1
|
Hong N, Yoon SR, Ahn JC. Photobiomodulation using an 830-nm laser alleviates hippocampal reactive gliosis and cognitive dysfunction in a mouse model of adolescent chronic alcohol exposure. Pharmacol Biochem Behav 2025; 248:173956. [PMID: 39793712 DOI: 10.1016/j.pbb.2025.173956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 08/14/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
Chronic alcoholism is known to have detrimental effects on the brain, including cognitive impairment, neurotransmitter imbalances, and brain atrophy. The hippocampus, crucial for spatial memory and cognitive functions, is particularly susceptible to alcohol-induced changes. Photobiomodulation (PBM), a non-invasive therapeutic method that utilizes red or near-infrared light, has shown promising applications in the central and peripheral nervous systems. Near-infrared (NIR) light, in particular, has been shown to prevent apoptosis, and neuroinflammation, as well as to improve cognitive functions. In this study, we aimed to investigate whether 830-nm laser irradiation could mitigate cognitive deficits in a chronic alcohol mouse model. Chronic alcoholism was induced in C57BL/6 mice through continuous ethanol gavage for 4 weeks at a dosage of 5 g/kg/day. Gavaging was performed 3 times per week for 4 weeks. Mice were transcranial irradiated by 830-nm laser, following making a chronic alcohol mouse model. Laser irradiation (50 mW/cm2) was performed 5 times per week for 3 weeks. To verify memory and cognitive defeats of a chronic alcohol mouse model, we performed animal behavior tasks such as Morris water maze, Y maze, and novel objective recognition. Our results confirmed the cognitive impairment in the chronic alcohol mouse model compared to the control group in conducted tasks. However, cognitive and spatial memory significantly improved following 830-nm laser irradiation. Additionally, we confirmed whether the behavior tasks result from histological changes. We performed immunofluorescence staining in the hippocampus region (CA3, CA1 and hilus) using astrocyte (GFAP) and microglia (Iba1) markers. As a result, reactive astrocyte was significantly increased in the chronic alcohol mouse model compared to control mice, whereas the number of GFAP-positive cells was significantly reduced by 830-nm laser irradiation. These findings indicate that chronic alcohol exposure induces spatial memory and cognitive impairment, which can be effectively rescued through near-infrared laser irradiation.
Collapse
Affiliation(s)
- Namgue Hong
- Medical Laser Research Center, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Sung-Ryeong Yoon
- Department of Medical Science, Graduate School of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Chul Ahn
- Medical Laser Research Center, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomedical Science, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
2
|
Macht V, de Castro S, Vetreno RP. Impact of Neuroimmune System Activation by Adolescent Binge Alcohol Exposure on Adult Neurobiology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:179-208. [PMID: 40128480 DOI: 10.1007/978-3-031-81908-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Adolescence is a conserved neurodevelopmental period encompassing maturation of glia and the innate immune system that parallels refinement of brain structures, neurotransmitter systems, and neurocircuitry. Given the vast neurodevelopmental processes occurring during adolescence, spanning brain structural and neurocircuitry refinement to maturation of neurotransmitter systems, glia, and the innate immune system, insults incurred during this critical period of neurodevelopment, could have profound effects on brain function and behavior that persist into adulthood. Adolescent binge drinking is common and associated with many adverse outcomes that may underlie the lifelong increased risk of alcohol-related problems and development of an alcohol use disorder (AUD). In this chapter, we examined the impact of adolescent binge drinking models using the adolescent intermittent ethanol (AIE) model on adult neurobiology. These studies implicate proinflammatory neuroimmune signaling across glia and neurons in persistent AIE-induced neuropathology. Some of these changes are reversible, providing unique opportunities for the development of treatments to prevent many of the long-term consequences of adolescent alcohol misuse.
Collapse
Affiliation(s)
- Victoria Macht
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sagan de Castro
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Song H, Yang P, Zhang X, Tao R, Zuo L, Liu W, Fu J, Kong Z, Tang R, Wu S, Pang L, Zhang X. Atypical effective connectivity from the frontal cortex to striatum in alcohol use disorder. Transl Psychiatry 2024; 14:381. [PMID: 39294121 PMCID: PMC11411137 DOI: 10.1038/s41398-024-03083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/23/2024] [Accepted: 08/30/2024] [Indexed: 09/20/2024] Open
Abstract
Alcohol use disorder (AUD) is a profound psychiatric condition marked by disrupted connectivity among distributed brain regions, indicating impaired functional integration. Previous connectome studies utilizing functional magnetic resonance imaging (fMRI) have predominantly focused on undirected functional connectivity, while the specific alterations in directed effective connectivity (EC) associated with AUD remain unclear. To address this issue, this study utilized multivariate pattern analysis (MVPA) and spectral dynamic causal modeling (DCM). We recruited 32 abstinent men with AUD and 30 healthy controls (HCs) men, and collected their resting-state fMRI data. A regional homogeneity (ReHo)-based MVPA method was employed to classify AUD and HC groups, as well as predict the severity of addiction in AUD individuals. The most informative brain regions identified by the MVPA were further investigated using spectral DCM. Our results indicated that the ReHo-based support vector classification (SVC) exhibits the highest accuracy in distinguishing individuals with AUD from HCs (classification accuracy: 98.57%). Additionally, our results demonstrated that ReHo-based support vector regression (SVR) could be utilized to predict the addiction severity (alcohol use disorders identification test, AUDIT, R2 = 0.38; Michigan alcoholism screening test, MAST, R2 = 0.29) of patients with AUD. The most informative brain regions for the prediction include left pre-SMA, right dACC, right LOFC, right putamen, and right NACC. These findings were validated in an independent data set (35 patients with AUD and 36 HCs, Classification accuracy: 91.67%; AUDIT, R2 = 0.17; MAST, R2 = 0.20). The results of spectral DCM analysis indicated that individuals with AUD exhibited decreased EC from the left pre-SMA to the right putamen, from the right dACC to the right putamen, and from the right LOFC to the right NACC compared to HCs. Moreover, the EC strength from the right NACC to left pre-SMA and from the right dACC to right putamen mediated the relationship between addiction severity (MAST scores) and behavioral measures (impulsive and compulsive scores). These findings provide crucial evidence for the underlying mechanism of impaired self-control, risk assessment, and impulsive and compulsive alcohol consumption in individuals with AUD, providing novel causal insights into both diagnosis and treatment.
Collapse
Affiliation(s)
- Hongwen Song
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Philosophy and Social Science of Anhui Province on Adolescent Mental Health and Crisis Intelligence Intervention, Hefei Normal University, Hefei, China
- The Institute of Linguistics and Applied Linguistics, Anhui Jianzhu University, Hefei, China
| | - Ping Yang
- Department of Psychology, School of Humanities and Social Science, University of Science and Technology of China, Hefei, China
| | - Xinyue Zhang
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Rui Tao
- Department of Substance-Related Disorders, Hefei Fourth People's Hospital, Hefei, China
| | - Lin Zuo
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Weili Liu
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaxin Fu
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhuo Kong
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Rui Tang
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Siyu Wu
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Liangjun Pang
- Department of Substance-Related Disorders, Hefei Fourth People's Hospital, Hefei, China.
| | - Xiaochu Zhang
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Psychology, School of Humanities and Social Science, University of Science and Technology of China, Hefei, China.
- School of Mental Health, Bengbu Medical College, Bengbu, China.
- Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China.
- Institute of Health and Medicine, Hefei Comprehensive Science Center, Hefei, China.
| |
Collapse
|
4
|
Crews FT, Coleman LG, Macht VA, Vetreno RP. Alcohol, HMGB1, and Innate Immune Signaling in the Brain. Alcohol Res 2024; 44:04. [PMID: 39135668 PMCID: PMC11318841 DOI: 10.35946/arcr.v44.1.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Binge drinking (i.e., consuming enough alcohol to achieve a blood ethanol concentration of 80 mg/dL, approximately 4-5 drinks within 2 hours), particularly in early adolescence, can promote progressive increases in alcohol drinking and alcohol-related problems that develop into compulsive use in the chronic relapsing disease, alcohol use disorder (AUD). Over the past decade, neuroimmune signaling has been discovered to contribute to alcohol-induced changes in drinking, mood, and neurodegeneration. This review presents a mechanistic hypothesis supporting high mobility group box protein 1 (HMGB1) and Toll-like receptor (TLR) signaling as key elements of alcohol-induced neuroimmune signaling across glia and neurons, which shifts gene transcription and synapses, altering neuronal networks that contribute to the development of AUD. This hypothesis may help guide further research on prevention and treatment. SEARCH METHODS The authors used the search terms "HMGB1 protein," "alcohol," and "brain" across PubMed, Scopus, and Embase to find articles published between 1991 and 2023. SEARCH RESULTS The database search found 54 references in PubMed, 47 in Scopus, and 105 in Embase. A total of about 100 articles were included. DISCUSSION AND CONCLUSIONS In the brain, immune signaling molecules play a role in normal development that differs from their functions in inflammation and the immune response, although cellular receptors and signaling are shared. In adults, pro-inflammatory signals have emerged as contributing to brain adaptation in stress, depression, AUD, and neurodegenerative diseases. HMGB1, a cytokine-like signaling protein released from activated cells, including neurons, is hypothesized to activate pro-inflammatory signals through TLRs that contribute to adaptations to binge and chronic heavy drinking. HMGB1 alone and in heteromers with other molecules activates TLRs and other immune receptors that spread signaling across neurons and glia. Both blood and brain levels of HMGB1 increase with ethanol exposure. In rats, an adolescent intermittent ethanol (AIE) binge drinking model persistently increases brain HMGB1 and its receptors; alters microglia, forebrain cholinergic neurons, and neuronal networks; and increases alcohol drinking and anxiety while disrupting cognition. Studies of human postmortem AUD brain have found elevated levels of HMGB1 and TLRs. These signals reduce cholinergic neurons, whereas microglia, the brain's immune cells, are activated by binge drinking. Microglia regulate synapses through complement proteins that can change networks affected by AIE that increase drinking, contributing to risks for AUD. Anti-inflammatory drugs, exercise, cholinesterase inhibitors, and histone deacetylase epigenetic inhibitors prevent and reverse the AIE-induced pathology. Further, HMGB1 antagonists and other anti-inflammatory treatments may provide new therapies for alcohol misuse and AUD. Collectively, these findings suggest that restoring the innate immune signaling balance is central to recovering from alcohol-related pathology.
Collapse
Affiliation(s)
- Fulton T. Crews
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Leon G. Coleman
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Victoria A. Macht
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
5
|
Wank I, Mittmann C, Kreitz S, Chestnykh D, Mühle C, Kornhuber J, Ludwig A, Kalinichenko LS, Müller CP, Hess A. Neutral sphingomyelinase controls acute and chronic alcohol effects on brain activity. Neuropharmacology 2024; 253:109948. [PMID: 38636728 DOI: 10.1016/j.neuropharm.2024.109948] [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: 01/11/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
Alcohol consumption is a widespread phenomenon throughout the world. However, how recreational alcohol use evolves into alcohol use disorder (AUD) remains poorly understood. The Smpd3 gene and its coded protein neutral sphingomyelinase (NSM) are associated with alcohol consumption in humans and alcohol-related behaviors in mice, suggesting a potential role in this transition. Using multiparametric magnetic resonance imaging, we characterized the role of NSM in acute and chronic effects of alcohol on brain anatomy and function in female mice. Chronic voluntary alcohol consumption (16 vol% for at least 6 days) affected brain anatomy in WT mice, reducing regional structure volume predominantly in cortical regions. Attenuated NSM activity prevented these anatomical changes. Functional MRI linked these anatomical adaptations to functional changes: Chronic alcohol consumption in mice significantly modulated resting state functional connectivity (RS FC) in response to an acute ethanol challenge (i.p. bolus of 2 g kg-1) in heterozygous NSM knockout (Fro), but not in WT mice. Acute ethanol administration in alcohol-naïve WT mice significantly decreased RS FC in cortical and brainstem regions, a key finding that was amplified in Fro mice. Regarding direct pharmacological effects, acute ethanol administration increased the regional cerebral blood volume (rCBV) in many brain areas. Here, chronic alcohol consumption otherwise attenuated the acute rCBV response in WT mice but enhanced it in Fro mice. Altogether, these findings suggest a differential role for NSM in acute and chronic functional brain responses to alcohol. Therefore, targeting NSM may be useful in the prevention or treatment of AUD.
Collapse
Affiliation(s)
- Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Claire Mittmann
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91045, Erlangen, Germany
| | - Silke Kreitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany; Department of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Daria Chestnykh
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91045, Erlangen, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91045, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91045, Erlangen, Germany
| | - Andreas Ludwig
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91045, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91045, Erlangen, Germany; Centre for Drug Research, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia; Institute of Psychopharmacology, Central Institute of Mental Health, University of Heidelberg, 68159, Heidelberg, Germany
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany; Department of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany; FAU NeW - Research Center for New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany.
| |
Collapse
|
6
|
Pochapski JA, Gómez-A A, Stringfield SJ, Jaggers H, Boettiger CA, Da Cunha C, Robinson DL. Adolescent alcohol exposure persistently alters orbitofrontal cortical encoding of Pavlovian conditional stimulus components in female rats. Sci Rep 2024; 14:13775. [PMID: 38877100 PMCID: PMC11178901 DOI: 10.1038/s41598-024-64036-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024] Open
Abstract
Exposure to alcohol during adolescence impacts cortical and limbic brain regions undergoing maturation. In rodent models, long-term effects on behavior and neurophysiology have been described after adolescent intermittent ethanol (AIE), especially in males. We hypothesized that AIE in female rats increases conditional approach to a reward-predictive cue and corresponding neuronal activity in the orbitofrontal cortex (OFC) and nucleus accumbens (NAc). We evaluated behavior and neuronal firing after AIE (5 g/kg intragastric) or water (CON) in adult female rats. Both AIE and CON groups expressed a ST phenotype, and AIE marginally increased sign-tracking (ST) and decreased goal-tracking (GT) metrics. NAc neurons exhibited phasic firing patterns to the conditional stimulus (CS), with no differences between groups. In contrast, neuronal firing in the OFC of AIE animals was greater at CS onset and offset than in CON animals. During reward omission, OFC responses to CS offset normalized to CON levels, but enhanced OFC firing to CS onset persisted in AIE. We suggest that the enhanced OFC neural activity observed in AIE rats to the CS could contribute to behavioral inflexibility. Ultimately, AIE persistently impacts the neurocircuitry of reward-motivated behavior in female rats.
Collapse
Affiliation(s)
- Jose A Pochapski
- Laboratorio de Fisiologia e Farmacologia do Sistema Nervoso Central, Department of Pharmacology, Universidade Federal do Parana, Curitiba, PR, Brazil
| | - Alexander Gómez-A
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Hannah Jaggers
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charlotte A Boettiger
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Claudio Da Cunha
- Laboratorio de Fisiologia e Farmacologia do Sistema Nervoso Central, Department of Pharmacology, Universidade Federal do Parana, Curitiba, PR, Brazil
- Department of Biochemistry, Universidade Federal do Parana, Curitiba, PR, Brazil
| | - Donita L Robinson
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Moreno-Fernández RD, Bernabéu-Brotons E, Carbonell-Colomer M, Buades-Sitjar F, Sampedro-Piquero P. Sex-related differences in young binge drinkers on the neurophysiological response to stress in virtual reality. Front Public Health 2024; 12:1348960. [PMID: 38947350 PMCID: PMC11211283 DOI: 10.3389/fpubh.2024.1348960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 06/06/2024] [Indexed: 07/02/2024] Open
Abstract
Background Stress is one of the main environmental factors involved in the onset of different psychopathologies. In youth, stressful life events can trigger inappropriate and health-damaging behaviors, such as binge drinking. This behavior, in turn, can lead to long-lasting changes in the neurophysiological response to stress and the development of psychological disorders late in life, e.g., alcohol use disorder. Our aim was to analyze the pattern of neurophysiological responses triggered with the exposition to a stressful virtual environment in young binge drinkers. Methods AUDIT-3 (third question from the full AUDIT) was used to detect binge drinking (BD) in our young sample (age 18-25 years). According to the score, participants were divided into control (CO) and BD group. Next, a standardized virtual reality (VR) scenario (Richie's Plank) was used for triggering the stress response while measuring the following neurophysiological variables: brain electrical activity by electroencephalogram (EEG) and cortisol levels through saliva samples both measurements registered before and after the stressful situation. Besides, heart rate (HR) with a pulsometer and electrodermal response (EDA) through electrodes placed on fingers were analyzed before, during and after the VR task. Results Regarding the behavior assessed during the VR task, BD group spent significantly less amount of time walking forward the table and a tendency toward more time walking backwards. There was no statistically significant difference between the BD and the CO group regarding time looking down, but when we controlled the variable sex, the BD women group displayed higher amount of time looking down than the rest of the groups. Neurophysiological measurements revealed that there was not any statistically significant difference between groups in any of the EEG registered measures, EDA response and cortisol levels. Sex-related differences were found in HR response to VR scenario, in which BD women displayed the highest peak of response to the stressor. Also, the change in heartbeat was higher in BD women than men. Conclusion Unveiling the neurophysiological alterations associated with BD can help us to prevent and detect early onset of alcohol use disorder. Also, from our data we conclude that participants' sex can modulate some stress responses, especially when unhealthy behaviors such as BD are present. Nevertheless, the moment of registration of the neurophysiological variables respect to the stressor seems to be a crucial variable.
Collapse
Affiliation(s)
| | | | | | - Francisco Buades-Sitjar
- Departamento de Psicología Biológica y de la Salud, Facultad de Psicología, Universidad Autónoma de Madrid, Madrid, Spain
| | - Patricia Sampedro-Piquero
- Departamento de Psicología Biológica y de la Salud, Facultad de Psicología, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
8
|
Grizzell JA, Vanbaelinghem M, Westerman J, Saddoris MP. Voluntary alcohol consumption during distinct phases of adolescence differentially alters adult fear acquisition, extinction and renewal in male and female rats. Stress 2023; 26:2278315. [PMID: 37916300 PMCID: PMC11042498 DOI: 10.1080/10253890.2023.2278315] [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: 07/17/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023] Open
Abstract
Alcohol use during adolescence coincides with elevated risks of stress-related impairment in adults, particularly via disrupted developmental trajectories of vulnerable corticolimbic and mesolimbic systems involved in fear processing. Prior work has investigated the impact of binge-like alcohol consumption on adult fear and stress, but less is known about whether voluntarily consumed alcohol imparts differential effects based on adolescence phases and biological sex. Here, adolescent male and female Long Evans rats were granted daily access to alcohol (15%) during either early (Early-EtOH; P25-45) or late adolescence (Late-EtOH; P45-55) using a modified drinking-in-the-dark design. Upon adulthood (P75-80), rats were exposed to a three-context (ABC) fear renewal procedure. We found that male and female Early-EtOH rats showed faster acquisition of fear but less freezing during early phases of extinction and throughout fear renewal. In the extinction period specifically, Early-EtOH rats showed normal levels of freezing in the presence of fear-associated cues, but abnormally low freezing immediately after cue offset, suggesting a key disruption in contextual processing and/or novelty seeking brought by early adolescent binge consumption. While the effects of alcohol were most pronounced in the Early-EtOH rats (particularly in females), Late-EtOH rats displayed some changes in fear behavior including slower fear acquisition, faster extinction, and reduced renewal compared with controls, but primarily in males. Our results suggest that early adolescence in males and females and, to a lesser extent, late adolescence in males is a particularly vulnerable period wherein alcohol use can promote stress-related dysfunction in adulthood. Furthermore, our results provide multiple bases for future research focused on developmental correlates of alcohol mediated disruption in the brain.
Collapse
Affiliation(s)
- J Alex Grizzell
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
- Dept of Neuroscience and Behavioral Biology, Emory University, Atlanta GA 30322
| | - Maryam Vanbaelinghem
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
| | - Jessica Westerman
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
| | - Michael P Saddoris
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
| |
Collapse
|
9
|
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
|
10
|
Alex Grizzell J, Vanbaelinghem M, Westerman J, Saddoris MP. Voluntary alcohol consumption during distinct phases of adolescence differentially alters adult fear acquisition, extinction and renewal in male and female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560757. [PMID: 37873067 PMCID: PMC10592894 DOI: 10.1101/2023.10.03.560757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Alcohol use during adolescence coincides with elevated risks of stress-related impairment in adults, particularly via disrupted developmental trajectories of vulnerable corticolimbic and mesolimbic systems involved in fear processing. Prior work has investigated the impact of binge-like alcohol consumption on adult fear and stress, but less is known about whether voluntarily consumed alcohol imparts differential effects based on adolescence phases and biological sex. Here, adolescent male and female Long Evans rats were granted daily access to alcohol (15%) during either early (Early-EtOH; P25-45) or late adolescence (Late-EtOH; P45-55) using a modified drinking-in-the-dark design. Upon adulthood (P75-80), rats were exposed to a three-context (ABC) fear renewal procedure. We found that male and female Early-EtOH rats showed faster acquisition of fear but less freezing during early phases of extinction and throughout fear renewal. In the extinction period specifically, Early-EtOH rats showed normal levels of freezing in the presence of fear-associated cues, but abnormally low freezing immediately after cue offset, suggesting a key disruption in contextual processing and/or novelty seeking brought by early adolescent binge consumption. While the effects of alcohol were most pronounced in the Early-EtOH rats (particularly in females), Late-EtOH rats displayed some changes in fear behavior including slower fear acquisition, faster extinction, and reduced renewal compared with controls, but primarily in males. Our results suggest that early adolescence in males and females and, to a lesser extent, late adolescence in males is a particularly vulnerable period wherein alcohol use can promote stress-related dysfunction in adulthood. Furthermore, our results provide multiple bases for future research focused on developmental correlates of alcohol mediated disruption in the brain.
Collapse
Affiliation(s)
- J Alex Grizzell
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
- Dept of Neuroscience and Behavioral Biology, Emory University, Atlanta GA 30322
| | - Maryam Vanbaelinghem
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
| | - Jessica Westerman
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
| | - Michael P Saddoris
- Dept Psychology & Neuroscience, University of Colorado Boulder, Boulder CO, 80301
| |
Collapse
|
11
|
Lee SH, Shnitko TA, Hsu LM, Broadwater MA, Sardinas M, Wang TWW, Robinson DL, Vetreno RP, Crews FT, Shih YYI. Acute alcohol induces greater dose-dependent increase in the lateral cortical network functional connectivity in adult than adolescent rats. ADDICTION NEUROSCIENCE 2023; 7:100105. [PMID: 37576436 PMCID: PMC10421607 DOI: 10.1016/j.addicn.2023.100105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Alcohol misuse and, particularly adolescent drinking, is a major public health concern. While evidence suggests that adolescent alcohol use affects frontal brain regions that are important for cognitive control over behavior little is known about how acute alcohol exposure alters large-scale brain networks and how sex and age may moderate such effects. Here, we employ a recently developed functional magnetic resonance imaging (fMRI) protocol to acquire rat brain functional connectivity data and use an established analytical pipeline to examine the effect of sex, age, and alcohol dose on connectivity within and between three major rodent brain networks: defaul mode, salience, and lateral cortical network. We identify the intra- and inter-network connectivity differences and establish moderation models to reveal significant influences of age on acute alcohol-induced lateral cortical network connectivity. Through this work, we make brain-wide isotropic fMRI data with acute alcohol challenge publicly available, with the hope to facilitate future discovery of brain regions/circuits that are causally relevant to the impact of acute alcohol use.
Collapse
Affiliation(s)
- Sung-Ho Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| | - Tatiana A. Shnitko
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Margaret A. Broadwater
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| | - Mabelle Sardinas
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Wen Winnie Wang
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Dai X, Yu J, Gao L, Zhang J, Li Y, Du B, Huang X, Zhang H. Cortical thickness and intrinsic activity changes in middle-aged men with alcohol use disorder. Alcohol 2023; 106:15-21. [PMID: 36272658 DOI: 10.1016/j.alcohol.2022.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Previous studies reported the alterations of brain structure or function in people with alcohol use disorder (AUD). However, a multi-modal approach combining structural and functional studies is essential to understanding the neural mechanisms of AUD. Hence, we examined regional differences in cortical thickness (CT) and amplitude of low-frequency fluctuation (ALFF) in patients with AUD. METHODS Thirty male patients with AUD and thirty age- and education-matched healthy male controls were recruited. High-resolution anatomical and resting-state functional MRI (rs-fMRI) data were collected, and the CT and ALFF were computed. RESULTS Behaviorally, males with AUD showed a cognitive decline in multiple domains. Structurally, they presented prominent reductions in CT in the bilateral temporal, insular, precentral, and dorsolateral prefrontal gyri (p < 0.05, voxel-wise family-wise error [FWE]). Functionally, a significant decrease in ALFF in the bilateral temporal, dorsolateral prefrontal, insular, putamen, cerebellum, right precuneus, mid-cingulate, and precentral gyri were observed (p < 0.05, FWE). CONCLUSIONS Our findings demonstrate the dual alterations of alcohol-related brain structure and function in male patients with AUD. These results may be useful in understanding the neural mechanisms in AUD.
Collapse
Affiliation(s)
- Xiyong Dai
- Department of Radiology, The Third People's Hospital of Zhongshan, Zhongshan City, Guangdong Province, China
| | - Jinming Yu
- Department of Psychiatry, The Third People's Hospital of Zhongshan, Zhongshan City, Guangdong Province, China
| | - Lei Gao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei Province, China
| | - Jianlong Zhang
- Department of Psychiatry, The Third People's Hospital of Zhongshan, Zhongshan City, Guangdong Province, China
| | - Yuanchun Li
- Department of Nursing, The Third People's Hospital of Zhongshan, Zhongshan City, Guangdong Province, China
| | - Baoguo Du
- Department of Psychiatry, The Third People's Hospital of Zhongshan, Zhongshan City, Guangdong Province, China
| | - Xiangyi Huang
- Department of Radiology, The Third People's Hospital of Zhongshan, Zhongshan City, Guangdong Province, China
| | - Haibo Zhang
- Department of Radiology, The Third People's Hospital of Zhongshan, Zhongshan City, Guangdong Province, China.
| |
Collapse
|
13
|
Obray JD, Landin JD, Vaughan DT, Scofield MD, Chandler LJ. Adolescent alcohol exposure reduces dopamine 1 receptor modulation of prelimbic neurons projecting to the nucleus accumbens and basolateral amygdala. ADDICTION NEUROSCIENCE 2022; 4:100044. [PMID: 36643604 PMCID: PMC9836047 DOI: 10.1016/j.addicn.2022.100044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Binge drinking during adolescence is highly prevalent despite increasing evidence of its long-term impact on behaviors associated with modulation of behavioral flexibility by the medial prefrontal cortex (mPFC). In the present study, male and female rats underwent adolescent intermittent ethanol (AIE) exposure by vapor inhalation. After aging to adulthood, retrograde bead labelling and viral tagging were used to identify populations of neurons in the prelimbic region (PrL) of the mPFC that project to specific subcortical targets. Electrophysiological recording from bead-labelled neurons in PrL slices revealed that AIE did not alter the intrinsic excitability of PrL neurons that projected to either the NAc or the BLA. Similarly, recordings of spontaneous inhibitory and excitatory post-synaptic currents revealed no AIE-induced changes in synaptic drive onto either population of projection neurons. In contrast, AIE exposure was associated with a loss of dopamine receptor 1 (D1), but no change in dopamine receptor 2 (D2), modulation of evoked firing of both populations of projection neurons. Lastly, confocal imaging of proximal and apical dendritic tufts of viral-labelled PrL neurons that projected to the nucleus accumbens (NAc) revealed AIE did not alter the density of dendritic spines. Together, these observations provide evidence that AIE exposure results in disruption of D1 receptor modulation of PrL inputs to at least two major subcortical target regions that have been implicated in AIE-induced long-term changes in behavioral control.
Collapse
Affiliation(s)
- J. Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Justine D. Landin
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Dylan T. Vaughan
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Michael D. Scofield
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA,Department of Anesthesiology, Medical University of South Carolina, Charleston SC, USA
| | - L. Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA,Corresponding author. (L.J. Chandler)
| |
Collapse
|
14
|
Walker CD, Sexton HG, Hyde J, Greene B, Risher ML. Diverging Effects of Adolescent Ethanol Exposure on Tripartite Synaptic Development across Prefrontal Cortex Subregions. Cells 2022; 11:3111. [PMID: 36231073 PMCID: PMC9561972 DOI: 10.3390/cells11193111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2022] Open
Abstract
Adolescence is a developmental period that encompasses, but is not limited to, puberty and continues into early adulthood. During this period, maturation and refinement are observed across brain regions such as the prefrontal cortex (PFC), which is critical for cognitive function. Adolescence is also a time when excessive alcohol consumption in the form of binge drinking peaks, increasing the risk of long-term cognitive deficits and the risk of developing an alcohol use disorder later in life. Animal models have revealed that adolescent ethanol (EtOH) exposure results in protracted disruption of neuronal function and performance on PFC-dependent tasks that require higher-order decision-making. However, the role of astrocytes in EtOH-induced disruption of prefrontal cortex-dependent function has yet to be elucidated. Astrocytes have complex morphologies with an extensive network of peripheral astrocyte processes (PAPs) that ensheathe pre- and postsynaptic terminals to form the 'tripartite synapse.' At the tripartite synapse, astrocytes play several critical roles, including synaptic maintenance, dendritic spine maturation, and neurotransmitter clearance through proximity-dependent interactions. Here, we investigate the effects of adolescent binge EtOH exposure on astrocyte morphology, PAP-synaptic proximity, synaptic stabilization proteins, and dendritic spine morphology in subregions of the PFC that are important in the emergence of higher cognitive function. We found that adolescent binge EtOH exposure resulted in subregion specific changes in astrocyte morphology and astrocyte-neuronal interactions. While this did not correspond to a loss of astrocytes, synapses, or dendritic spines, there was a corresponding region-specific and EtOH-dependent shift in dendritic spine phenotype. Lastly, we found that changes in astrocyte-neuronal interactions were not a consequence of changes in the expression of key synaptic structural proteins neurexin, neuroligin 1, or neuroligin 3. These data demonstrate that adolescent EtOH exposure results in enduring effects on neuron-glia interactions that persist into adulthood in a subregion-specific PFC manner, suggesting selective vulnerability. Further work is necessary to understand the functional and behavioral implications.
Collapse
Affiliation(s)
- Christopher Douglas Walker
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| | - Hannah Gray Sexton
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| | - Jentre Hyde
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Brittani Greene
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Mary-Louise Risher
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| |
Collapse
|
15
|
Quijano Cardé NA, Shaw J, Carter C, Kim S, Stitzel JA, Venkatesh SK, Ramchandani VA, De Biasi M. Mutation of the α5 nicotinic acetylcholine receptor subunit increases ethanol and nicotine consumption in adolescence and impacts adult drug consumption. Neuropharmacology 2022; 216:109170. [PMID: 35752273 PMCID: PMC9308728 DOI: 10.1016/j.neuropharm.2022.109170] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/16/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
Alcohol and nicotine are commonly used during adolescence, establishing long-lasting neuroplastic alterations that influence subsequent drug use and abuse. Drinking- and smoking-related traits have been extensively associated with variation in CHRNA5 - the gene that encodes the α5 subunit of neuronal nicotinic acetylcholine receptors (nAChRs). The single nucleotide polymorphism (SNP) rs16969968 in CHRNA5 encodes an amino acid substitution (D398N) that alters the function and pharmacokinetics of α5-containing nAChR. When expressed in rodents, this variant results in increased ethanol and nicotine operant self-administration. How disruption of α5-containing nAChRs influences adolescent ethanol and nicotine intake, and how it modulates interactions between these drugs has not been previously explored. In the present study, we examined volitional ethanol and nicotine consumption in adolescent mice (post-natal day 30-43) of both sexes with mutated (SNP) or lacking (KO) the α5 nAChR subunit. The effect of adolescent alcohol or nicotine exposure on home cage consumption of the opposite drug in adulthood and its modulation by Chrna5 mutation and sex were examined. During adolescence, we found that α5 nAChR disruption increases nicotine intake in mice of both sexes, but the effect on alcohol intake was only observed in females. The sex-specific increase in alcohol consumption in α5 SNP and KO was replicated in adulthood. The effect of adolescent alcohol or nicotine exposure on subsequent intake of the opposite drug in adulthood is modulated by sex and Chrna5 mutation. These observations suggest sex differences in the genetic architecture of alcohol dependence, and modulators of alcohol and nicotine interactions.
Collapse
Affiliation(s)
- Natalia A Quijano Cardé
- Pharmacology Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica Shaw
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina Carter
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Seung Kim
- Neuroscience Program, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Shyamala K Venkatesh
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Laboratory of Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Vijay A Ramchandani
- Laboratory of Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Mariella De Biasi
- Pharmacology Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Zhang WT, Chao THH, Yang Y, Wang TW, Lee SH, Oyarzabal EA, Zhou J, Nonneman R, Pegard NC, Zhu H, Cui G, Shih YYI. Spectral fiber photometry derives hemoglobin concentration changes for accurate measurement of fluorescent sensor activity. CELL REPORTS METHODS 2022; 2:100243. [PMID: 35880016 PMCID: PMC9308135 DOI: 10.1016/j.crmeth.2022.100243] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/08/2022] [Accepted: 06/08/2022] [Indexed: 12/22/2022]
Abstract
Fiber photometry is an emerging technique for recording fluorescent sensor activity in the brain. However, significant hemoglobin absorption artifacts in fiber photometry data may be misinterpreted as sensor activity changes. Because hemoglobin exists widely in the brain, and its concentration varies temporally, such artifacts could impede the accuracy of photometry recordings. Here we present use of spectral photometry and computational methods to quantify photon absorption effects by using activity-independent fluorescence signals, which can be used to derive oxy- and deoxy-hemoglobin concentration changes. Although these changes are often temporally delayed compared with the fast-responding fluorescence spikes, we found that erroneous interpretation may occur when examining pharmacology-induced sustained changes and that sometimes hemoglobin absorption could flip the GCaMP signal polarity. We provide hemoglobin-based correction methods to restore fluorescence signals and compare our results with other commonly used approaches. We also demonstrated the utility of spectral fiber photometry for delineating regional differences in hemodynamic response functions.
Collapse
Affiliation(s)
- Wei-Ting Zhang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yue Yang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tzu-Wen Wang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Esteban A. Oyarzabal
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jingheng Zhou
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Randy Nonneman
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nicolas C. Pegard
- Department of Applied Physical Sciences, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Guohong Cui
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
17
|
Degiorgis L, Arefin TM, Ben-Hamida S, Noblet V, Antal C, Bienert T, Reisert M, von Elverfeldt D, Kieffer BL, Harsan LA. Translational Structural and Functional Signatures of Chronic Alcohol Effects in Mice. Biol Psychiatry 2022; 91:1039-1050. [PMID: 35654559 DOI: 10.1016/j.biopsych.2022.02.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Alcohol acts as an addictive substance that may lead to alcohol use disorder. In humans, magnetic resonance imaging showed diverse structural and functional brain alterations associated with this complex pathology. Single magnetic resonance imaging modalities are used mostly but are insufficient to portray and understand the broad neuroadaptations to alcohol. Here, we combined structural and functional magnetic resonance imaging and connectome mapping in mice to establish brain-wide fingerprints of alcohol effects with translatable potential. METHODS Mice underwent a chronic intermittent alcohol drinking protocol for 6 weeks before being imaged under medetomidine anesthesia. We performed open-ended multivariate analysis of structural data and functional connectivity mapping on the same subjects. RESULTS Structural analysis showed alcohol effects for the prefrontal cortex/anterior insula, hippocampus, and somatosensory cortex. Integration with microglia histology revealed distinct alcohol signatures, suggestive of advanced (prefrontal cortex/anterior insula, somatosensory cortex) and early (hippocampus) inflammation. Functional analysis showed major alterations of insula, ventral tegmental area, and retrosplenial cortex connectivity, impacting communication patterns for salience (insula), reward (ventral tegmental area), and default mode (retrosplenial cortex) networks. The insula appeared as a most sensitive brain center across structural and functional analyses. CONCLUSIONS This study demonstrates alcohol effects in mice, which possibly underlie lower top-down control and impaired hedonic balance documented at the behavioral level, and aligns with neuroimaging findings in humans despite the potential limitation induced by medetomidine sedation. This study paves the way to identify further biomarkers and to probe neurobiological mechanisms of alcohol effects using genetic and pharmacological manipulations in mouse models of alcohol drinking and dependence.
Collapse
Affiliation(s)
- Laetitia Degiorgis
- Integrative Multimodal Imaging in Healthcare team, UMR 7357, Laboratory of Engineering, Informatics and Imaging (ICube); Department of Psychiatry, University of Strasbourg, Strasbourg, France
| | - Tanzil Mahmud Arefin
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany; Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York
| | - Sami Ben-Hamida
- INSERM U1114, University Hospital of Strasbourg, Strasbourg, France; INSERM U1247, research group on alcohol and pharmacodependance (GRAP), University of Picardie Jules-Verne, Amiens, France
| | - Vincent Noblet
- Images, Learning, Geometry and Statistics team, UMR 7357, Laboratory of Engineering, Informatics and Imaging (ICube); Department of Psychiatry, University of Strasbourg, Strasbourg, France
| | - Cristina Antal
- Integrative Multimodal Imaging in Healthcare team, UMR 7357, Laboratory of Engineering, Informatics and Imaging (ICube); Department of Psychiatry, University of Strasbourg, Strasbourg, France; Faculty of Medicine, Histology Institute and Unité Fonctionnelle de Foetopathologie, University Hospital of Strasbourg, Strasbourg, France
| | - Thomas Bienert
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Marco Reisert
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Dominik von Elverfeldt
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | | | - Laura-Adela Harsan
- Integrative Multimodal Imaging in Healthcare team, UMR 7357, Laboratory of Engineering, Informatics and Imaging (ICube); Department of Psychiatry, University of Strasbourg, Strasbourg, France; Department of Biophysics and Nuclear Medicine, University Hospital of Strasbourg, Strasbourg, France.
| |
Collapse
|
18
|
Sicher AR, Duerr A, Starnes WD, Crowley NA. Adolescent Alcohol and Stress Exposure Rewires Key Cortical Neurocircuitry. Front Neurosci 2022; 16:896880. [PMID: 35655755 PMCID: PMC9152326 DOI: 10.3389/fnins.2022.896880] [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: 03/15/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022] Open
Abstract
Human adolescence is a period of development characterized by wide ranging emotions and behavioral risk taking, including binge drinking (Konrad et al., 2013). These behavioral manifestations of adolescence are complemented by growth in the neuroarchitecture of the brain, including synaptic pruning (Spear, 2013) and increases in overall white matter volume (Perrin et al., 2008). During this period of profound physiological maturation, the adolescent brain has a unique vulnerability to negative perturbations. Alcohol consumption and stress exposure, both of which are heightened during adolescence, can individually and synergistically alter these neurodevelopmental trajectories in positive and negative ways (conferring both resiliency and susceptibility) and influence already changing neurotransmitter systems and circuits. Importantly, the literature is rapidly changing and evolving in our understanding of basal sex differences in the brain, as well as the interaction between biological sex and life experiences. The animal literature provides the distinctive opportunity to explore sex-specific stress- and alcohol- induced changes in neurocircuits on a relatively rapid time scale. In addition, animal models allow for the investigation of individual neurons and signaling molecules otherwise inaccessible in the human brain. Here, we review the human and rodent literature with a focus on cortical development, neurotransmitters, peptides, and steroids, to characterize the field's current understanding of the interaction between adolescence, biological sex, and exposure to stress and alcohol.
Collapse
Affiliation(s)
- Avery R. Sicher
- The Pennsylvania State University, University Park, PA, United States
- Department of Biology, The Pennsylvania State University, University Park, PA, United States
| | - Arielle Duerr
- Department of Biology, The Pennsylvania State University, University Park, PA, United States
| | - William D. Starnes
- Department of Biology, The Pennsylvania State University, University Park, PA, United States
| | - Nicole A. Crowley
- The Pennsylvania State University, University Park, PA, United States
- Department of Biology, The Pennsylvania State University, University Park, PA, United States
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
19
|
Dannenhoffer CA, Gómez-A A, Macht VA, Jawad R, Sutherland EB, Vetreno RP, Crews FT, Boettiger CA, Robinson DL. Impact of adolescent intermittent ethanol exposure on interneurons and their surrounding perineuronal nets in adulthood. Alcohol Clin Exp Res 2022; 46:759-769. [PMID: 35307830 PMCID: PMC9117471 DOI: 10.1111/acer.14810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/24/2022] [Accepted: 03/15/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Binge alcohol exposure during adolescence results in long-lasting alterations in the brain and behavior. For example, adolescent intermittent ethanol (AIE) exposure in rodents results in long-term loss of functional connectivity among prefrontal cortex (PFC) and striatal regions as well as a variety of neurochemical, molecular, and epigenetic alterations. Interneurons in the PFC and striatum play critical roles in behavioral flexibility and functional connectivity. For example, parvalbumin (PV) interneurons are known to contribute to neural synchrony and cholinergic interneurons contribute to strategy selection. Furthermore, extracellular perineuronal nets (PNNs) that surround some interneurons, particularly PV+ interneurons, further regulate cellular plasticity. The effect of AIE exposure on the expression of these markers within the PFC is not well understood. METHODS The present study tested the hypothesis that AIE exposure reduces the expression of PV+ and choline acetyltransferase (ChAT)+ interneurons in the adult PFC and striatum and increases the related expression of PNNs (marked by binding of Wisteria floribunda agglutinin lectin) in adulthood. Male rats were exposed to AIE (5 g/kg/day, 2-days-on/2-days-off, i.e., P25 to P54) or water (CON), and brain tissue was harvested in adulthood (>P80). Immunohistochemistry and co-immunofluorescence were used to assess the expression of ChAT, PV, and PNNs within the adult PFC and striatum following AIE exposure. RESULTS ChAT and PV interneuron densities in the striatum and PFC were unchanged after AIE exposure. However, PNN density in the PFC of AIE-exposed rats was greater than in CON rats. Moreover, significantly more PV neurons were surrounded by PNNs in AIE-exposed subjects than controls in both PFC subregions assessed: orbitofrontal cortex (CON = 34%; AIE = 40%) and medial PFC (CON = 10%; AIE = 14%). CONCLUSIONS These findings indicate that, following AIE exposure, PV interneuron expression in the adult PFC and striatum is unaltered, while PNNs surrounding these neurons are increased. This increase in PNNs may restrict the plasticity of the ensheathed neurons, thereby contributing to impaired microcircuitry in frontostriatal connectivity and related behavioral impairments.
Collapse
Affiliation(s)
- Carol A. Dannenhoffer
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
| | - Alexander Gómez-A
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
| | - Victoria A. Macht
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
| | - Rayyanoor Jawad
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
| | - E. Blake Sutherland
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill
| | - Charlotte A. Boettiger
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill
- Neuroscience Curriculum, University of North Carolina at Chapel Hill
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill
- Neuroscience Curriculum, University of North Carolina at Chapel Hill
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill
| |
Collapse
|
20
|
Altered dynamic interactions within frontostriatal circuits reflect disturbed craving processing in internet gaming disorder. CNS Spectr 2022; 27:109-117. [PMID: 32951628 DOI: 10.1017/s1092852920001832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Individuals with internet gaming disorder (IGD) are generally characterized by impaired executive control, persistent game-craving, and excessive reward-seeking behaviors. However, the causal interactions within the frontostriatal circuits underlying these problematic behaviors remain unclear. Here, spectral dynamic causal modeling (spDCM) was implemented to explore this issue. METHODS Resting-state functional magnetic resonance imaging data from 317 online game players (148 IGD subjects and 169 recreational game users (RGUs)) were collected. Using independent component analysis, we determined six region of interests within frontostriatal circuits for further spDCM analysis, and further statistical analyses based on the parametric empirical Bayes framework were performed. RESULTS Compared with RGUs, IGD subjects showed inhibitory effective connectivity from the right orbitofrontal cortex (OFC) to the right caudate and from the right dorsolateral prefrontal cortex to the left OFC; at the same time, excitatory effective connectivity was observed from the thalamus to the left OFC. Correlation analyses results showed that the directional connection from the right OFC to the right caudate was negatively associated with addiction severity. CONCLUSIONS These results suggest that the disrupted causal interactions between specific regions might contribute to dysfunctions within frontostriatal circuits in IGD, and the pathway from the right OFC to the right caudate could serve as a target for brain modulation in future IGD interventions.
Collapse
|
21
|
Gómez-A A, Dannenhoffer CA, Elton A, Lee SH, Ban W, Shih YYI, Boettiger CA, Robinson DL. Altered Cortico-Subcortical Network After Adolescent Alcohol Exposure Mediates Behavioral Deficits in Flexible Decision-Making. Front Pharmacol 2021; 12:778884. [PMID: 34912227 PMCID: PMC8666507 DOI: 10.3389/fphar.2021.778884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/03/2021] [Indexed: 12/24/2022] Open
Abstract
Behavioral flexibility, the ability to modify behavior according to changing conditions, is essential to optimize decision-making. Deficits in behavioral flexibility that persist into adulthood are one consequence of adolescent alcohol exposure, and another is decreased functional connectivity in brain structures involved in decision-making; however, a link between these two outcomes has not been established. We assessed effects of adolescent alcohol and sex on both Pavlovian and instrumental behaviors and resting-state functional connectivity MRI in adult animals to determine associations between behavioral flexibility and resting-state functional connectivity. Alcohol exposure impaired attentional set reversals and decreased functional connectivity among cortical and subcortical regions-of-interest that underlie flexible behavior. Moreover, mediation analyses indicated that adolescent alcohol-induced reductions in functional connectivity within a subnetwork of affected brain regions statistically mediated errors committed during reversal learning. These results provide a novel link between persistent reductions in brain functional connectivity and deficits in behavioral flexibility resulting from adolescent alcohol exposure.
Collapse
Affiliation(s)
- Alexander Gómez-A
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States
| | - Carol A. Dannenhoffer
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States
| | - Amanda Elton
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, United States
| | - Sung-Ho Lee
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, United States
- Department of Neurology, University of North Carolina, Chapel Hill, NC, United States
| | - Woomi Ban
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, United States
| | - Yen-Yu Ian Shih
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, United States
- Department of Neurology, University of North Carolina, Chapel Hill, NC, United States
- Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, United States
| | - Charlotte A. Boettiger
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, United States
- Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, United States
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States
- Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, United States
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
22
|
Lee SH, Broadwater MA, Ban W, Wang TWW, Kim HJ, Dumas JS, Vetreno RP, Herman MA, Morrow AL, Besheer J, Kash TL, Boettiger CA, Robinson DL, Crews FT, Shih YYI. An isotropic EPI database and analytical pipelines for rat brain resting-state fMRI. Neuroimage 2021; 243:118541. [PMID: 34478824 PMCID: PMC8561231 DOI: 10.1016/j.neuroimage.2021.118541] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/08/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Resting-state functional magnetic resonance imaging (fMRI) has drastically expanded the scope of brain research by advancing our knowledge about the topologies, dynamics, and interspecies translatability of functional brain networks. Several databases have been developed and shared in accordance with recent key initiatives in the rodent fMRI community to enhance the transparency, reproducibility, and interpretability of data acquired at various sites. Despite these pioneering efforts, one notable challenge preventing efficient standardization in the field is the customary choice of anisotropic echo planar imaging (EPI) schemes with limited spatial coverage. Imaging with anisotropic resolution and/or reduced brain coverage has significant shortcomings including reduced registration accuracy and increased deviation in brain feature detection. Here we proposed a high-spatial-resolution (0.4 mm), isotropic, whole-brain EPI protocol for the rat brain using a horizontal slicing scheme that can maintain a functionally relevant repetition time (TR), avoid high gradient duty cycles, and offer unequivocal whole-brain coverage. Using this protocol, we acquired resting-state EPI fMRI data from 87 healthy rats under the widely used dexmedetomidine sedation supplemented with low-dose isoflurane on a 9.4 T MRI system. We developed an EPI template that closely approximates the Paxinos and Watson's rat brain coordinate system and demonstrated its ability to improve the accuracy of group-level approaches and streamline fMRI data pre-processing. Using this database, we employed a multi-scale dictionary-learning approach to identify reliable spatiotemporal features representing rat brain intrinsic activity. Subsequently, we performed k-means clustering on those features to obtain spatially discrete, functional regions of interest (ROIs). Using Euclidean-based hierarchical clustering and modularity-based partitioning, we identified the topological organizations of the rat brain. Additionally, the identified group-level FC network appeared robust across strains and sexes. The "triple-network" commonly adapted in human fMRI were resembled in the rat brain. Through this work, we disseminate raw and pre-processed isotropic EPI data, a rat brain EPI template, as well as identified functional ROIs and networks in standardized rat brain coordinates. We also make our analytical pipelines and scripts publicly available, with the hope of facilitating rat brain resting-state fMRI study standardization.
Collapse
Affiliation(s)
- Sung-Ho Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA,Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA,Department of Neurology, University of North Carolina, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Corresponding authors at: Center for Animal MRI, 125 Mason Farm Road, CB# 7513, University of North Carolina, Chapel Hill, NC 27599, USA. (S.-H. Lee), (Y.-Y.I. Shih)
| | - Margaret A. Broadwater
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA,Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA,Department of Neurology, University of North Carolina, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| | - Woomi Ban
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA,Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Wen Winnie Wang
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA,Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Hyeon-Joong Kim
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA,Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA,Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Jaiden Seongmi Dumas
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA,Department of Neurology, University of North Carolina, Chapel Hill, NC, USA,Department of Quantitative Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Melissa A. Herman
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Charlotte A. Boettiger
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA,Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA,Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA,Department of Neurology, University of North Carolina, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA,Corresponding authors at: Center for Animal MRI, 125 Mason Farm Road, CB# 7513, University of North Carolina, Chapel Hill, NC 27599, USA. (S.-H. Lee), (Y.-Y.I. Shih)
| |
Collapse
|
23
|
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
|
24
|
Melbourne JK, Chandler CM, Van Doorn CE, Bardo MT, Pauly JR, Peng H, Nixon K. Primed for addiction: A critical review of the role of microglia in the neurodevelopmental consequences of adolescent alcohol drinking. Alcohol Clin Exp Res 2021; 45:1908-1926. [PMID: 34486128 PMCID: PMC8793635 DOI: 10.1111/acer.14694] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/22/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022]
Abstract
Alcohol is one of the most widely used recreational substances worldwide, with drinking frequently initiated during adolescence. The developmental state of the adolescent brain makes it vulnerable to initiating alcohol use, often in high doses, and particularly susceptible to alcohol-induced brain changes. Microglia, the brain parenchymal macrophages, have been implicated in mediating some of these effects, though the role that these cells play in the progression from alcohol drinking to dependence remains unclear. Microglia are uniquely positioned to sense and respond to central nervous system insult, and are now understood to exhibit innate immune memory, or "priming," altering their future functional responses based on prior exposures. In alcohol use disorders (AUDs), the role of microglia is debated. Whereas microglial activation can be pathogenic, contributing to neuroinflammation, tissue damage, and behavioral changes, or protective, it can also engage protective functions, providing support and mediating the resolution of damage. Understanding the role of microglia in adolescent AUDs is complicated by the fact that microglia are thought to be involved in developmental processes such as synaptic refinement and myelination, which underlie the functional maturation of multiple brain systems in adolescence. Thus, the role microglia play in the impact of alcohol use in adolescence is likely multifaceted. Long-term sequelae may be due to a failure to recover from EtOH-induced tissue damage, altered neurodevelopmental trajectories, and/or persistent changes to microglial responsivity and function. Here, we review critically the literature surrounding the effects of alcohol on microglia in models of adolescent alcohol misuse. We attempt to disentangle what is known about microglia from other neuroimmune effectors, to which we apply recent discoveries on the role of microglia in development and plasticity. Considered altogether, these studies challenge assumptions that proinflammatory microglia drive addiction. Alcohol priming microglia and thereby perturbing their homeostatic roles in neurodevelopment, especially during critical periods of plasticity such as adolescence, may have more serious implications for the neuropathogenesis of AUDs in adolescents.
Collapse
Affiliation(s)
- Jennifer K. Melbourne
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Cassie M. Chandler
- Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| | | | - Michael T. Bardo
- Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| | - James R. Pauly
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Hui Peng
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Kimberly Nixon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
25
|
Kipp BT, Nunes PT, Galaj E, Hitchcock B, Nasra T, Poynor KR, Heide SK, Reitz NL, Savage LM. Adolescent Ethanol Exposure Alters Cholinergic Function and Apical Dendritic Branching Within the Orbital Frontal Cortex. Neuroscience 2021; 473:52-65. [PMID: 34450212 DOI: 10.1016/j.neuroscience.2021.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
During adolescence, heavy binge-like ethanol consumption can lead to frontocortical structural and functional impairments. These impairments are likely driven by adolescence being a critical time point for maturation of brain regions associated with higher-order cognitive functioning. Rodent models of heavy binge-like ethanol exposure show consistent disruptions to the typical development of the prefrontal cortex (PFC). All deep cortical layers receive cholinergic projections that originate from the Nucleus basalis of Meynert (NbM) complex. These cholinergic projections are highly involved in learning, memory, and attention. Adolescent intermittent ethanol exposure (AIE) induces cholinergic dysfunction as a result of an epigenetic suppression of the genes that drive the cholinergic phenotype. The current study used a model of AIE to assess structural and functional changes to the frontal cortex and NbM following binge-like ethanol exposure in adolescence. Western blot analysis revealed long-term disruptions of the cholinergic circuit following AIE: choline acetyltransferase (ChAT) was suppressed in the NbM and vesicular acetylcholine transporter (VAChT) was suppressed in the orbitofrontal cortex (OFC). In vivo microdialysis for acetylcholine efflux during a spatial memory task determined changes in cholinergic modulation within the PFC following AIE. However, AIE spared performance on the spatial memory task and on an operant reversal task. In a second study, Golgi-Cox staining determined that AIE increased apical dendritic complexity in the OFC, with sex influencing whether the increase in branching occurred near or away from the soma. Spine density or maturity was not affected, likely compensating for a disruption in neurotransmitter function following AIE.
Collapse
Affiliation(s)
- B T Kipp
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - P T Nunes
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - E Galaj
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - B Hitchcock
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - T Nasra
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - K R Poynor
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - S K Heide
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - N L Reitz
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - L M Savage
- Department of Psychology, Binghamton University of the State University of New York, New York, USA.
| |
Collapse
|
26
|
Dannenhoffer CA, Robertson MM, Macht VA, Mooney SM, Boettiger CA, Robinson DL. Chronic alcohol exposure during critical developmental periods differentially impacts persistence of deficits in cognitive flexibility and related circuitry. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:117-173. [PMID: 34696872 DOI: 10.1016/bs.irn.2021.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cognitive flexibility in decision making depends on prefrontal cortical function and is used by individuals to adapt to environmental changes in circumstances. Cognitive flexibility can be measured in the laboratory using a variety of discrete, translational tasks, including those that involve reversal learning and/or set-shifting ability. Distinct components of flexible behavior rely upon overlapping brain circuits, including different prefrontal substructures that have separable impacts on decision making. Cognitive flexibility is impaired after chronic alcohol exposure, particularly during development when the brain undergoes rapid maturation. This review examines how cognitive flexibility, as indexed by reversal and set-shifting tasks, is impacted by chronic alcohol exposure in adulthood, adolescent, and prenatal periods in humans and animal models. We also discuss areas for future study, including mechanisms that may contribute to the persistence of cognitive deficits after developmental alcohol exposure and the compacting consequences from exposure across multiple critical periods.
Collapse
Affiliation(s)
- C A Dannenhoffer
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - M M Robertson
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, United States
| | - Victoria A Macht
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - S M Mooney
- Nutrition Research Institute and Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - C A Boettiger
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, United States; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, United States; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, United States
| | - Donita L Robinson
- 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, Chapel Hill, NC, United States; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
27
|
One-pot synthesis of carboxymethyl-dextran coated iron oxide nanoparticles (CION) for preclinical fMRI and MRA applications. Neuroimage 2021; 238:118213. [PMID: 34116153 PMCID: PMC8418149 DOI: 10.1016/j.neuroimage.2021.118213] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/15/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022] Open
Abstract
Superparamagnetic iron-oxide nanoparticles are robust contrast agents for magnetic resonance imaging (MRI) used for sensitive structural and functional mapping of the cerebral blood volume (CBV) when administered intravenously. To date, many CBV-MRI studies are conducted with Feraheme, manufactured for the clinical treatment of iron-deficiency. Unfortunately, Feraheme is currently not available outside the United States due to commercial and regulatory constraints, making CBV-MRI methods either inaccessible or very costly to achieve. To address this barrier, we developed a simple, one-pot recipe to synthesize Carboxymethyl-dextran coated Iron Oxide Nanoparticles, namely, “CION”, suitable for preclinical CBV-MRI applications. Here we disseminate a step-by-step instruction of our one-pot synthesis protocol, which allows CION to be produced in laboratories with minimal cost. We also characterized different CION-conjugations by manipulating polymer to metal stoichiometric ratio in terms of their size, surface chemistry, and chemical composition, and shifts in MR relaxivity and pharmacokinetics. We performed several proof-of-concept experiments in vivo, demonstrating the utility of CION for functional and structural MRI applications, including hypercapnic CO2 challenge, visual stimulation, targeted optogenetic stimulation, and microangiography. We also present evidence that CION can serve as a cross-modality research platform by showing concurrent in vivo optical and MRI measurement of CBV using fluorescent-labeled CION. The simplicity and cost-effectiveness of our one-pot synthesis method should allow researchers to reproduce CION and tailor the relaxivity and pharmacokinetics according to their imaging needs. It is our hope that this work makes CBV-MRI more openly available and affordable for a variety of research applications.
Collapse
|
28
|
Araujo I, Henriksen A, Gamsby J, Gulick D. Impact of Alcohol Abuse on Susceptibility to Rare Neurodegenerative Diseases. Front Mol Biosci 2021; 8:643273. [PMID: 34179073 PMCID: PMC8220155 DOI: 10.3389/fmolb.2021.643273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/14/2021] [Indexed: 12/22/2022] Open
Abstract
Despite the prevalence and well-recognized adverse effects of prenatal alcohol exposure and alcohol use disorder in the causation of numerous diseases, their potential roles in the etiology of neurodegenerative diseases remain poorly characterized. This is especially true of the rare neurodegenerative diseases, for which small population sizes make it difficult to conduct broad studies of specific etiological factors. Nonetheless, alcohol has potent and long-lasting effects on neurodegenerative substrates, at both the cellular and systems levels. This review highlights the general effects of alcohol in the brain that contribute to neurodegeneration across diseases, and then focuses on specific diseases in which alcohol exposure is likely to play a major role. These specific diseases include dementias (alcohol-induced, frontotemporal, and Korsakoff syndrome), ataxias (cerebellar and frontal), and Niemann-Pick disease (primarily a Type B variant and Type C). We conclude that there is ample evidence to support a role of alcohol abuse in the etiology of these diseases, but more work is needed to identify the primary mechanisms of alcohol's effects.
Collapse
Affiliation(s)
- Iskra Araujo
- Gulick Laboratory, Byrd Neuroscience Institute, University of South Florida Health, Tampa, FL, United States
| | - Amy Henriksen
- Gulick Laboratory, Byrd Neuroscience Institute, University of South Florida Health, Tampa, FL, United States
| | - Joshua Gamsby
- Gulick Laboratory, Byrd Neuroscience Institute, University of South Florida Health, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South FL, Tampa, FL, United States
| | - Danielle Gulick
- Gulick Laboratory, Byrd Neuroscience Institute, University of South Florida Health, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South FL, Tampa, FL, United States
| |
Collapse
|
29
|
Liu W, Rohlman AR, Vetreno R, Crews FT. Expression of Oligodendrocyte and Oligoprogenitor Cell Proteins in Frontal Cortical White and Gray Matter: Impact of Adolescent Development and Ethanol Exposure. Front Pharmacol 2021; 12:651418. [PMID: 34025418 PMCID: PMC8134748 DOI: 10.3389/fphar.2021.651418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Adolescent development of prefrontal cortex (PFC) parallels maturation of executive functions as well as increasing white matter and myelination. Studies using MRI and other methods find that PFC white matter increases across adolescence into adulthood in both humans and rodents. Adolescent binge drinking is common and has been found to alter adult behaviors and PFC functions. This study examines development of oligoprogenitor (OPC) and oligodendrocytes (OLs) in Wistar rats from adolescence to adulthood within PFC white matter, corpus callosum forceps minor (fmi), PFC gray matter, and the neurogenic subventricular zone (SVZ) using immunohistochemistry for marker proteins. In addition, the effects of adolescent intermittent ethanol exposure [AIE; 5.0 g/kg/day, intragastric, 2 days on/2 days off on postnatal day (P)25-54], which is a weekend binge drinking model, were determined. OPC markers NG2+, PDGFRα+ and Olig2+IHC were differentially impacted by both age and PFC region. In both fmi and SVZ, NG2+IHC cells declined from adolescence to adulthood with AIE increasing adult NG2+IHC cells and their association with microglial marker Iba1. PFC gray matter decline in NG2+IHC in adulthood was not altered by AIE. Both adult maturation and AIE impacted OL expression of PLP+, MBP+, MAG+, MOG+, CNPase+, Olig1+, and Olig2+IHC in all three PFC regions, but in region- and marker-specific patterns. These findings are consistent with PFC region-specific changes in OPC and OL markers from adolescence to adulthood as well as following AIE that could contribute to lasting changes in PFC function.
Collapse
Affiliation(s)
| | | | | | - Fulton T. Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
30
|
Towner TT, Spear LP. Rats exposed to intermittent ethanol during late adolescence exhibit enhanced habitual behavior following reward devaluation. Alcohol 2021; 91:11-20. [PMID: 33031883 DOI: 10.1016/j.alcohol.2020.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 12/29/2022]
Abstract
The brain undergoes substantial maturation during adolescence, and repeated exposure to ethanol at this time has been shown to result in long-lasting behavioral and neural consequences. During the broad period of adolescence, different neuronal populations and circuits are refined between early and late adolescence, suggesting the possibility that ethanol exposure at these differing times may lead to differential outcomes. The goal of the current study was to evaluate the impact of adolescent intermittent ethanol (AIE) during early and late adolescence on the formation of goal-directed and habitual behavior in adulthood. Male and female Sprague-Dawley rats were exposed to ethanol via intragastric gavage (4.0 g/kg, 25% v/v) every other day from postnatal day (P) 25-45 or P45-65, considered early and late adolescence, respectively. In adulthood (~P70 early or ~ P90 late), rats were gradually food-restricted and began operant training on a fixed ratio 1 schedule. Rats were then transitioned onto random interval schedules and eventually underwent a sensory-specific satiation procedure as a model of reward devaluation. Few differences as a result of adolescent ethanol exposure were found during instrumental training. Following reward devaluation, rats exposed to water and ethanol during early adolescence exhibited reductions in lever pressing, suggestive of a goal-directed response pattern. In contrast, late AIE males and females demonstrated persistent responding following both devalued and non-devalued trials, findings representative of a habitual behavior pattern. The shifts from goal-directed to habitual behavior noted only following late AIE contribute to the growing literature identifying specific behavioral consequences as a result of ethanol exposure during distinct developmental periods within adolescence. More work is needed to determine whether the greater habit formation following late AIE is also associated with elevated habitual ethanol consumption.
Collapse
Affiliation(s)
- Trevor Theodore Towner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, United States.
| | - Linda Patia Spear
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, United States
| |
Collapse
|
31
|
Chen J, Li X, Zhang Q, Zhou Y, Wang R, Tian C, Xiang H. Impulsivity and Response Inhibition Related Brain Networks in Adolescents With Internet Gaming Disorder: A Preliminary Study Utilizing Resting-State fMRI. Front Psychiatry 2021; 11:618319. [PMID: 33519558 PMCID: PMC7843793 DOI: 10.3389/fpsyt.2020.618319] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/16/2020] [Indexed: 01/31/2023] Open
Abstract
Background and Aims: Internet gaming disorder (IGD), as a relapse disease, has become a common mental health problem among Asian teenagers. Functional connections in the prefrontal lobo-striatum affect changes in impulsivity and inhibition. Therefore, exploration of the directional connections of the relevant brain regions in the prefrontal-striatal circuit and the synchronization level of the two hemispheres will help us to further understand the neural mechanism of IGD, which can provide guidance for the development of prevention and intervention strategies. Methods: Twenty-two adolescents with IGD, recruited through various channels, composed the IGD group. Twenty-six subjects, matching age, gender, and education level, were included in a recreational internet game users (RGUs) control group. Impulsivity and response inhibition were tested via general questionnaire, the Internet Addiction Test (IAT), the Barratt impulsivity scale-11 (BIS-11), and a Stroop color-word task. A Granger causality analysis (GCA) was used to calculate the directional connection between the prefrontal and striatum with the dorsolateral prefrontal cortex (DLPFC) as a region of interest (ROI). We chose voxel-mirrored homotopic connectivity (VMHC) to determine brain hemisphere functional connectivity in the prefrontal-striatal circuits. Results: We found significant differences in impulsivity between the IGD group and RGU group, with members of the IGD group exhibiting higher impulsivity. Additionally, the response inhibition of adolescents with IGD in the Stroop color-word task was impaired. There was a significant difference in the directed connection of the left DLPFC and dorsal striatum between the IGD group and the RGU group. Conclusions: This study confirmed the role of prefrontal-striatal circuits in the neural mechanism of IGD in adolescents. In the IGD group, bilateral cerebral medial orbitofrontal cortex (mOFC) synchronization was significantly reduced, which indicated that mOFC signal transmission in both hemispheres of the brain might be affected by impulse behavior and impaired response inhibition.
Collapse
Affiliation(s)
- Jieyu Chen
- Department of Medical Psychology, College of Medical Humanities, Guizhou Medical University, Guiyang, China
| | - Xinyi Li
- Department of Medical Psychology, College of Medical Humanities, Guizhou Medical University, Guiyang, China
- Binzhou Medical University, Binzhou, China
| | - Qun Zhang
- Department of Psychology, Guizhou Normal University, Guiyang, China
| | - Yu Zhou
- Department of Psychiatry, Guizhou Provincial People's Hospital, Guiyang, China
| | - Rongpin Wang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Chong Tian
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hui Xiang
- Department of Psychiatry, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
32
|
Waddell J, Hill E, Tang S, Jiang L, Xu S, Mooney SM. Choline Plus Working Memory Training Improves Prenatal Alcohol-Induced Deficits in Cognitive Flexibility and Functional Connectivity in Adulthood in Rats. Nutrients 2020; 12:E3513. [PMID: 33202683 PMCID: PMC7696837 DOI: 10.3390/nu12113513] [Citation(s) in RCA: 10] [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: 09/30/2020] [Revised: 10/27/2020] [Accepted: 11/12/2020] [Indexed: 12/28/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is the leading known cause of intellectual disability, and may manifest as deficits in cognitive function, including working memory. Working memory capacity and accuracy increases during adolescence when neurons in the prefrontal cortex undergo refinement. Rats exposed to low doses of ethanol prenatally show deficits in working memory during adolescence, and in cognitive flexibility in young adulthood. The cholinergic system plays a crucial role in learning and memory processes. Here we report that the combination of choline and training on a working memory task during adolescence significantly improved cognitive flexibility (performance on an attentional set shifting task) in young adulthood: 92% of all females and 81% of control males formed an attentional set, but only 36% of ethanol-exposed males did. Resting state functional magnetic resonance imaging showed that functional connectivity among brain regions was different between the sexes, and was altered by prenatal ethanol exposure and by choline + training. Connectivity, particularly between prefrontal cortex and striatum, was also different in males that formed a set compared with those that did not. Together, these findings indicate that prenatal exposure to low doses of ethanol has persistent effects on brain functional connectivity and behavior, that these effects are sex-dependent, and that an adolescent intervention could mitigate some of the effects of prenatal ethanol exposure.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (E.H.); (S.M.M.)
| | - Elizabeth Hill
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (E.H.); (S.M.M.)
| | - Shiyu Tang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.T.); (L.J.); (S.X.)
| | - Li Jiang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.T.); (L.J.); (S.X.)
| | - Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.T.); (L.J.); (S.X.)
| | - Sandra M. Mooney
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (E.H.); (S.M.M.)
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
33
|
Macht V, Elchert N, Crews F. Adolescent Alcohol Exposure Produces Protracted Cognitive-Behavioral Impairments in Adult Male and Female Rats. Brain Sci 2020; 10:brainsci10110785. [PMID: 33126417 PMCID: PMC7692738 DOI: 10.3390/brainsci10110785] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/20/2022] Open
Abstract
Binge drinking is common in adolescence. Rodent studies modeling adolescent binge drinking find persistent effects on the brain's physiology, including increased expression of neuroimmune genes, impaired neurogenesis, and changes in behavioral flexibility. This study used females and males to investigate the effects of adolescent intermittent ethanol (AIE) on a battery of behaviors assessing spatial navigation using a radial arm water maze, working memory using the Hebb-Williams maze, non-spatial long-term memory using novel object recognition, and dominance using a tube dominance test. Results indicate that AIE impairs adult acquisition in spatial navigational learning with deficits predominantly driven by females. Surprisingly, AIE slowed the transition from random to serial search strategies in both sexes, suggesting AIE impairs flexibility in problem-solving processing. In the Hebb-Williams maze working memory task, adult AIE rats exhibited deficits in problem solving, resulting in more errors across the 12 maze configurations, independent of sex. Conversely, AIE decreased dominance behaviors in female rats, and at 7 months post-alcohol, female AIE rats continued to exhibit deficits in novel object recognition. These results suggest that cognitive-behavioral alterations after adolescent binge drinking persist well into middle age, despite abstinence. Future studies should focus on intervening treatment strategies in both females and males.
Collapse
Affiliation(s)
- Victoria Macht
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, USA; (V.M.); (N.E.)
| | - Natalie Elchert
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, USA; (V.M.); (N.E.)
| | - Fulton Crews
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, USA; (V.M.); (N.E.)
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +1-919-966-5678
| |
Collapse
|
34
|
Hsu LM, Wang S, Ranadive P, Ban W, Chao THH, Song S, Cerri DH, Walton LR, Broadwater MA, Lee SH, Shen D, Shih YYI. Automatic Skull Stripping of Rat and Mouse Brain MRI Data Using U-Net. Front Neurosci 2020; 14:568614. [PMID: 33117118 PMCID: PMC7575753 DOI: 10.3389/fnins.2020.568614] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/13/2020] [Indexed: 11/13/2022] Open
Abstract
Accurate removal of magnetic resonance imaging (MRI) signal outside the brain, a.k.a., skull stripping, is a key step in the brain image pre-processing pipelines. In rodents, this is mostly achieved by manually editing a brain mask, which is time-consuming and operator dependent. Automating this step is particularly challenging in rodents as compared to humans, because of differences in brain/scalp tissue geometry, image resolution with respect to brain-scalp distance, and tissue contrast around the skull. In this study, we proposed a deep-learning-based framework, U-Net, to automatically identify the rodent brain boundaries in MR images. The U-Net method is robust against inter-subject variability and eliminates operator dependence. To benchmark the efficiency of this method, we trained and validated our model using both in-house collected and publicly available datasets. In comparison to current state-of-the-art methods, our approach achieved superior averaged Dice similarity coefficient to ground truth T2-weighted rapid acquisition with relaxation enhancement and T2∗-weighted echo planar imaging data in both rats and mice (all p < 0.05), demonstrating robust performance of our approach across various MRI protocols.
Collapse
Affiliation(s)
- Li-Ming Hsu
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Shuai Wang
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Paridhi Ranadive
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Woomi Ban
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tzu-Hao Harry Chao
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sheng Song
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Domenic Hayden Cerri
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lindsay R. Walton
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Margaret A. Broadwater
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sung-Ho Lee
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dinggang Shen
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Brain and Cognitive Engineering, Korea University, Seoul, South Korea
| | - Yen-Yu Ian Shih
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
35
|
Ehlers CL, Wills DN, Karriker-Jaffe KJ, Gilder DA, Phillips E, Bernert RA. Delta Event-Related Oscillations Are Related to a History of Extreme Binge Drinking in Adolescence and Lifetime Suicide Risk. Behav Sci (Basel) 2020; 10:E154. [PMID: 33036364 PMCID: PMC7599813 DOI: 10.3390/bs10100154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 11/18/2022] Open
Abstract
Alcohol exposure typically begins in adolescence, and heavy binge drinking is associated with health risk behaviors. Event-related oscillations (EROs) may represent sensitive biomarkers or endophenotypes for early alcohol exposure as well as other risk behaviors such as suicidal thoughts and actions. In this study, young adults (age 18-30 years) of American Indian (AI) (n = 479) and Mexican American (MA) (n = 705) ancestry were clinically assessed, and EROs were generated to happy, sad and neutral faces. Extreme adolescent binge drinking (10+ drinks) was common (20%) in this population of AI/MA and associated with a significantly increased risk of a lifetime history of suicidal acts (SA, suicide attempts, deaths) but not suicidal thoughts (ST, ideation, plans). ST were reported among MA participants, whereas SA were more common among AI young adults. Extreme adolescent binge drinking was also associated with errors in detection of sad and neutral faces, increases in delta ERO energy, and decreases in phase locking (PL), particularly in parietal areas. A lifetime history of ST was associated with increases in delta ERO energy and PL, whereas SA were associated with decreases in both. These studies suggest that ERO measures may represent important potential biomarkers of adolescent extreme binge drinking and risk for suicidal behaviors.
Collapse
Affiliation(s)
- Cindy L. Ehlers
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; (D.N.W.); (D.A.G.); (E.P.)
| | - Derek N. Wills
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; (D.N.W.); (D.A.G.); (E.P.)
| | | | - David A. Gilder
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; (D.N.W.); (D.A.G.); (E.P.)
| | - Evelyn Phillips
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; (D.N.W.); (D.A.G.); (E.P.)
| | - Rebecca A. Bernert
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
36
|
Shields CN, Gremel CM. Review of Orbitofrontal Cortex in Alcohol Dependence: A Disrupted Cognitive Map? Alcohol Clin Exp Res 2020; 44:1952-1964. [PMID: 32852095 PMCID: PMC8261866 DOI: 10.1111/acer.14441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022]
Abstract
Alcoholism is a persistent worldwide problem associated with long-lasting impairments to decision making processes. Some aspects of dysfunction are thought to reflect alcohol-induced changes to relevant brain areas such as the orbitofrontal cortex (OFC). In this review, we will examine how chronic alcohol exposure alters OFC function to potentially contribute to maladaptive decision making, and explore experimental behavioral approaches that may be better suited to test whether alcohol dependence disrupts OFC's function. We argue that although past works suggest impairments in aspects of OFC function, more information may be gained by specifically targeting tasks to the broader function of OFC as put forth by the recent hypothesis of OFC as a "cognitive map" of task space. Overall, we suggest that such a focus could provide a better understanding of how OFC function changes in alcohol dependence, and could inform better assessment tools and treatment options for clinicians working with this population.
Collapse
Affiliation(s)
- Chloe N. Shields
- Department of Psychology, University of California San Diego, La Jolla, CA 92093, USA
| | - Christina M. Gremel
- Department of Psychology, University of California San Diego, La Jolla, CA 92093, USA
- The Neurosciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
37
|
Galaj E, Barrera E, Morris D, Ma YY, Ranaldi R. Aberrations in Incentive Learning and Responding to Heroin in Male Rats After Adolescent or Adult Chronic Binge-Like Alcohol Exposure. Alcohol Clin Exp Res 2020; 44:1214-1223. [PMID: 32311102 PMCID: PMC7313436 DOI: 10.1111/acer.14341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/08/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE Binge drinking is a serious problem among adolescents and young adults despite its adverse consequences on the brain and behavior. One area that remains poorly understood concerns the impact of chronic intermittent ethanol (CIE) exposure on incentive learning. METHODS Here, we examined the effects of CIE exposure during different developmental stages on conditioned approach and conditioned reward learning in rats experiencing acute or protracted withdrawal from alcohol. Two or 21 days after adolescent or adult CIE exposure, male rats were exposed to pairings of a light stimulus (CS) and food pellets for 3 consecutive daily sessions (30 CS-food pellet pairings per session). This was followed by conditioned approach testing measuring responses (food trough head entries) to the CS-only presentations and by conditioned reward testing measuring responses on a lever producing the CS and on another producing a tone. We then measured behavioral sensitization to repeated injections of heroin (2 mg/kg/d for 9 days). RESULTS Adolescent and adult alcohol-treated rats showed significantly impaired conditioned reward learning regardless of withdrawal period (acute or prolonged). We found no evidence of changes to conditioned approach learning after adolescent or adult exposure to CIE. Finally, in addition to producing long-term impairments in incentive learning, CIE exposure enhanced locomotor activity in response to heroin and had no effect on behavioral sensitization to heroin regardless of age and withdrawal period. CONCLUSIONS Our work sets a framework for identifying CIE-induced alterations in incentive learning and inducing susceptibility to subsequent opioid effects.
Collapse
Affiliation(s)
- Ewa Galaj
- National Institute on Drug Abuse Intramural Research Program, Molecular Targets and Medication Discovery Branch, 251 Bayview Blvd, Baltimore, MD, 21224, US
| | - Eddy Barrera
- Queens College, City University of New York, Department of Psychology, 65-30 Kissena Blvd., Flushing, NY, 11367, US
| | - Debra Morris
- Queens College, City University of New York, Department of Psychology, 65-30 Kissena Blvd., Flushing, NY, 11367, US
| | - Yao-Ying Ma
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, 635 Barnhill Drive, Indianapolis, IN, 46202, US
| | - Robert Ranaldi
- Queens College, City University of New York, Department of Psychology, 65-30 Kissena Blvd., Flushing, NY, 11367, US
| |
Collapse
|
38
|
Scuppa G, Tambalo S, Pfarr S, Sommer WH, Bifone A. Aberrant insular cortex connectivity in abstinent alcohol-dependent rats is reversed by dopamine D3 receptor blockade. Addict Biol 2020; 25:e12744. [PMID: 30907042 PMCID: PMC7187338 DOI: 10.1111/adb.12744] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/12/2019] [Accepted: 02/12/2019] [Indexed: 12/23/2022]
Abstract
A few studies have reported aberrant functional connectivity in alcoholic patients, but the specific neural circuits involved remain unknown. Moreover, it is unclear whether these alterations can be reversed upon treatment. Here, we used functional MRI to study resting state connectivity in rats following chronic intermittent exposure to ethanol. Further, we evaluated the effects of SB-277011-a, a selective dopamine D3 receptor antagonist, known to decrease ethanol consumption. Alcohol-dependent and control rats (N = 13/14 per group), 3 weeks into abstinence, were administered SB-277011-a or vehicle before fMRI sessions. Resting state connectivity networks were extracted by independent component analysis. A dual-regression analysis was performed using independent component maps as spatial regressors, and the effects of alcohol history and treatment on connectivity were assessed. A history of alcohol dependence caused widespread reduction of the internal coherence of components. Weaker correlation was also found between the insula cortex (IC) and cingulate cortices, key constituents of the salience network. Similarly, reduced connectivity was observed between a component comprising the anterior insular cortex, together with the caudate putamen (CPu-AntIns), and the posterior part of the IC. On the other hand, postdependent rats showed strengthened connectivity between salience and reward networks. In particular, higher connectivity was observed between insula and nucleus accumbens, between the ventral tegmental area and the cingulate cortex and between the VTA and CPu-AntIns. Interestingly, aberrant connectivity in postdependent rats was partially restored by acute administration of SB-277011-a, which, conversely, had no significant effects in naïve rats.
Collapse
Affiliation(s)
- Giulia Scuppa
- Center for Neuroscience and Cognitive SystemsIstituto Italiano di TecnologiaRoveretoItaly
| | - Stefano Tambalo
- Center for Neuroscience and Cognitive SystemsIstituto Italiano di TecnologiaRoveretoItaly
| | - Simone Pfarr
- Institute of Psychopharmacology, Central Institute of Mental HealthUniversity of HeidelbergMannheimGermany
| | - Wolfgang H. Sommer
- Institute of Psychopharmacology, Central Institute of Mental HealthUniversity of HeidelbergMannheimGermany
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental HealthUniversity of HeidelbergMannheimGermany
| | - Angelo Bifone
- Center for Neuroscience and Cognitive SystemsIstituto Italiano di TecnologiaRoveretoItaly
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
| |
Collapse
|
39
|
Lees B, Meredith LR, Kirkland AE, Bryant BE, Squeglia LM. Effect of alcohol use on the adolescent brain and behavior. Pharmacol Biochem Behav 2020; 192:172906. [PMID: 32179028 PMCID: PMC7183385 DOI: 10.1016/j.pbb.2020.172906] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/31/2020] [Accepted: 03/12/2020] [Indexed: 12/18/2022]
Abstract
Adolescence is a particularly vulnerable neurodevelopmental period marked by high rates of engagement with risky alcohol use. This review summarizes the cognitive and neural consequences following alcohol use during adolescence from longitudinal design studies in humans and animals. Findings from human adolescent studies suggest that binge drinking and heavy alcohol use is associated with poorer cognitive functioning on a broad range of neuropsychological assessments, including learning, memory, visuospatial functioning, psychomotor speed, attention, executive functioning, and impulsivity. Alcohol use during adolescence is associated with accelerated decreases in gray matter and attenuated increases in white matter volume, and aberrant neural activity during executive functioning, attentional control, and reward sensitivity tasks, when compared to non-drinking adolescents. Animal studies in rodents and non-human primates have replicated human findings, and suggest cognitive and neural consequences of adolescent alcohol use may persist into adulthood. Novel rodent studies demonstrate that adolescent alcohol use may increase reward responsiveness of the dopamine system to alcohol later in life, as well as disrupt adolescent neurogenesis, potentially through neuroinflammation, with long-lasting neural and behavioral effects into adulthood. Larger longitudinal human cognitive and neuroimaging studies with more diverse samples are currently underway which will improve understanding of the impact of polysubstance use, as well as the interactive effects of substance use, physical and mental health, and demographic factors on cognition and neurodevelopment.
Collapse
Affiliation(s)
- Briana Lees
- The Matilda Centre for Research in Mental Health and Substance Use, University of Sydney, Australia.
| | - Lindsay R Meredith
- University of California, Los Angeles, Department of Psychology, United States of America
| | - Anna E Kirkland
- American University, Department of Psychology, United States of America
| | - Brittany E Bryant
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, United States of America
| | - Lindsay M Squeglia
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, United States of America
| |
Collapse
|
40
|
Ehlers CL, Phillips E, Wills D, Benedict J, Sanchez-Alavez M. Phase locking of event-related oscillations is decreased in both young adult humans and rats with a history of adolescent alcohol exposure. Addict Biol 2020; 25:e12732. [PMID: 30884076 PMCID: PMC6751029 DOI: 10.1111/adb.12732] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 01/25/2023]
Abstract
Alcohol exposure typically begins in adolescence, and frequent binge drinking has been associated with health risk behaviors including alcohol use disorders (AUDs). Few studies have documented the effects of a history of adolescent binge drinking on neurophysiological consequences in young adulthood. Synchrony of phase (phase locking (PL)) of event-related oscillations (EROs) within and between different brain areas reflects communication exchange between neural networks and is a sensitive measure of adolescent development in both rats and humans, and thus may be a good translational measure of the potential harmful effects of alcohol exposure during adolescence. In this study, EROs were collected from 1041 young adults of Mexican American and American Indian ancestry (age 18-30 years) with and without a history of adolescent binge drinking (five drinks for boys and four for girls per occasion at least once per month) and in 74 young adult rats with and without a history of 5 weeks of adolescent alcohol vapor exposure. PL of theta and beta frequencies between frontal and parietal cortex were estimated using an auditory-oddball paradigm in the rats and a visual facial expression paradigm in the humans. Significantly lower PL between frontal and parietal cortices in the theta frequencies was seen in both the humans and the rats with a history of adolescent alcohol exposure as compared with their controls. These findings suggest that alcohol exposure during adolescence may result in decreases in synchrony between cortical neuronal networks, suggesting a developmental delay, in young adult humans and in rats.
Collapse
Affiliation(s)
- Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Evie Phillips
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Derek Wills
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Jessica Benedict
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
41
|
Age-dependent impairment of metabotropic glutamate receptor 2-dependent long-term depression in the mouse striatum by chronic ethanol exposure. Alcohol 2020; 82:11-21. [PMID: 31233806 PMCID: PMC6925350 DOI: 10.1016/j.alcohol.2019.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022]
Abstract
Chronic alcohol exposure is associated with increased reliance on behavioral strategies involving the dorsolateral striatum (DLS), including habitual or stimulus-response behaviors. Presynaptic G protein-coupled receptors (GPCRs) on cortical and thalamic inputs to the DLS inhibit glutamate release, and alcohol-induced disruption of presynaptic GPCR function represents a mechanism by which alcohol could disinhibit DLS neurons and thus bias toward use of DLS-dependent behaviors. Metabotropic glutamate receptor 2 (mGlu2) is a Gi/o-coupled GPCR that robustly modulates glutamate transmission in the DLS, inducing long-term depression (LTD) at both cortical and thalamic synapses. Loss of mGlu2 function has recently been associated with increased ethanol seeking and consumption, but the ability of alcohol to produce adaptations in mGlu2 function in the DLS has not been investigated. We exposed male C57Bl/6J mice to a 2-week chronic intermittent ethanol (CIE) paradigm followed by a brief withdrawal period, then used whole-cell patch clamp recordings of glutamatergic transmission in the striatum to assess CIE effects on mGlu2-mediated synaptic plasticity. We report that CIE differentially disrupts mGlu2-mediated long-term depression in the DLS vs. dorsomedial striatum (DMS). Interestingly, CIE-induced impairment of mGlu2-LTD in the dorsolateral striatum is only observed when alcohol exposure occurs during adolescence. Incubation of striatal slices from CIE-exposed adolescent mice with a positive allosteric modulator of mGlu2 fully rescues mGlu2-LTD. In contrast to the 2-week CIE paradigm, acute exposure of striatal slices to ethanol concentrations that mimic ethanol levels during CIE exposure fails to disrupt mGlu2-LTD. We did not observe a reduction of mGlu2 mRNA or protein levels following CIE exposure, suggesting that alcohol effects on mGlu2 occur at the functional level. Our findings contribute to growing evidence that adolescents are uniquely vulnerable to certain alcohol-induced neuroadaptations, and identify enhancement of mGlu2 activity as a strategy to reverse the effects of adolescent alcohol exposure on DLS physiology.
Collapse
|
42
|
Salmanzadeh H, Ahmadi-Soleimani SM, Pachenari N, Azadi M, Halliwell RF, Rubino T, Azizi H. Adolescent drug exposure: A review of evidence for the development of persistent changes in brain function. Brain Res Bull 2020; 156:105-117. [PMID: 31926303 DOI: 10.1016/j.brainresbull.2020.01.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/28/2019] [Accepted: 01/06/2020] [Indexed: 12/24/2022]
Abstract
Over the past decade, many studies have indicated that adolescence is a critical period of brain development and maturation. The refinement and maturation of the central nervous system over this prolonged period, however, makes the adolescent brain highly susceptible to perturbations from acute and chronic drug exposure. Here we review the preclinical literature addressing the long-term consequences of adolescent exposure to common recreational drugs and drugs-of-abuse. These studies on adolescent exposure to alcohol, nicotine, opioids, cannabinoids and psychostimulant drugs, such as cocaine and amphetamine, reveal a variety of long-lasting behavioral and neurobiological consequences. These agents can affect development of the prefrontal cortex and mesolimbic dopamine pathways and modify the reward systems, socio-emotional processing and cognition. Other consequences include disruption in working memory, anxiety disorders and an increased risk of subsequent drug abuse in adult life. Although preventive and control policies are a valuable approach to reduce the detrimental effects of drugs-of-abuse on the adolescent brain, a more profound understanding of their neurobiological impact can lead to improved strategies for the treatment and attenuation of the detrimental neuropsychiatric sequelae.
Collapse
Affiliation(s)
- Hamed Salmanzadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; TJ Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA, USA
| | | | - Narges Pachenari
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Azadi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Robert F Halliwell
- TJ Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA, USA
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio, VA, Italy
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
43
|
Sanchez-Alavez M, Nguyen W, Mori S, Wills DN, Otero D, Aguirre CA, Singh M, Ehlers CL, Conti B. Time Course of Blood and Brain Cytokine/Chemokine Levels Following Adolescent Alcohol Exposure and Withdrawal in Rats. Alcohol Clin Exp Res 2019; 43:2547-2558. [PMID: 31589333 PMCID: PMC6904424 DOI: 10.1111/acer.14209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 10/01/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Adolescence is a critical period for neural development, and alcohol exposure during adolescence can lead to an elevated risk for health consequences as well as alcohol use disorders. Clinical and experimental data suggest that chronic alcohol exposure may produce immunomodulatory effects that can lead to the activation of pro-inflammatory cytokine pathways as well as microglial markers. The present study evaluated, in brain and blood, the effects of adolescent alcohol exposure and withdrawal on microglia and on the most representative pro- and anti-inflammatory cytokines and major chemokines that can contribute to the establishing of a neuroinflammatory environment. METHODS Wistar rats (males, n = 96) were exposed to ethanol (EtOH) vapors, or air control, for 5 weeks over adolescence (PD22-PD58). Brains and blood samples were collected at 3 time points: (i) after 35 days of vapor/air exposure (PD58); (ii) after 1 day of withdrawal (PD59), and (iii) 28 days after withdrawal (PD86). The ionized calcium-binding adapter molecule 1 (Iba-1) was used to index microglial activation, and cytokine/chemokine responses were analyzed using magnetic bead panels. RESULTS After 35 days of adolescent vapor exposure, a significant increase in Iba-1 immunoreactivity was seen in amygdala, frontal cortex, hippocampus, and substantia nigra. However, Iba-1 density returned to control levels at both 1 day and 28 days of withdrawal except in the hippocampus where Iba-1 density was significantly lower than controls. In serum, adolescent EtOH exposure induced a reduction in IL-13 and an increase in fractalkine at day 35. After 1 day of withdrawal, IL-18 was reduced, and IP-10 was elevated, whereas both IP-10 and IL-10 were elevated at 28 days following withdrawal. In the frontal cortex, adolescent EtOH exposure induced an increase in IL-1β at day 35, and 28 days of withdrawal, and IL-10 was increased after 28 days of withdrawal. CONCLUSION These data demonstrate that EtOH exposure during adolescence produces significant microglial activation; however, inflammatory markers seen in the blood appear to differ from those observed in the brain.
Collapse
Affiliation(s)
| | - William Nguyen
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Simone Mori
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Derek N Wills
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Dennis Otero
- Infectious and Inflammatory Disease Center and National Cancer Institute (NCI)-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Research Institute, La Jolla, California
| | - Carlos A Aguirre
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Mona Singh
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Bruno Conti
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
44
|
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
|
45
|
Gallant C, Good D. Alcohol misuse and traumatic brain injury: a review of the potential roles of dopaminergic dysfunction and physiological underarousal post-injury. APPLIED NEUROPSYCHOLOGY-ADULT 2019; 28:501-511. [PMID: 31561716 DOI: 10.1080/23279095.2019.1670181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although many researchers have demonstrated an increase in alcohol use following traumatic brain injury (TBI), there is also a body of research indicating that alcohol misuse predisposes one to injury and precedes TBI. Accordingly, various mechanisms have been proposed (e.g., self-medication, dampened levels of arousal, dopaminergic dysfunction, etc.) and variable results have emerged. This paper reviews the empirical evidence, for and against, TBI as a risk factor for alcohol misuse. In particular, this paper focuses on the brain-behavior relationships involved and examines the roles of physiological underarousal and dopaminergic dysfunction in the development of alcohol misuse after injury. Alcohol misuse impedes community reintegration among TBI survivors and creates additional rehabilitative challenges. Thus, in order to inform and improve treatment outcomes among this vulnerable population, a deeper understanding of the neural mechanisms implicated is needed.
Collapse
Affiliation(s)
- Caitlyn Gallant
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Dawn Good
- Department of Psychology, Brock University, St. Catharines, ON, Canada.,Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
46
|
Henricks AM, Dwiel LL, Deveau NH, Simon AA, Ruiz-Jaquez MJ, Green AI, Doucette WT. Corticostriatal Oscillations Predict High vs. Low Drinkers in a Rat Model of Limited Access Alcohol Consumption. Front Syst Neurosci 2019; 13:35. [PMID: 31456669 PMCID: PMC6700217 DOI: 10.3389/fnsys.2019.00035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 07/24/2019] [Indexed: 01/23/2023] Open
Abstract
Individuals differ in their vulnerability to develop alcohol dependence, which is determined by innate and environmental factors. The corticostriatal circuit is heavily involved in the development of alcohol dependence and may contain neural information regarding vulnerability to drink excessively. In the current experiment, we hypothesized that we could characterize high and low alcohol-drinking rats (HD and LD, respectively) based on corticostriatal oscillations and that these subgroups would differentially respond to corticostriatal brain stimulation. Male Sprague–Dawley rats (n = 13) were trained to drink 10% alcohol in a limited access paradigm. In separate sessions, local field potentials (LFPs) were recorded from the nucleus accumbens shell (NAcSh) and medial prefrontal cortex (mPFC). Based on training alcohol consumption levels, we classified rats using a median split as HD or LD. Then, using machine-learning, we built predictive models to classify rats as HD or LD by corticostriatal LFPs and compared the model performance from real data to the performance of models built on data permutations. Additionally, we explored the impact of NAcSh or mPFC stimulation on alcohol consumption in HD vs. LD. Corticostriatal LFPs were able to predict HD vs. LD group classification with greater accuracy than expected by chance (>80% accuracy). Moreover, NAcSh stimulation significantly reduced alcohol consumption in HD, but not LD (p < 0.05), while mPFC stimulation did not alter drinking behavior in either HD or LD (p > 0.05). These data collectively show that the corticostriatal circuit is differentially involved in regulating alcohol intake in HD vs. LD rats, and suggests that corticostriatal activity may have the potential to predict a vulnerability to develop alcohol dependence in a clinical population.
Collapse
Affiliation(s)
- Angela M Henricks
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Lucas L Dwiel
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Nicholas H Deveau
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Amanda A Simon
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Metztli J Ruiz-Jaquez
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Alan I Green
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.,The Dartmouth Clinical and Translational Science Institute, Dartmouth College, Hanover, NH, United States
| | - Wilder T Doucette
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.,The Dartmouth Clinical and Translational Science Institute, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
47
|
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: 122] [Impact Index Per Article: 20.3] [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
|
48
|
Henricks AM, Dwiel LL, Deveau NH, Simon AA, Ruiz-Jaquez MJ, Green AI, Doucette WT. Corticostriatal Oscillations Predict High vs. Low Drinkers in a Rat Model of Limited Access Alcohol Consumption. Front Syst Neurosci 2019; 13:35. [PMID: 31456669 PMCID: PMC6700217 DOI: 10.3389/fnsys.2019.00035 10.3389/fnsys.2019.00035/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 07/24/2019] [Indexed: 06/16/2024] Open
Abstract
Individuals differ in their vulnerability to develop alcohol dependence, which is determined by innate and environmental factors. The corticostriatal circuit is heavily involved in the development of alcohol dependence and may contain neural information regarding vulnerability to drink excessively. In the current experiment, we hypothesized that we could characterize high and low alcohol-drinking rats (HD and LD, respectively) based on corticostriatal oscillations and that these subgroups would differentially respond to corticostriatal brain stimulation. Male Sprague-Dawley rats (n = 13) were trained to drink 10% alcohol in a limited access paradigm. In separate sessions, local field potentials (LFPs) were recorded from the nucleus accumbens shell (NAcSh) and medial prefrontal cortex (mPFC). Based on training alcohol consumption levels, we classified rats using a median split as HD or LD. Then, using machine-learning, we built predictive models to classify rats as HD or LD by corticostriatal LFPs and compared the model performance from real data to the performance of models built on data permutations. Additionally, we explored the impact of NAcSh or mPFC stimulation on alcohol consumption in HD vs. LD. Corticostriatal LFPs were able to predict HD vs. LD group classification with greater accuracy than expected by chance (>80% accuracy). Moreover, NAcSh stimulation significantly reduced alcohol consumption in HD, but not LD (p < 0.05), while mPFC stimulation did not alter drinking behavior in either HD or LD (p > 0.05). These data collectively show that the corticostriatal circuit is differentially involved in regulating alcohol intake in HD vs. LD rats, and suggests that corticostriatal activity may have the potential to predict a vulnerability to develop alcohol dependence in a clinical population.
Collapse
Affiliation(s)
- Angela M. Henricks
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Lucas L. Dwiel
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Nicholas H. Deveau
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Amanda A. Simon
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Metztli J. Ruiz-Jaquez
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Alan I. Green
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- The Dartmouth Clinical and Translational Science Institute, Dartmouth College, Hanover, NH, United States
| | - Wilder T. Doucette
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- The Dartmouth Clinical and Translational Science Institute, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
49
|
Sey NYA, Gómez-A A, Madayag AC, Boettiger CA, Robinson DL. Adolescent intermittent ethanol impairs behavioral flexibility in a rat foraging task in adulthood. Behav Brain Res 2019; 373:112085. [PMID: 31319133 DOI: 10.1016/j.bbr.2019.112085] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/19/2022]
Abstract
Alcohol exposure is linked to behavioral flexibility deficits in humans, but it is unclear when the critical exposure occurred or if alcohol exposure alone is sufficient to produce behavior deficits. Increasing evidence shows that binge levels of alcohol during adolescence are particularly harmful to the brain, producing physiological and behavioral effects that can persist into adulthood. The present study determined whether adolescent intermittent ethanol (AIE) in rats impaired action selection in a discriminative stimulus task using a foraging response. Rats were exposed to ethanol during adolescence (5 g/kg/day, IG, 2-days-on/2-days-off, postnatal day 25-54). In adulthood, they learned to dig for food reward buried in one of two media, cued with one of two odors. AIE and control rats both learned to discriminate between olfactory cues, but AIE rats were impaired when reversing that learned association (first intra-dimensional reversal). However, AIE rats were faster to reinstate the original odor discrimination rule (second reversal), suggesting perseverative behavior. Next, the reward location was cued by digging media rather than odor. Both groups learned this extra-dimensional shift; however, control rats were slower to reach criterion. These findings are consistent with studies of people with substance abuse disorder, who learn new stimulus-response associations similarly to, or better than, control subjects, but perseverate when attempting to replace a well-learned association. These data suggest that adolescent binge-alcohol exposure contributes to behavioral flexibility deficits observed in adulthood.
Collapse
Affiliation(s)
- Nancy Y A Sey
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, USA
| | - Alexander Gómez-A
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA
| | - Aric C Madayag
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA
| | - Charlotte A Boettiger
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, USA; Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Donita L Robinson
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, USA; Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
50
|
Fritz M, Klawonn AM, Zahr NM. Neuroimaging in alcohol use disorder: From mouse to man. J Neurosci Res 2019; 100:1140-1158. [PMID: 31006907 DOI: 10.1002/jnr.24423] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
This article provides an overview of recent advances in understanding the effects of alcohol use disorders (AUD) on the brain from the perspective of magnetic resonance imaging (MRI) research in preclinical models and clinical studies. As a noninvasive investigational tool permitting assessment of morphological, metabolic, and hemodynamic changes over time, MRI offers insight into the dynamic course of alcoholism beginning with initial exposure through periods of binge drinking and escalation, sobriety, and relapse and has been useful in differential diagnosis of neurological diseases associated with AUD. Structural MRI has revealed acute and chronic effects of alcohol on both white and gray matter volumes. MR Spectroscopy, able to quantify brain metabolites in vivo, has shed light on biochemical alterations associated with alcoholism. Diffusion tensor imaging permits microstructural characterization of white matter fiber tracts. Functional MRI has allowed for elucidation of hemodynamic responses at rest and during task engagement. Positron emission tomography, a non-MRI imaging tool, has led to a deeper understanding of alcohol-induced receptor and neurotransmitter changes during various stages of drinking and abstinence. Together, such in vivo imaging tools have expanded our understanding of the dynamic course of alcoholism including evidence for regional specificity of the effects of AUD, hints at mechanisms underlying the shift from casual to compulsive use of alcohol, and profound recovery with sustained abstinence.
Collapse
Affiliation(s)
- Michael Fritz
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Anna M Klawonn
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Natalie M Zahr
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California.,Neuroscience Program, SRI International, Menlo Park, California
| |
Collapse
|