandey SC, Scott Swartzwelder H, Maldonado-Devincci AM. Adolescent intermittent ethanol exposure alters adult exploratory and affective behaviors, and cerebellar Grin2b expression in C57BL/6J mice. Drug Alcohol Depend 2023; 253:111026. [PMID: 38006668 PMCID: PMC10990063 DOI: 10.1016/j.drugalcdep.2023.111026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 09/17/2023] [Accepted: 11/01/2023] [Indexed: 11/27/2023]
Abstract
Binge drinking is one of the most common patterns (more than 90%) of alcohol consumption by young people. During adolescence, the brain undergoes maturational changes that influence behavioral control and affective behaviors, such as cerebellar brain volume and function in adulthood. We investigated long-term impacts of adolescent binge ethanol exposure on affective and exploratory behaviors and cerebellar gene expression in adult male and female mice. Further, the cerebellum is increasingly recognized as a brain region integrating a multitude of behaviors that span from the traditional primary sensory-motor to affective functions, such as anxiety and stress reactivity. Therefore, we investigated the persistent effects of adolescent intermittent ethanol (AIE) on exploratory and affective behaviors and began to elucidate the role of the cerebellum in these behaviors through excitatory signaling gene expression. We exposed C57BL/6J mice to AIE or air (control) vapor inhalation from postnatal day 28-42. After prolonged abstinence (>34 days), in young adulthood (PND 77+) we assessed behavior in the open field, light/dark, tail suspension, and forced swim stress tests to determine changes in affective behaviors including anxiety-like, depressive-like, and stress reactivity behavior. Excitatory signaling gene mRNA levels of fragile X messenger ribonucleoprotein (FMR1), glutamate receptors (Grin2a, Grin2b and Grm5) and excitatory synaptic markers (PSD-95 and Eaat1) were measured in the cerebellum of adult control and AIE-exposed mice. AIE-exposed mice showed decreased exploratory behaviors in the open field test (OFT) where both sexes show reduced ambulation, however only females exhibited a reduction in rearing. Additionally, in the OFT, AIE-exposed females also exhibited increased anxiety-like behavior (entries to center zone). In the forced swim stress test, AIE-exposed male mice, but not females, spent less time immobile compared to their same-sex controls, indicative of sex-specific changes in stress reactivity. Male and female AIE-exposed mice showed increased Grin2b (Glutamate Ionotropic Receptor NMDA Type Subunit 2B) mRNA levels in the cerebellum compared to their same-sex controls. Together, these data show that adolescent binge-like ethanol exposure altered both exploratory and affective behaviors in a sex-specific manner and modified cerebellar Grin2b expression in adult mice. This indicates the cerebellum may serve as an important brain region that is susceptible to long-term molecular changes after AIE.
Collapse
Affiliation(s)
- Kati Healey
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, 323 Foster St., Durham, NC 27701, United States
| | - Renee C Waters
- Department of Psychology, Hairston College of Health and Human Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States; Department of Psychology, Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540, United States
| | - Sherilynn G Knight
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Gabriela M Wandling
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois, Chicago, IL, United States
| | - Nzia I Hall
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States; University of North Carolina at Chapel Hill School of Medicine, NC 27516, United States
| | - Brooke N Jones
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Mariah J Shobande
- Department of Chemical, Biological and Bioengineering, College of Engineering, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Jaela G Melton
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois, Chicago, IL, United States; Jesse Brown VA Medical Center, Chicago, IL, United States
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, 323 Foster St., Durham, NC 27701, United States
| | - Antoniette M Maldonado-Devincci
- Department of Psychology, Hairston College of Health and Human Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States.
| |
Collapse
|
5
|
Hauser SR, Waeiss RA, Deehan GA, Engleman EA, Bell RL, Rodd ZA. Adolescent alcohol and nicotine exposure alters the adult response to alcohol use. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11880. [PMID: 38389816 PMCID: PMC10880795 DOI: 10.3389/adar.2023.11880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 02/24/2024]
Abstract
Adolescence through young adulthood is a unique period of neuronal development and maturation. Numerous agents can alter this process, resulting in long-term neurological and biological consequences. In the clinical literature, it is frequently reported that adolescent alcohol consumption increases the propensity to develop addictions, including alcohol use disorder (AUD), during adulthood. A general limitation of both clinical and human pre-clinical adolescent alcohol research is the high rate of co-using/abusing more than one drug during adolescence, such as co-using/abusing alcohol with nicotine. A primary goal of basic research is elucidating neuroadaptations produced by adolescent alcohol exposure/consumption that promote alcohol and other drug self-administration in adulthood. The long-term goal is to develop pharmacotherapeutics for the prevention or amelioration of these neuroadaptations. This review will focus on studies that have examined the effects of adolescent alcohol and nicotine exposure on adult alcohol consumption, the hypersensitivity of the mesolimbic dopaminergic system, and enhanced responses not only to alcohol but also to nicotine during adulthood. Again, the long-term goal is to identify potential cholinergic agents to prevent or ameliorate the consequences of, peri-adolescent alcohol abuse.
Collapse
Affiliation(s)
- Sheketha R Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert A Waeiss
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Gerald A Deehan
- Department of Psychology, East Tennessee State University, Johnson City, TN, United States
| | - Eric A Engleman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zachary A Rodd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
6
|
Steinfeld MR, Torregrossa MM. Consequences of adolescent drug use. Transl Psychiatry 2023; 13:313. [PMID: 37802983 PMCID: PMC10558564 DOI: 10.1038/s41398-023-02590-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/22/2023] [Accepted: 08/23/2023] [Indexed: 10/08/2023] Open
Abstract
Substance use in adolescence is a known risk factor for the development of neuropsychiatric and substance use disorders in adulthood. This is in part due to the fact that critical aspects of brain development occur during adolescence, which can be altered by drug use. Despite concerted efforts to educate youth about the potential negative consequences of substance use, initiation remains common amongst adolescents world-wide. Additionally, though there has been substantial research on the topic, many questions remain about the predictors and the consequences of adolescent drug use. In the following review, we will highlight some of the most recent literature on the neurobiological and behavioral effects of adolescent drug use in rodents, non-human primates, and humans, with a specific focus on alcohol, cannabis, nicotine, and the interactions between these substances. Overall, consumption of these substances during adolescence can produce long-lasting changes across a variety of structures and networks which can have enduring effects on behavior, emotion, and cognition.
Collapse
Affiliation(s)
- Michael R Steinfeld
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA.
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA
| |
Collapse
|
7
|
Healey K, Waters RC, Knight SG, Wandling GM, Hall NI, Jones BN, Shobande MJ, Melton JG, Pandey SC, Scott Swartzwelder H, Maldonado-Devincci AM. Adolescent Intermittent Ethanol Exposure Alters Adult Exploratory and Affective Behaviors, and Cerebellar Grin2B Expression in C57BL/6J Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528396. [PMID: 36824954 PMCID: PMC9949091 DOI: 10.1101/2023.02.13.528396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Binge drinking is one of the most common patterns (more than 90%) of alcohol consumption by young people. During adolescence, the brain undergoes maturational changes that influence behavioral control and affective behaviors, such as cerebellar brain volume and function in adulthood. We investigated long-term impacts of adolescent binge ethanol exposure on affective and exploratory behaviors and cerebellar gene expression in adult male and female mice. Further, the cerebellum is increasingly recognized as a brain region integrating a multitude of behaviors that span from the traditional primary sensory-motor to affective functions, such as anxiety and stress reactivity. Therefore, we investigated the persistent effects of adolescent intermittent ethanol (AIE) on exploratory and affective behaviors and began to elucidate the role of the cerebellum in these behaviors through excitatory signaling gene expression. We exposed C57BL/6J mice to AIE or air (control) vapor inhalation from postnatal day 28-42. After prolonged abstinence (>34 days), in young adulthood (PND 77+) we assessed behavior in the open field, light/dark, tail suspension, and forced swim stress tests to determine changes in affective behaviors including anxiety-like, depressive-like, and stress reactivity behavior. Excitatory signaling gene mRNA levels of fragile X messenger ribonucleoprotein ( FMR1) , glutamate receptors ( Grin2a , Grin2B and Grm5 ) and excitatory synaptic markers (PSD-95 and Eaat1) were measured in the cerebellum of adult control and AIE-exposed mice. AIE-exposed mice showed decreased exploratory behaviors in the open field test (OFT) where both sexes show reduced ambulation, however only females exhibited a reduction in rearing. Additionally, in the OFT, AIE-exposed females also exhibited increased anxiety-like behavior (entries to center zone). In the forced swim stress test, AIE-exposed male mice, but not females, spent less time immobile compared to their same-sex controls, indicative of sex-specific changes in stress reactivity. Male and female AIE-exposed mice showed increased Grin2B (Glutamate Ionotropic Receptor NMDA Type Subunit 2B) mRNA levels in the cerebellum compared to their same-sex controls. Together, these data show that adolescent binge-like ethanol exposure altered both exploratory and affective behaviors in a sex-specific manner and modified cerebellar Grin2B expression in adult mice. This indicates the cerebellum may serve as an important brain region that is susceptible to long-term molecular changes after AIE. Highlights Adolescent intermittent ethanol (AIE) exposure decreased exploratory behavior in adult male and female mice.In females, but not males, AIE increased anxiety-like behavior.In males, but not females, AIE reduced stress reactivity in adulthood.These findings indicate sex differences in the enduring effects of AIE on exploratory and affective behaviors. Cerebellar Grin2B mRNA levels were increased in adulthood in both male and female AIE-exposed mice. These findings add to the small, but growing literature on behavioral AIE effects in mice, and establish cerebellar excitatory synaptic gene expression as an enduring effect of adolescent ethanol exposure.
Collapse
|
8
|
Nwachukwu KN, Mohammed HE, Mebane DR, Barber AW, Swartzwelder HS, Marshall SA. Acute and Chronic Ethanol Effects during Adolescence on Neuroimmune Responses: Consequences and Potential Pharmacologic Interventions. Cells 2023; 12:1423. [PMID: 37408257 PMCID: PMC10217092 DOI: 10.3390/cells12101423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 07/07/2023] Open
Abstract
Heavy ethanol consumption during adolescence has been linked to neuroimmune response dysregulation and cognitive deficits in the developing adolescent brain. During adolescence, the brain is particularly susceptible to the pharmacological effects of ethanol that are induced by acute and chronic bouts of exposure. Numerous preclinical rodent model studies have used different ethanol administration techniques, such as intragastric gavage, self-administration, vapor, intraperitoneal, and free access, and while most models indicated proinflammatory neuroimmune responses in the adolescent brain, there are various factors that appear to influence this observation. This review synthesizes the most recent findings of the effects of adolescent alcohol use on toll-like receptors, cytokines, and chemokines, as well as the activation of astrocytes and microglia with an emphasis on differences associated with the duration of ethanol exposure (acute vs. chronic), the amount of exposure (e.g., dose or blood ethanol concentrations), sex differences, and the timing of the neuroimmune observation (immediate vs. persistent). Finally, this review discusses new therapeutics and interventions that may ameliorate the dysregulation of neuroimmune maladaptations after ethanol exposure.
Collapse
Affiliation(s)
- Kala N. Nwachukwu
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
- Integrated Biosciences PhD Program, North Carolina Central University, Durham, NC 27707, USA
| | - Hassan E. Mohammed
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - DaQuan R. Mebane
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - Andrew W. Barber
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - H. Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27708, USA
| | - S. Alex Marshall
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
9
|
Rodd ZA, Swartzwelder HS, Waeiss RA, Soloviov SO, Lahiri DK, Engleman EA, Truitt WA, Bell RL, Hauser SR. Negative and positive allosteric modulators of the α7 nicotinic acetylcholine receptor regulates the ability of adolescent binge alcohol exposure to enhance adult alcohol consumption. Front Behav Neurosci 2023; 16:954319. [PMID: 37082421 PMCID: PMC10113115 DOI: 10.3389/fnbeh.2022.954319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 12/09/2022] [Indexed: 04/07/2023] Open
Abstract
Rationale and Objectives: Ethanol acts directly on the α7 Nicotinic acetylcholine receptor (α7). Adolescent-binge alcohol exposure (ABAE) produces deleterious consequences during adulthood, and data indicate that the α7 receptor regulates these damaging events. Administration of an α7 Negative Allosteric Modulator (NAM) or the cholinesterase inhibitor galantamine can prophylactically prevent adult consequences of ABAE. The goals of the experiments were to determine the effects of co-administration of ethanol and a α7 agonist in the mesolimbic dopamine system and to determine if administration of an α7 NAM or positive allosteric modulator (PAM) modulates the enhancement of adult alcohol drinking produced by ABAE. Methods: In adult rats, ethanol and the α7 agonist AR-R17779 (AR) were microinjected into the posterior ventral tegmental area (VTA), and dopamine levels were measured in the nucleus accumbens shell (AcbSh). In adolescence, rats were treated with the α7 NAM SB-277011-A (SB) or PNU-120596 (PAM) 2 h before administration of EtOH (ABAE). Ethanol consumption (acquisition, maintenance, and relapse) during adulthood was characterized. Results: Ethanol and AR co-administered into the posterior VTA stimulated dopamine release in the AcbSh in a synergistic manner. The increase in alcohol consumption during the acquisition and relapse drinking during adulthood following ABAE was prevented by administration of SB, or enhanced by administration of PNU, prior to EtOH exposure during adolescence. Discussion: Ethanol acts on the α7 receptor, and the α7 receptor regulates the critical effects of ethanol in the brain. The data replicate the findings that cholinergic agents (α7 NAMs) can act prophylactically to reduce the alterations in adult alcohol consumption following ABAE.
Collapse
Affiliation(s)
- Zachary A. Rodd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - H. Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - R. Aaron Waeiss
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Serhii O. Soloviov
- Department of Pharmacy, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
- Department of Industrial Biotechnology and Biopharmacy, National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, Kyiv, Ukraine
| | - Debomoy K. Lahiri
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Eric A. Engleman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - William A. Truitt
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Richard L. Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sheketha R. Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
10
|
Healey KL, Kibble S, Dubester K, Bell A, Swartzwelder HS. Adolescent intermittent ethanol exposure enhances adult stress effects in male rats. Pharmacol Biochem Behav 2023; 223:173513. [PMID: 36610590 PMCID: PMC10028459 DOI: 10.1016/j.pbb.2022.173513] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
Binge patterns of alcohol use, prevalent among adolescents, are associated with a higher probability of developing alcohol use disorders (AUD) and other psychiatric disorders, like anxiety and depression. Additionally, adverse life events strongly predict AUD and other psychiatric disorders. As such, the combined fields of stress and AUD have been well established, and animal models indicate that both binge-like alcohol exposure and stress exposure elevate anxiety-like behaviors. However, few have investigated the interaction of adolescent intermittent ethanol (AIE) and adult stressors. We hypothesized that AIE would increase vulnerability to restraint-induced stress (RS), manifested as increased anxiety-like behavior. After AIE exposure, in adulthood, animals were tested on forced swim (FST) and saccharin preference (SP) and then exposed to either RS (90 min/5 days) or home-cage control. Twenty-four hours after the last RS session, animals began testing on the elevated plus maze (EPM), and were re-tested on FST and SP. A separate group of animals were sacrificed in adulthood after AIE and RS, and brains were harvested for immunoblot analysis of dorsal and ventral hippocampus. Consistent with previous reports, AIE had no significant effect on closed arm time in the EPM (anxiety-like behavior). However, in male rats the interaction of AIE and adult RS increased time spent in the closed arms. No effect was observed among female animals. AIE and RS-specific alterations were found in glial and synaptic markers (GLT-1, FMRP and PSD-95) in male animals. These findings indicate AIE has sex-specific effects on both SP and the interaction of AIE and adult RS, which induces a propensity toward anxiety-like behavior in males. Also, AIE produces persistent hippocampal deficits that may interact with adult RS to cause increased anxiety-like behaviors. Understanding the mechanisms behind this AIE-induced increase in stress vulnerability may provide insight into treatment and prevention strategies for alcohol use disorders.
Collapse
Affiliation(s)
- Kati L Healey
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, United States of America.
| | - Sandra Kibble
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, United States of America
| | - Kira Dubester
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, United States of America
| | - Amelia Bell
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, United States of America
| | - H S Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27710, United States of America
| |
Collapse
|
11
|
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: 2.5] [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
|
12
|
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: 2.0] [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
|
13
|
Amodeo LR, Jennings SD, Mulholland PJ, Ehlers CL. Chronic intermittent ethanol during adolescence and adulthood alters dendritic spines in the primary motor and visual cortex in rats. Alcohol 2021; 97:67-74. [PMID: 34626787 DOI: 10.1016/j.alcohol.2021.09.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/14/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022]
Abstract
Prolonged adolescent binge drinking can disrupt sleep quality and increase the likelihood of alcohol-induced sleep disruptions in young adulthood in rodents and in humans. Striking changes in spine density and morphology have been seen in many cortical and subcortical regions after adolescent alcohol exposure in rats. However, there is little known about the impact of alcohol exposure on dendritic spines in the same motor and sensory cortices that EEG sleep is typically recorded from in rats. The aim of this study is to investigate whether an established model of chronic intermittent ethanol vapor in rats that has been demonstrated to disrupt sleep during adolescence or adulthood, also significantly alters cortical dendritic spine density and morphology. To this end, adolescent and adult Wistar rats were exposed to 5 weeks of ethanol vapor or control air exposure. After a 13-day withdrawal, primary motor cortex (M1) and primary/secondary visual cortex (V1/V2) layer V dendrites were analyzed for differences in spine density and morphology. Spines were classified into four categories (stubby, long, filopodia, and mushroom) based on the spine length and the width of the spine head and neck. The main results indicate an age-specific effect of adolescent intermittent ethanol exposure decreasing spine density in the M1 cortex compared to age-matched controls. Reductions in the density of M1 long-shaped spine subclassifications were seen in adolescent ethanol-exposed rats, but not adult-exposed rats, compared to their air-controls. Irrespective of age, there was an overall reduction produced by ethanol exposure on the density of filopodia and the length of long-shaped spines in V1/V2 cortex as compared to their air-exposed controls. Together, these data add to growing evidence that some cortical circuits are vulnerable to the effects of alcohol during adolescence and begin to elucidate potential mechanisms that may influence brain plasticity following early alcohol use.
Collapse
Affiliation(s)
- Leslie R Amodeo
- Department of Psychology, California State University San Bernardino, San Bernardino, CA, 92407, United States
| | - Sarah D Jennings
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, United States
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, United States
| | - Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, United States.
| |
Collapse
|
14
|
Hauser SR, Mulholland PJ, Truitt WA, Waeiss RA, Engleman EA, Bell RL, Rodd ZA. Adolescent Intermittent Ethanol (AIE) Enhances the Dopaminergic Response to Ethanol within the Mesolimbic Pathway during Adulthood: Alterations in Cholinergic/Dopaminergic Genes Expression in the Nucleus Accumbens Shell. Int J Mol Sci 2021; 22:11733. [PMID: 34769161 PMCID: PMC8584082 DOI: 10.3390/ijms222111733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
A consistent preclinical finding is that exposure to alcohol during adolescence produces a persistent hyperdopaminergic state during adulthood. The current experiments determine that effects of Adolescent Intermittent Ethanol (AIE) on the adult neurochemical response to EtOH administered directly into the mesolimbic dopamine system, alterations in dendritic spine and gene expression within the nucleus accumbens shell (AcbSh), and if treatment with the HDACII inhibitor TSA could normalize the consequences of AIE. Rats were exposed to the AIE (4 g/kg ig; 3 days a week) or water (CON) during adolescence, and all testing occurred during adulthood. CON and AIE rats were microinjected with EtOH directly into the posterior VTA and dopamine and glutamate levels were recorded in the AcbSh. Separate groups of AIE and CON rats were sacrificed during adulthood and Taqman arrays and dendritic spine morphology assessments were performed. The data indicated that exposure to AIE resulted in a significant leftward and upward shift in the dose-response curve for an increase in dopamine in the AcbSh following EtOH microinjection into the posterior VTA. Taqman array indicated that AIE exposure affected the expression of target genes (Chrna7, Impact, Chrna5). The data indicated no alterations in dendritic spine morphology in the AcbSh or any alteration in AIE effects by TSA administration. Binge-like EtOH exposure during adolescence enhances the response to acute ethanol challenge in adulthood, demonstrating that AIE produces a hyperdopaminergic mesolimbic system in both male and female Wistar rats. The neuroadaptations induced by AIE in the AcbSh could be part of the biological basis of the observed negative consequences of adolescent binge-like alcohol exposure on adult drug self-administration behaviors.
Collapse
Affiliation(s)
- Sheketha R. Hauser
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.H.); (W.A.T.); (R.A.W.); (E.A.E.); (R.L.B.)
| | - Patrick J. Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - William A. Truitt
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.H.); (W.A.T.); (R.A.W.); (E.A.E.); (R.L.B.)
| | - R. Aaron Waeiss
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.H.); (W.A.T.); (R.A.W.); (E.A.E.); (R.L.B.)
| | - Eric A. Engleman
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.H.); (W.A.T.); (R.A.W.); (E.A.E.); (R.L.B.)
| | - Richard L. Bell
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.H.); (W.A.T.); (R.A.W.); (E.A.E.); (R.L.B.)
| | - Zachary A. Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.H.); (W.A.T.); (R.A.W.); (E.A.E.); (R.L.B.)
| |
Collapse
|
15
|
Coleman LG, Crews FT, Vetreno RP. The persistent impact of adolescent binge alcohol on adult brain structural, cellular, and behavioral pathology: A role for the neuroimmune system and epigenetics. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:1-44. [PMID: 34696871 DOI: 10.1016/bs.irn.2021.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adolescence is a critical neurodevelopmental window for maturation of brain structure, neurocircuitry, and glia. This development is sculpted by an individual's unique experiences and genetic background to establish adult level cognitive function and behavioral makeup. Alcohol abuse during adolescence is associated with an increased lifetime risk for developing an alcohol use disorder (AUD). Adolescents participate in heavy, episodic binge drinking that causes persistent changes in neurocircuitry and behavior. These changes may underlie the increased risk for AUD and might also promote cognitive deficits later in life. In this chapter, we have examined research on the persistent effects of adolescent binge-drinking both in humans and in rodent models. These studies implicate roles for neuroimmune signaling as well as epigenetic reprogramming of neurons and glia, which create a vulnerable neuroenvironment. Some of these changes are reversible, giving hope for future treatments to prevent many of the long-term consequences of adolescent alcohol abuse.
Collapse
Affiliation(s)
- Leon G Coleman
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| | - Fulton T Crews
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
16
|
Dulman RS, Auta J, Wandling GM, Patwell R, Zhang H, Pandey SC. Persistence of cerebellar ataxia during chronic ethanol exposure is associated with epigenetic up-regulation of Fmr1 gene expression in rat cerebellum. Alcohol Clin Exp Res 2021; 45:2006-2016. [PMID: 34453331 PMCID: PMC8602769 DOI: 10.1111/acer.14691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Alcohol intoxication produces ataxia by affecting the cerebellum, which coordinates movements. Fragile X mental retardation (FMR) protein is a complex regulator of RNA and synaptic plasticity implicated in fragile X-associated tremor/ataxia syndrome, which features ataxia and increased Fmr1 mRNA expression resulting from epigenetic dysregulation of FMRP. We recently demonstrated that acute ethanol-induced ataxia is associated with increased cerebellar Fmr1 gene expression via histone modifications in rats, but it is unknown whether similar behavioral and molecular changes occur following chronic ethanol exposure. Here, we investigated the effects of chronic ethanol exposure on ataxia and epigenetically regulated changes in Fmr1 expression in the cerebellum. METHODS Male adult Sprague-Dawley rats were trained on the accelerating rotarod and then fed with chronic ethanol or a control Lieber-DeCarli diet while undergoing periodic behavioral testing for ataxia during ethanol exposure and withdrawal. Cerebellar tissues were analyzed for expression of the Fmr1 gene and its targets using a real-time quantitative polymerase chain reaction assay. The epigenetic regulation of Fmr1 was also investigated using a chromatin immunoprecipitation assay. RESULTS Ataxic behavior measured by the accelerating rotarod behavioral test developed during chronic ethanol treatment and persisted at both the 8-h and 24-h withdrawal time points compared to control diet-fed rats. In addition, chronic ethanol treatment resulted in up-regulated expression of Fmr1 mRNA and increased activating epigenetic marks H3K27 acetylation and H3K4 trimethylation at 2 sites within the Fmr1 promoter. Finally, measurement of the expression of relevant FMRP mRNA targets in the cerebellum showed that chronic ethanol up-regulated cAMP response element binding (CREB) Creb1, Psd95, Grm5, and Grin2b mRNA expression without altering Grin2a, Eaa1, or histone acetyltransferases CREB binding protein (Cbp) or p300 mRNA transcripts. CONCLUSIONS These results suggest that epigenetic regulation of Fmr1 and subsequent FMRP regulation of target mRNA transcripts constitute neuroadaptations in the cerebellum that may underlie the persistence of ataxic behavior during chronic ethanol exposure and withdrawal.
Collapse
Affiliation(s)
- Russell S. Dulman
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - James Auta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - Gabriela M. Wandling
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - Ryan Patwell
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, 60612 USA
| | - Subhash C. Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, 60612 USA
| |
Collapse
|
17
|
Hauser SR, Rodd ZA, Deehan GA, Liang T, Rahman S, Bell RL. Effects of adolescent substance use disorders on central cholinergic function. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:175-221. [PMID: 34696873 DOI: 10.1016/bs.irn.2021.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adolescence is a transitional period between childhood and adulthood, in which the individual undergoes significant cognitive, behavioral, physical, emotional, and social developmental changes. During this period, adolescents engage in experimentation and risky behaviors such as licit and illicit drug use. Adolescents' high vulnerability to abuse drugs and natural reinforcers leads to greater risk for developing substance use disorders (SUDs) during adulthood. Accumulating evidence indicates that the use and abuse of licit and illicit drugs during adolescence and emerging adulthood can disrupt the cholinergic system and its processes. This review will focus on the effects of peri-adolescent nicotine and/or alcohol use, or exposure, on the cholinergic system during adulthood from preclinical and clinical studies. This review further explores potential cholinergic agents and pharmacological manipulations to counteract peri-adolescent nicotine and/or alcohol abuse.
Collapse
Affiliation(s)
- S R Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - Z A Rodd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - G A Deehan
- Department of Psychology, East Tennessee State University, Johnson City, TN, United States
| | - T Liang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
18
|
Walter N, Cervera-Juanes R, Zheng C, Darakjian P, Lockwood D, Cuzon-Carlson V, Ray K, Fei S, Conrad D, Searles R, Grant K, Hitzemann R. Effect of chronic ethanol consumption in rhesus macaques on the nucleus accumbens core transcriptome. Addict Biol 2021; 26:e13021. [PMID: 33942443 PMCID: PMC8588809 DOI: 10.1111/adb.13021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
The nucleus accumbens core (NAcc) has been repeatedly demonstrated to be a key component of the circuitry associated with excessive ethanol consumption. Previous studies have illustrated that in a nonhuman primate (NHP) model of chronic ethanol consumption, there is significant epigenetic remodeling of the NAcc. In the current study, RNA-Seq was used to examine genome-wide gene expression in eight each of control, low/binge (LD*), and high/very high (HD*) rhesus macaque drinkers. Using an FDR < 0.05, zero genes were significantly differentially expressed (DE) between LD* and controls, six genes between HD* and LD*, and 734 genes between HD* and controls. Focusing on HD* versus control DE genes, the upregulated genes (N = 366) were enriched in genes with annotations associated with signal recognition particle (SRP)-dependent co-translational protein targeting to membrane (FDR < 3 × 10-59 ), structural constituent of ribosome (FDR < 3 × 10-47 ), and ribosomal subunit (FDR < 5 × 10-48 ). Downregulated genes (N = 363) were enriched in annotations associated with behavior (FDR < 2 × 10-4 ), membrane organization (FDR < 1 × 10-4 ), inorganic cation transmembrane transporter activity (FDR < 2 × 10-3 ), synapse part (FDR < 4 × 10-10 ), glutamatergic synapse (FDR < 1 × 10-6 ), and GABAergic synapse (FDR < 6 × 10-4 ). Ingenuity Pathway Analysis (IPA) revealed that EIF2 signaling and mTOR pathways were significantly upregulated in HD* animals (FDR < 3 × 10-33 and <2 × 10-16 , respectively). Overall, the data supported our working hypothesis; excessive consumption would be associated with transcriptional differences in GABA/glutamate-related genes.
Collapse
Affiliation(s)
- Nicole Walter
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Rita Cervera-Juanes
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Christina Zheng
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Priscila Darakjian
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Denesa Lockwood
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Verginia Cuzon-Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Karina Ray
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Suzanne Fei
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Don Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Robert Searles
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, Oregon, USA
| | - Kathleen Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
19
|
Robinson DL, Amodeo LR, Chandler LJ, Crews FT, Ehlers CL, Gómez-A A, Healey KL, Kuhn CM, Macht VA, Marshall SA, Swartzwelder HS, Varlinskaya EI, Werner DF. The role of sex in the persistent effects of adolescent alcohol exposure on behavior and neurobiology in rodents. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:305-340. [PMID: 34696877 DOI: 10.1016/bs.irn.2021.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alcohol drinking is often initiated during adolescence, and this frequently escalates to binge drinking. As adolescence is also a period of dynamic neurodevelopment, preclinical evidence has highlighted that some of the consequences of binge drinking can be long lasting with deficits persisting into adulthood in a variety of cognitive-behavioral tasks. However, while the majority of preclinical work to date has been performed in male rodents, the rapid increase in binge drinking in adolescent female humans has re-emphasized the importance of addressing alcohol effects in the context of sex as a biological variable. Here we review several of the consequences of adolescent ethanol exposure in light of sex as a critical biological variable. While some alcohol-induced outcomes, such as non-social approach/avoidance behavior and sleep disruption, are generally consistent across sex, others are variable across sex, such as alcohol drinking, sensitivity to ethanol, social anxiety-like behavior, and induction of proinflammatory markers.
Collapse
Affiliation(s)
- Donita L Robinson
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| | - Leslie R Amodeo
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Department of Psychology, California State University, San Bernardino, CA, United States
| | - L Judson Chandler
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Fulton T Crews
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cindy L Ehlers
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Department of Neuroscience, Scripps Research, La Jolla, CA, United States
| | - Alexander Gómez-A
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kati L Healey
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Department of Psychiatry and Behavioral Sciences, School of Medicine, Duke University, Durham, NC, United States
| | - Cynthia M Kuhn
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, NC, United States
| | - Victoria A Macht
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - S Alexander Marshall
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Biological and Biomedical Sciences Department, North Carolina Central University, Durham, NC, United States
| | - H Scott Swartzwelder
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Department of Psychiatry and Behavioral Sciences, School of Medicine, Duke University, Durham, NC, United States
| | - Elena I Varlinskaya
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, United States
| | - David F Werner
- Neurobiology of Adolescent Drinking in Adulthood Consortium (NADIA), United States; Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, United States
| |
Collapse
|
20
|
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: 7] [Impact Index Per Article: 1.8] [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
|
21
|
Crews FT, Fisher R, Deason C, Vetreno RP. Loss of Basal Forebrain Cholinergic Neurons Following Adolescent Binge Ethanol Exposure: Recovery With the Cholinesterase Inhibitor Galantamine. Front Behav Neurosci 2021; 15:652494. [PMID: 33716687 PMCID: PMC7953159 DOI: 10.3389/fnbeh.2021.652494] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Binge drinking and alcohol abuse are common during adolescence and cause both cognitive deficits and lasting cholinergic pathology in the adult basal forebrain. Acetylcholine is anti-inflammatory and studies using the preclinical adolescent intermittent ethanol (AIE; 5.0 g/kg, i.g., 2 day on/2 day off from postnatal day [P]25 to P54) model of human adolescent binge drinking report decreased basal forebrain cholinergic neurons (BFCNs) and induction of proinflammatory genes that persist long into adulthood. Recent studies link AIE-induced neuroimmune activation to cholinergic pathology, but the underlying mechanisms contributing to the persistent loss of BFCNs are unknown. We report that treatment with the cholinesterase inhibitor galantamine (4.0 mg/kg, i.p.) administered during AIE (i.e., P25-P54) or following the conclusion of AIE (i.e., P57-P72) recovered the persistent loss of cholinergic neuron phenotype markers (i.e., ChAT, TrkA, and p75NTR) and somal shrinkage of residual ChAT + neurons known to persist in AIE-exposed adults. Galantamine treatment also recovered the AIE-increased expression of the proinflammatory receptors TLR4 and RAGE, the endogenous TLR4/RAGE agonist HMGB1, and the transcription activation marker pNF-κB p65. Interestingly, we find BFCNs express TLR4 and RAGE, and that AIE treatment increased pNF-κB p65 expression in adult ChAT + IR neurons, consistent with intracellular HMGB1-TLR4/RAGE signaling within BFCNs. AIE increased epigenetic transcription silencing markers (i.e., H3K9me2 and H3K9me3) in the adult basal forebrain and H3K9me2 occupancy at cholinergic phenotype gene promoters (i.e., ChAT and TrkA). The finding of no AIE-induced changes in total basal forebrain NeuN + neurons with galantamine reversal of AIE-induced ChAT + neuron loss, TLR4/RAGE-pNF-κB p65 signals, and epigenetic transcription silencing markers suggests that AIE does not cause cell death, but rather the loss of the cholinergic phenotype. Together, these data suggest that AIE induces HMGB1-TLR4/RAGE-pNF-κB p65 signals, causing the loss of cholinergic phenotype (i.e., ChAT, TrkA, and p75NTR) through epigenetic histone transcription silencing that result in the loss of the BFCN phenotype that can be prevented and restored by galantamine.
Collapse
Affiliation(s)
- Fulton T. Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rachael Fisher
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chloe Deason
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
22
|
Cannady R, Nguyen T, Padula AE, Rinker JA, Lopez MF, Becker HC, Woodward JJ, Mulholland PJ. Interaction of chronic intermittent ethanol and repeated stress on structural and functional plasticity in the mouse medial prefrontal cortex. Neuropharmacology 2021; 182:108396. [PMID: 33181147 PMCID: PMC7942177 DOI: 10.1016/j.neuropharm.2020.108396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/05/2020] [Accepted: 11/06/2020] [Indexed: 01/27/2023]
Abstract
Stress is a risk factor that plays a considerable role in the development and maintenance of alcohol (ethanol) abuse and relapse. Preclinical studies examining ethanol-stress interactions have demonstrated elevated ethanol drinking, cognitive deficits, and negative affective behaviors in mice. However, the neural adaptations in prefrontal cortical regions that drive these aberrant behaviors produced by ethanol-stress interactions are unknown. In this study, male C57BL/6J mice were exposed to chronic intermittent ethanol (CIE) and repeated forced swim stress (FSS). After two cycles of CIE x FSS, brain slices containing the prelimbic (PrL) and infralimbic (IfL) cortex were prepared for analysis of adaptations in dendritic spines and synaptic plasticity. In the PrL cortex, total spine density was increased in mice exposed to CIE. Immediately following induction of long-term potentiation (LTP), the fEPSP slope was increased in the PrL of CIE x FSS treated mice, indicative of a presynaptic adaptation on post-tetanic potentiation (PTP). In the IfL cortex, CIE exposure regardless of FSS experience resulted in an increase in spine density. FSS alone or when combined with CIE exposure increased PTP following LTP induction. Repeated FSS episodes increased IfL cortical paired-pulse facilitation, a second measure of presynaptic plasticity. In summary, CIE exposure resulted in structural adaptations while repeated stress exposure drove metaplastic changes in presynaptic function, demonstrating distinct morphological and functional changes in PrL and IfL cortical neurons. Thus, the structural and functional adaptations may be one mechanism underlying the development of excessive drinking and cognitive deficits associated with ethanol-stress interactions.
Collapse
Affiliation(s)
- Reginald Cannady
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA; Department of Biology, College of Science and Technology, North Carolina Agricultural & Technical State University, 1601 East Market Street, Barnes Hall 215, Greensboro, NC, 27411, USA
| | - Tiffany Nguyen
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Audrey E Padula
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Jennifer A Rinker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Marcelo F Lopez
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Howard C Becker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - John J Woodward
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA.
| |
Collapse
|
23
|
Bell MD, Pittman B, Petrakis I, Yoon G. Donepezil and cognitive remediation therapy to augment treatment of alcohol use disorder related mild cognitive impairment (AUD-MCI): An open label pilot study with historical controls. Subst Abus 2020; 42:412-416. [PMID: 33284058 DOI: 10.1080/08897077.2020.1844847] [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: 10/22/2022]
Abstract
BACKGROUND Pharmacological and psychosocial interventions have only modest efficacy for Alcohol Use Disorder (AUD), and continued alcohol use has brain effects on neurocognition, which place heavy drinkers at increased risk of early onset dementia. The most common neurocognitive deficits are in the domains of executive function and memory, and these deficits may impact AUD treatment outcomes. AUD related Mild Cognitive Impairment (AUD-MCI) is a diagnosis in DSM-V and ICD-10. Donepezil, a cholinesterase inhibitor, is currently FDA approved for treatment of dementia, and recent preclinical research suggests anticholinesterase agents may treat alcoholism. Another approach to cognitive recovery is Cognitive Remediation Therapy (CRT). Recent research supports CRT efficacy in schizophrenia and related disorders, and some research suggests that CRT could improve neurocognition in substance abuse disorders (SUDs). This is the first report of an open-label clinical trial that combined donepezil with CRT to improve neurocognitive and clinical outcomes in the early phase of AUD recovery. Methods: Eleven older male US Veterans with AUD-MCI as determined by Level II neurocognitive criteria and who had recently relapsed participated in an open-label trial of 13 weeks of donepezil combined with CRT. They were compared with matched historical control samples on neurocognitive and clinical outcomes. Results: Participants had excellent adherence to donepezil and CRT. Neurocognitive improvements were highly significant on a composite score of learning and memory and executive function measures (p < .0001) and was significantly better than historical matched controls (p < .001). On a Clinical Global Impression scale, 90.9% of participants had a good clinical recovery compared with 59.5% of historical matched controls (p <.052). Conclusions: AUD-MCI has not received much research attention but is of considerable public health importance. Findings in this open-label trial of donepezil + CRT should encourage further investigation into the clinical benefit of this combined treatment for AUD-MCI.
Collapse
Affiliation(s)
- Morris D Bell
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA.,Psychiatry Service, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Brian Pittman
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ismene Petrakis
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA.,Psychiatry Service, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Gihyun Yoon
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA.,Psychiatry Service, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
24
|
Healey KL, Kibble S, Bell A, Hodges S, Swartzwelder HS. Effects of adolescent intermittent ethanol on hippocampal expression of glutamate homeostasis and astrocyte-neuronal tethering proteins in male and female rats. J Neurosci Res 2020; 99:1908-1921. [PMID: 33217775 DOI: 10.1002/jnr.24758] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/02/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
Adolescent alcohol drinking is widely recognized as a significant public health problem, and evidence is accumulating that sufficient levels of consumption during this critical period of brain development have an enduring impact on neural and behavioral function. Recent studies have indicated that adolescent intermittent ethanol (AIE) exposure alters astrocyte function, astrocyte-neuronal interactions, and related synaptic regulation and activity. However, few of those studies have included female animals, and a broader assessment of AIE effects on the proteins mediating astrocyte-mediated glutamate dynamics and synaptic function is needed. We measured synaptic membrane expression of several such proteins in the dorsal and ventral regions of the hippocampal formation (DH, VH) from male and female rats exposed to AIE or adolescent intermittent water. In the DH, AIE caused elevated expression of glutamate transporter 1 (GLT-1) in both males and females, elevated postsynaptic density 95 expression in females only, and diminished NMDA receptor subunit 2A expression in males only. AIE and sex interactively altered ephrin receptor A4 (EphA4) expression in the DH. In the VH, AIE elevated expression of the cystine/glutamate antiporter and the glutamate aspartate transporter 1 (GLAST) in males only. Compared to males, female animals expressed lower levels of GLT-1 in the DH and greater levels of ephrin receptor B6 (EphB6) in the VH, in the absence of AIE effects. These results support the growing literature indicating that adolescent alcohol exposure produces long-lasting effects on astrocyte function and astrocyte-neuronal interactions. The sex and subregion specificity of these effects have mechanistic implications for our understanding of AIE effects generally.
Collapse
Affiliation(s)
- Kati L Healey
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Sandra Kibble
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Amelia Bell
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Sierra Hodges
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
25
|
Bohnsack JP, Pandey SC. Histone modifications, DNA methylation, and the epigenetic code of alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 156:1-62. [PMID: 33461661 DOI: 10.1016/bs.irn.2020.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder (AUD) is a leading cause of morbidity and mortality. Despite AUD's substantial contributions to lost economic productivity and quality of life, there are only a limited number of approved drugs for treatment of AUD in the United States. This chapter will update progress made on the epigenetic basis of AUD, with particular focus on histone post-translational modifications and DNA methylation and how these two epigenetic mechanisms interact to contribute to neuroadaptive processes leading to initiation, maintenance and progression of AUD pathophysiology. We will also evaluate epigenetic therapeutic strategies that have arisen from preclinical models of AUD and epigenetic biomarkers that have been discovered in human populations with AUD.
Collapse
Affiliation(s)
- John Peyton Bohnsack
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States; Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
26
|
Rodd ZA, Hauser SR, Swartzwelder HS, Waeiss RA, Lahiri DK, Bell RL. Regulation of the deleterious effects of binge-like exposure to alcohol during adolescence by α7 nicotinic acetylcholine receptor agents: prevention by pretreatment with a α7 negative allosteric modulator and emulation by a α7 agonist in alcohol-preferring (P) male and female rats. Psychopharmacology (Berl) 2020; 237:2601-2611. [PMID: 32607619 PMCID: PMC7502519 DOI: 10.1007/s00213-020-05557-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/18/2020] [Indexed: 12/23/2022]
Abstract
RATIONALE AND OBJECTIVES Binge-like alcohol consumption during adolescence associates with several deleterious consequences during adulthood including an increased risk for developing alcohol use disorder (AUD) and other addictions. Replicated preclinical data has indicated that adolescent exposure to binge-like levels of alcohol results in a reduction of choline acetyltransferase (ChAT) and an upregulation in the α7 nicotinic receptor (α7). From this information, we hypothesized that the α7 plays a critical role in mediating the effects of adolescent alcohol exposure. METHODS Male and female P rats were injected with the α7 agonist AR-R17779 (AR) once during 6 time points between post-natal days (PND) 29-37. Separate groups were injected with the α7 negative allosteric modulator (NAM) dehydronorketamine (DHNK) 2 h before administration of 4 g/kg EtOH (14 total exposures) during PND 28-48. On PND 75, all rats were given access to water and ethanol (15 and 30%) for 6 consecutive weeks (acquisition). All rats were then deprived of EtOH for 2 weeks and then, alcohol was returned (relapse). RESULTS Administration of AR during adolescence significantly increased acquisition of alcohol consumption during adulthood and prolonged relapse drinking in P rats. In contrast, administration of DHNK prior to binge-like EtOH exposure during adolescence prevented the increase in alcohol consumption observed during acquisition of alcohol consumption and the enhancement of relapse drinking observed during adulthood. DISCUSSION The data indicate that α7 mediates the effects of alcohol during adolescence. The data also indicate that α7 NAMs are potential prophylactic agents to reduce the deleterious effects of adolescent alcohol abuse.
Collapse
Affiliation(s)
- Zachary A Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA.
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA.
| | - Sheketha R Hauser
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - H Scott Swartzwelder
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - R Aaron Waeiss
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Debomoy K Lahiri
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Richard L Bell
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
27
|
Healey KL, Kibble S, Hodges S, Reissner KJ, Testen A, Wills TA, Acheson SK, Siemsen BM, McFaddin JA, Scofield MD, Swartzwelder HS. Enduring alterations in hippocampal astrocytesynaptic proximity following adolescent alcohol exposure: reversal by gabapentin. Neural Regen Res 2020; 15:1496-1501. [PMID: 31997814 PMCID: PMC7059577 DOI: 10.4103/1673-5374.274339] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 09/29/2019] [Accepted: 10/07/2019] [Indexed: 11/27/2022] Open
Abstract
Adolescent alcohol abuse is a substantive public health problem that has been the subject of intensive study in recent years. Despite reports of a wide range of effects of adolescent intermittent ethanol (AIE) exposure on brain and behavior, little is known about the mechanisms that may underlie those effects, and even less about treatments that might reverse them. Recent studies from our laboratory have indicated that AIE produced enduring changes in astrocyte function and synaptic activity in the hippocampal formation, suggesting the possibility of an alteration in astrocyte-neuronal connectivity and function. We utilized astrocyte-specific, membrane restricted viral labeling paired with immunohistochemistry to perform confocal single cell astrocyte imaging, three-dimensional reconstruction, and quantification of astrocyte morphology in hippocampal area CA1 from adult rats after AIE. Additionally, we assessed the colocalization of astrocyte plasma membrane labeling with immunoreactivity for AMPA-(α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) glutamate receptor 1, an AMPA receptor subunit and established neuronal marker of excitatory synapses, as a metric of astrocyte-synapse proximity. AIE significantly reduced the colocalization of the astrocyte plasma membrane with synaptic marker puncta in adulthood. This is striking in that it suggests not only an alteration of the physical association of astrocytes with synapses by AIE, but one that lasts into adulthood - well after the termination of alcohol exposure. Perhaps even more notable, the AIE-induced reduction of astrocyte-synapse interaction was reversed by sub-chronic treatment with the clinically used agent, gabapentin (Neurontin), in adulthood. This suggests that a medication in common clinical use may have the potential to reverse some of the enduring effects of adolescent alcohol exposure on brain function. All animal experiments conducted were approved by the Duke University Institutional Animal Care and Use Committee (Protocol Registry Number A159-18-07) on July 27, 2018.
Collapse
Affiliation(s)
- Kati L. Healey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Sandra Kibble
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Sierra Hodges
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Kathryn J. Reissner
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Anze Testen
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Tiffany A. Wills
- Department of Cell Biology and Anatomy, LSU School of Medicine, New Orleans, LA, USA
| | - Shawn K. Acheson
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Benjamin M. Siemsen
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - John A. McFaddin
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Michael D. Scofield
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - H. Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
28
|
Brucato FH, Benjamin DE. Synaptic Pruning in Alzheimer's Disease: Role of the Complement System. GLOBAL JOURNAL OF MEDICAL RESEARCH 2020; 20:10.34257/gjmrfvol20is6pg1. [PMID: 32982106 PMCID: PMC7518506 DOI: 10.34257/gjmrfvol20is6pg1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alz heimer’s disease (AD) continues to threaten aged individuals and health care systems around the world. Human beings have been trying to postpone, reduce, or eliminate the primary risk factor for AD, aging, throughout history. Despite this, there is currently only symptomatic treatment for AD and this treatment is limited to only a handful of FDA approved AD drugs.
Collapse
Affiliation(s)
- Frederic H Brucato
- Cascade Biotechnology Inc., Princeton Corporate Plaza 1 Deer Park Dr., Suite D5. Monmouth Junction NJ 08852
| | - Daniel E Benjamin
- Cascade Biotechnology Inc., Princeton Corporate Plaza 1 Deer Park Dr., Suite D5. Monmouth Junction NJ 08852
| |
Collapse
|
29
|
Biological intersection of sex, age, and environment in the corticotropin releasing factor (CRF) system and alcohol. Neuropharmacology 2020; 170:108045. [PMID: 32217364 DOI: 10.1016/j.neuropharm.2020.108045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/13/2020] [Accepted: 03/06/2020] [Indexed: 01/21/2023]
Abstract
The neuropeptide corticotropin-releasing factor (CRF) is critical in neural circuit function and behavior, particularly in the context of stress, anxiety, and addiction. Despite a wealth of preclinical evidence for the efficacy of CRF receptor 1 antagonists in reducing behavioral pathology associated with alcohol exposure, several clinical trials have had disappointing outcomes, possibly due to an underappreciation of the role of biological variables. Although he National Institutes of Health (NIH) now mandate the inclusion of sex as a biological variable in all clinical and preclinical research, the current state of knowledge in this area is based almost entirely on evidence from male subjects. Additionally, the influence of biological variables other than sex has received even less attention in the context of neuropeptide signaling. Age (particularly adolescent development) and housing conditions have been shown to affect CRF signaling and voluntary alcohol intake, and the interaction between these biological variables is particularly relevant to the role of the CRF system in the vulnerability or resilience to the development of alcohol use disorder (AUD). Going forward, it will be important to include careful consideration of biological variables in experimental design, reporting, and interpretation. As new research uncovers conditions in which sex, age, and environment play major roles in physiological and/or pathological processes, our understanding of the complex interaction between relevant biological variables and critical signaling pathways like the CRF system in the cellular and behavioral consequences of alcohol exposure will continue to expand ultimately improving the ability of preclinical research to translate to the clinic. This article is part of the special issue on Neuropeptides.
Collapse
|
30
|
Mira RG, Lira M, Tapia-Rojas C, Rebolledo DL, Quintanilla RA, Cerpa W. Effect of Alcohol on Hippocampal-Dependent Plasticity and Behavior: Role of Glutamatergic Synaptic Transmission. Front Behav Neurosci 2020; 13:288. [PMID: 32038190 PMCID: PMC6993074 DOI: 10.3389/fnbeh.2019.00288] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022] Open
Abstract
Problematic alcohol drinking and alcohol dependence are an increasing health problem worldwide. Alcohol abuse is responsible for approximately 5% of the total deaths in the world, but addictive consumption of it has a substantial impact on neurological and memory disabilities throughout the population. One of the better-studied brain areas involved in cognitive functions is the hippocampus, which is also an essential brain region targeted by ethanol. Accumulated evidence in several rodent models has shown that ethanol treatment produces cognitive impairment in hippocampal-dependent tasks. These adverse effects may be related to the fact that ethanol impairs the cellular and synaptic plasticity mechanisms, including adverse changes in neuronal morphology, spine architecture, neuronal communication, and finally an increase in neuronal death. There is evidence that the damage that occurs in the different brain structures is varied according to the stage of development during which the subjects are exposed to ethanol, and even much earlier exposure to it would cause damage in the adult stage. Studies on the cellular and cognitive deficiencies produced by alcohol in the brain are needed in order to search for new strategies to reduce alcohol neuronal toxicity and to understand its consequences on memory and cognitive performance with emphasis on the crucial stages of development, including prenatal events to adulthood.
Collapse
Affiliation(s)
- Rodrigo G Mira
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile
| | - Matias Lira
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile.,Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Providencia, Chile
| | - Daniela L Rebolledo
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Escuela de Obstetricia y Puericultura and Centro Integrativo de Biología y Química Aplicada (CIBQA), Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Rodrigo A Quintanilla
- Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile.,Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Providencia, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
31
|
McCool BA, McGinnis MM. Adolescent Vulnerability to Alcohol Use Disorder: Neurophysiological Mechanisms from Preclinical Studies. Handb Exp Pharmacol 2020; 258:421-442. [PMID: 31595414 DOI: 10.1007/164_2019_296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Adolescent alcohol use in human populations dramatically increases the likelihood of adult alcohol use disorder. This adolescent vulnerability is recapitulated in preclinical models which provide important opportunities to understand basic neurobiological mechanisms. We provide here an overview of GABAergic and glutamatergic neurotransmission and our current understanding of the sensitivity of these systems to adolescent ethanol exposure. As a whole, the preclinical literature suggests that adolescent vulnerability may be directly related to region-specific neurobiological processes that continue to develop during adolescence. These processes include the activity of intrinsic circuits within diverse brain regions (primarily represented by GABAergic neurotransmission) and activity-dependent regulation of synaptic strength at glutamatergic synapses. Furthermore, GABAergic and glutamatergic neurotransmission within regions/circuits that regulate cognitive function, emotion, and their integration appears to be the most vulnerable to adolescent ethanol exposure. Finally, using documented behavioral differences between adolescents and adults with respect to acute ethanol, we highlight additional circuits and regions for future study.
Collapse
Affiliation(s)
- Brian A McCool
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Molly M McGinnis
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
32
|
Nunes PT, Kipp BT, Reitz NL, Savage LM. Aging with alcohol-related brain damage: Critical brain circuits associated with cognitive dysfunction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:101-168. [PMID: 31733663 PMCID: PMC7372724 DOI: 10.1016/bs.irn.2019.09.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcoholism is associated with brain damage and impaired cognitive functioning. The relative contributions of different etiological factors, such as alcohol, thiamine deficiency and age vulnerability, to the development of alcohol-related neuropathology and cognitive impairment are still poorly understood. One reason for this quandary is that both alcohol toxicity and thiamine deficiency produce brain damage and cognitive problems that can be modulated by age at exposure, aging following alcohol toxicity or thiamine deficiency, and aging during chronic alcohol exposure. Pre-clinical models of alcohol-related brain damage (ARBD) have elucidated some of the contributions of ethanol toxicity and thiamine deficiency to neuroinflammation, neuronal loss and functional deficits. However, the critical variable of age at the time of exposure or long-term aging with ARBD has been relatively ignored. Acute thiamine deficiency created a massive increase in neuroimmune genes and proteins within the thalamus and significant increases within the hippocampus and frontal cortex. Chronic ethanol treatment throughout adulthood produced very minor fluctuations in neuroimmune genes, regardless of brain region. Intermittent "binge-type" ethanol during the adolescent period established an intermediate neuroinflammatory response in the hippocampus and frontal cortex, that can persist into adulthood. Chronic excessive drinking throughout adulthood, adolescent intermittent ethanol exposure, and thiamine deficiency all led to a loss of the cholinergic neuronal phenotype within the basal forebrain, reduced hippocampal neurogenesis, and alterations in the frontal cortex. Only thiamine deficiency results in gross pathological lesions of the thalamus. The behavioral impairment following these types of treatments is hierarchical: Thiamine deficiency produces the greatest impairment of hippocampal- and prefrontal-dependent behaviors, chronic ethanol drinking ensues mild impairments on both types of tasks and adolescent intermittent ethanol exposure leads to impairments on frontocortical tasks, with sparing on most hippocampal-dependent tasks. However, our preliminary data suggest that as rodents age following adolescent intermittent ethanol exposure, hippocampal functional deficits began to emerge. A necessary requirement for the advancement of understanding the neural consequences of alcoholism is a more comprehensive assessment and understanding of how excessive alcohol drinking at different development periods (adolescence, early adulthood, middle-aged and aged) influences the trajectory of the aging process, including pathological aging and disease.
Collapse
Affiliation(s)
- Polliana Toledo Nunes
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Brian T Kipp
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Nicole L Reitz
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Lisa M Savage
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States.
| |
Collapse
|
33
|
Hauser SR, Knight CP, Truitt WA, Waeiss RA, Holt IS, Carvajal GB, Bell RL, Rodd ZA. Adolescent Intermittent Ethanol Increases the Sensitivity to the Reinforcing Properties of Ethanol and the Expression of Select Cholinergic and Dopaminergic Genes within the Posterior Ventral Tegmental Area. Alcohol Clin Exp Res 2019; 43:1937-1948. [DOI: 10.1111/acer.14150] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/10/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Sheketha R. Hauser
- Department of Psychiatry Indiana University School of Medicine Indianapolis Indiana
| | | | - William A. Truitt
- Department of Psychiatry Indiana University School of Medicine Indianapolis Indiana
| | - Robert Aaron Waeiss
- Program in Medical Neuroscience Paul and Carole Stark Neurosciences Research Institute Indiana University School of Medicine Indianapolis Indiana
| | - Ian S. Holt
- Department of Psychiatry Indiana University School of Medicine Indianapolis Indiana
| | - Gustavo B. Carvajal
- Department of Psychiatry Indiana University School of Medicine Indianapolis Indiana
| | - Richard L. Bell
- Department of Psychiatry Indiana University School of Medicine Indianapolis Indiana
| | - Zachary A. Rodd
- Department of Psychiatry Indiana University School of Medicine Indianapolis Indiana
| |
Collapse
|
34
|
Swartzwelder HS, Healey KL, Liu W, Dubester K, Miller KM, Crews FT. Changes in Neuroimmune and Neuronal Death Markers after Adolescent Alcohol Exposure in Rats are Reversed by Donepezil. Sci Rep 2019; 9:12110. [PMID: 31431637 PMCID: PMC6702347 DOI: 10.1038/s41598-019-47039-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 06/25/2019] [Indexed: 12/29/2022] Open
Abstract
Adolescent intermittent ethanol (AIE) exposure diminishes neurogenesis and dendritic spine density in the dentate gyrus. The cholinesterase inhibitor, donepezil (Aricept), reverses AIE effects on dendritic spines, possibly by interacting with inflammatory and/or epigenetic mediators after AIE exposure. This study tests the hypothesis that donepezil reverses AIE-induced neuroimmune, and epigenetic changes in the adult dentate gyrus. Adolescent Sprague-Dawley male rats (PD30-43) were given 10 intermittent, intragastric doses of ethanol (5.0 g/kg) or isovolumetric water (AIW). Twenty-one days later half of the animals from each group were treated with either donepezil or isovolumetric water (i.g.) once daily for four days. Two hours after the last donepezil or water dose animals were sacrificed and brains prepared for immunohistochemical analyses. AIE reduced immunoreactivity for doublecortin (DCX) and increased immunoreactivity for activated caspase-3 and death receptor-3 in adulthood, suggesting an enduring attenuation of neurogenesis and an increase in progenitor death. These effects were reversed by donepezil treatment in adulthood. AIE also increased immunoreactivity for the inflammatory signaling molecules HMGB1 and RAGE, as well as the activated phosphorylated transcription factor pNFκB p65, and the gene silencing marker dimethylated histone H3K9. All of these AIE effects were also reversed by donepezil, with the exception of HMGB1.
Collapse
Affiliation(s)
- H S Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, N.C., USA.
| | - Kati L Healey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, N.C., USA
| | - Wen Liu
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, N.C., USA
| | - Kira Dubester
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, N.C., USA
| | - Kelsey M Miller
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, N.C., USA
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, N.C., USA
| |
Collapse
|
35
|
Crews FT, Robinson DL, Chandler LJ, Ehlers CL, Mulholland PJ, Pandey SC, Rodd ZA, Spear LP, Swartzwelder HS, Vetreno RP. Mechanisms of Persistent Neurobiological Changes Following Adolescent Alcohol Exposure: NADIA Consortium Findings. Alcohol Clin Exp Res 2019; 43:1806-1822. [PMID: 31335972 PMCID: PMC6758927 DOI: 10.1111/acer.14154] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 07/12/2019] [Indexed: 12/13/2022]
Abstract
The Neurobiology of Adolescent Drinking in Adulthood (NADIA) Consortium has focused on the impact of adolescent binge drinking on brain development, particularly on effects that persist into adulthood. Adolescent binge drinking is common, and while many factors contribute to human brain development and alcohol use during adolescence, animal models are critical for understanding the specific consequences of alcohol exposure during this developmental period and the underlying mechanisms. Using adolescent intermittent ethanol (AIE) exposure models, NADIA investigators identified long-lasting AIE-induced changes in adult behavior that are consistent with observations in humans, such as increased alcohol drinking, increased anxiety (particularly social anxiety), increased impulsivity, reduced behavioral flexibility, impaired memory, disrupted sleep, and altered responses to alcohol. These behavioral changes are associated with multiple molecular, cellular, and physiological alterations in the brain that persist long after AIE exposure. At the molecular level, AIE results in long-lasting changes in neuroimmune/trophic factor balance and epigenetic-microRNA (miRNA) signaling across glia and neurons. At the cellular level, AIE history is associated in adulthood with reduced expression of cholinergic, serotonergic, and dopaminergic neuron markers, attenuated cortical thickness, decreased neurogenesis, and altered dendritic spine and glial morphology. This constellation of molecular and cellular adaptations to AIE likely contributes to observed alterations in neurophysiology, measured by synaptic physiology, EEG patterns, and functional connectivity. Many of these AIE-induced brain changes replicate findings seen in postmortem brains of humans with alcohol use disorder (AUD). NADIA researchers are now elucidating mechanisms of these adaptations. Emerging data demonstrate that exercise, antiinflammatory drugs, anticholinesterases, histone deacetylase inhibitors, and other pharmacological compounds are able to prevent (administered during AIE) and/or reverse (given after AIE) AIE-induced pathology in adulthood. These studies support hypotheses that adolescent binge drinking increases risk of adult hazardous drinking and influences brain development, and may provide insight into novel therapeutic targets for AIE-induced neuropathology and AUDs.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Donita L Robinson
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - L Judson Chandler
- Department of Neuroscience, Charleston Alcohol Research Center, Charleston, South Carolina
| | - Cindy L Ehlers
- Department of Neurosciences, The Scripps Research Institute, La Jolla, California
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Charleston, South Carolina
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Zachary A Rodd
- Department of Psychiatry and Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Linda P Spear
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
36
|
Dulman RS, Auta J, Teppen T, Pandey SC. Acute Ethanol Produces Ataxia and Induces Fmr1 Expression via Histone Modifications in the Rat Cerebellum. Alcohol Clin Exp Res 2019; 43:1191-1198. [PMID: 30969437 DOI: 10.1111/acer.14044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/04/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The cerebellum is fundamental for motor coordination and therefore crucial in ethanol (EtOH)-induced ataxia. EtOH contributes to cerebellar pathophysiology. Fragile-X mental retardation protein (FMRP) is a complex regulator of RNA and synaptic plasticity implicated in fragile-X tremor and ataxia syndrome, a phenotype featuring increased Fmr1 mRNA expression. Recent studies have implicated glutamatergic targets of FMRP in hereditary cerebellar ataxias including the main cerebellar excitatory amino acid (Eaa1) transporter and a subtype of metabotropic glutamate receptor (Grm5). However, EtOH-induced changes in cerebellar Fmr1 expression and its epigenetic regulation have not been investigated. Here, we examined the effects of acute EtOH exposure on ataxic behavior, gene expression, and epigenetic regulation of the Fmr1 gene and its glutamatergic targets in the rat cerebellum. METHODS Male adult Sprague Dawley rats received acute EtOH (2 g/kg) intraperitoneally 1 hour prior to ataxic behavioral testing on the accelerating rotarod and were sacrificed immediately thereafter. Cerebellar tissues were analyzed for gene expression and epigenetic regulation of the Fmr1 gene and its glutamatergic targets in the rat cerebellum using real-time quantitative polymerase chain reaction (PCR) and chromatin immunoprecipitation. RESULTS Acute EtOH exposure caused marked ataxia on the accelerating rotarod test compared with saline-treated controls. This ataxic response was associated with increases in mRNA levels of Fmr1, postsynaptic density 95 (Psd95), Eaa1, and Grm5 in the cerebellum. In addition, we found increased H3K27 acetylation both at the promoter region of Fmr1 and at a proposed cyclic adenosine monophosphate (cAMP) response-element binding (CREB) site downstream of the Fmr1 transcription start site. Furthermore, acute EtOH exposure significantly increased Creb1 and the histone acetyltransferases (HAT) CREB binding protein (Cbp), and p300 mRNA transcripts. CONCLUSIONS Overall, EtOH regulates cerebellar Fmr1 expression most likely via HAT-mediated increase in histone acetylation. We propose that FMRP regulation of glutamatergic transcripts plays an important role in disrupting the excitatory-inhibitory balance in the cerebellum underlying EtOH-induced ataxia.
Collapse
Affiliation(s)
- Russell S Dulman
- Department of Psychiatry, Center for Alcohol Research in Epigenetics, University of Illinois at Chicago, Chicago, Illinois
| | - James Auta
- Department of Psychiatry, Center for Alcohol Research in Epigenetics, University of Illinois at Chicago, Chicago, Illinois
| | - Tara Teppen
- Department of Psychiatry, Center for Alcohol Research in Epigenetics, University of Illinois at Chicago, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Subhash C Pandey
- Department of Psychiatry, Center for Alcohol Research in Epigenetics, University of Illinois at Chicago, Chicago, Illinois.,Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
37
|
Cacabelos R, Carril JC, Sanmartín A, Cacabelos P. Pharmacoepigenetic Processors: Epigenetic Drugs, Drug Resistance, Toxicoepigenetics, and Nutriepigenetics. PHARMACOEPIGENETICS 2019:191-424. [DOI: 10.1016/b978-0-12-813939-4.00006-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
38
|
Getachew B, Hudson T, Heinbockel T, Csoka AB, Tizabi Y. Protective Effects of Donepezil Against Alcohol-Induced Toxicity in Cell Culture: Role of Caspase-3. Neurotox Res 2018; 34:757-762. [PMID: 29804239 DOI: 10.1007/s12640-018-9913-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/29/2018] [Accepted: 05/17/2018] [Indexed: 01/12/2023]
Abstract
Ethanol (EtOH) is one of the most frequently abused drugs with heavy health, economic, and societal burdens. Although moderate to low EtOH may have some neuroprotective effects, heavy EtOH consumption associated with high blood alcohol level (BAL) can be quite detrimental. The brain is particularly vulnerable to the damaging effects of high BAL, leading to neuronal loss, cognitive, and behavioral deficits. Although the exact causes of these detriments are not fully elucidated, it is believed that damage to the cholinergic system is at least partially responsible for the cognitive impairment. Thus, high BAL may result in selective apoptotic damage to the cholinergic neurons. Donepezil (DON), a centrally acting, reversible and non-competitive acetylcholinesterase (AChE) inhibitor, approved for use in Alzheimer's disease (AD), may also attenuate EtOH-induced cognitive impairment. Cognitive effects of DON might be due to an anti-apoptotic activity as some AChE inhibitors have been shown to have this property. The aim of this study was to determine whether DON might protect against EtOH-induced toxicity and whether such protection might be apoptotically mediated. We exposed the human neuroblastoma-derived, SH-SY5Y cells to a relatively high concentration of EtOH (500 mM) for 24 h and evaluated the effects of two concentrations of DON (0.1 and 1.0 μM) on alcohol-induced toxicity and caspase-3, an apoptotic marker. We found a dose-dependent protection of DON against EtOH-induced toxicity as well as dose-dependent attenuation of EtOH-induced increases in caspase-3 levels. Thus, DON may inhibit apoptosis as well as alcohol-induced toxicity.
Collapse
Affiliation(s)
- Bruk Getachew
- Department of Pharmacology, Howard University College Medicine, 520 W Street NW, Washington, DC, 20059, USA
| | - Tamaro Hudson
- Department of Pharmacology, Howard University College Medicine, 520 W Street NW, Washington, DC, 20059, USA
| | - Thomas Heinbockel
- Department of Anatomy, Howard University College Medicine, Washington, DC, 20059, USA
| | - Antonei B Csoka
- Department of Anatomy, Howard University College Medicine, Washington, DC, 20059, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College Medicine, 520 W Street NW, Washington, DC, 20059, USA.
| |
Collapse
|