1
|
Angeles-Valdez D, López-Castro A, Rasgado-Toledo J, Naranjo-Albarrán L, Garza-Villarreal EA. Improved classification of alcohol intake groups in the Intermittent-Access Two-Bottle choice rat model using a latent class linear mixed model. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111397. [PMID: 40354870 DOI: 10.1016/j.pnpbp.2025.111397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Alcohol use disorder (AUD) is a major public health problem in which preclinical models allow the study of AUD development, phenotypes, and the exploration of potential new treatments. The intermittent access two-bottle choice (IA2BC) model is a validated preclinical model for studying alcohol intake patterns similar to human AUD clinical studies. Typically, the mean/median of overall alcohol intake or the last drinking sessions is used as a threshold to divide groups of animals into high or low alcohol consumers. Nevertheless, this approach has the potential for introducing bias due to the a priori selection of a threshold, as opposed to measuring the consumption drinking pattern along the protocol and subgrouping accordingly. This study aimed to assess the efficacy of utilizing longitudinal data of all drinking sessions to classify the population into high or low alcohol intake groups, employing a latent class linear mixed model (LCLMM). We compared LCLMM with traditional classification methods: (i) percentiles, (ii) K-means clustering, and (iii) hierarchical clustering. In addition, we used simulations to compare the accuracy, specificity, and sensitivity of these methods. By considering the entire trajectory of alcohol intake, LCLMM provides a more robust classification based on accuracy (0.94) between high and low alcohol classes. We recommend the use of longitudinal statistical models in research on substance use disorders in preclinical studies, since they could improve the classification of subpopulations.
Collapse
Affiliation(s)
- Diego Angeles-Valdez
- Institute of Neurobiology, Universidad Nacional Autónoma de México, campus Juriquilla, Querétaro, Mexico; Cognitive Neuroscience Center, Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, the Netherlands; Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, the Netherlands.
| | - Alejandra López-Castro
- Institute of Neurobiology, Universidad Nacional Autónoma de México, campus Juriquilla, Querétaro, Mexico
| | - Jalil Rasgado-Toledo
- Institute of Neurobiology, Universidad Nacional Autónoma de México, campus Juriquilla, Querétaro, Mexico
| | - Lizbeth Naranjo-Albarrán
- Department of Mathematics, Faculty of Sciences, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Eduardo A Garza-Villarreal
- Institute of Neurobiology, Universidad Nacional Autónoma de México, campus Juriquilla, Querétaro, Mexico.
| |
Collapse
|
2
|
Peng B, Wu XB, Zhang ZJ, Cao DL, Zhao LX, Wu H, Gao YJ. Anterior Cingulate Cortex Contributes to the Hyperlocomotion under Nitrogen Narcosis. Neurosci Bull 2025; 41:775-789. [PMID: 39158823 PMCID: PMC12015000 DOI: 10.1007/s12264-024-01278-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/20/2024] [Indexed: 08/20/2024] Open
Abstract
Nitrogen narcosis is a neurological syndrome that manifests when humans or animals encounter hyperbaric nitrogen, resulting in a range of motor, emotional, and cognitive abnormalities. The anterior cingulate cortex (ACC) is known for its significant involvement in regulating motivation, cognition, and action. However, its specific contribution to nitrogen narcosis-induced hyperlocomotion and the underlying mechanisms remain poorly understood. Here we report that exposure to hyperbaric nitrogen notably increased the locomotor activity of mice in a pressure-dependent manner. Concurrently, this exposure induced heightened activation among neurons in both the ACC and dorsal medial striatum (DMS). Notably, chemogenetic inhibition of ACC neurons effectively suppressed hyperlocomotion. Conversely, chemogenetic excitation lowered the hyperbaric pressure threshold required to induce hyperlocomotion. Moreover, both chemogenetic inhibition and genetic ablation of activity-dependent neurons within the ACC reduced the hyperlocomotion. Further investigation revealed that ACC neurons project to the DMS, and chemogenetic inhibition of ACC-DMS projections resulted in a reduction in hyperlocomotion. Finally, nitrogen narcosis led to an increase in local field potentials in the theta frequency band and a decrease in the alpha frequency band in both the ACC and DMS. These results collectively suggest that excitatory neurons within the ACC, along with their projections to the DMS, play a pivotal role in regulating the hyperlocomotion induced by exposure to hyperbaric nitrogen.
Collapse
Affiliation(s)
- Bin Peng
- Medical School, Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, 226019, China
| | - Xiao-Bo Wu
- Medical School, Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, 226019, China
| | - Zhi-Jun Zhang
- Medical School, Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, 226019, China
| | - De-Li Cao
- Medical School, Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, 226019, China
| | - Lin-Xia Zhao
- Medical School, Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, 226019, China
| | - Hao Wu
- Department of Otolaryngology-Head Neck Surgery, the Affiliated Hospital of Nantong University, Jiangsu, 226001, China
| | - Yong-Jing Gao
- Medical School, Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, 226019, China.
| |
Collapse
|
3
|
Duncan LE, Li T, Salem M, Li W, Mortazavi L, Senturk H, Shahverdizadeh N, Vesuna S, Shen H, Yoon J, Wang G, Ballon J, Tan L, Pruett BS, Knutson B, Deisseroth K, Giardino WJ. Mapping the cellular etiology of schizophrenia and complex brain phenotypes. Nat Neurosci 2025; 28:248-258. [PMID: 39833308 PMCID: PMC11802450 DOI: 10.1038/s41593-024-01834-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/29/2024] [Indexed: 01/22/2025]
Abstract
Psychiatric disorders are multifactorial and effective treatments are lacking. Probable contributing factors to the challenges in therapeutic development include the complexity of the human brain and the high polygenicity of psychiatric disorders. Combining well-powered genome-wide and brain-wide genetics and transcriptomics analyses can deepen our understanding of the etiology of psychiatric disorders. Here, we leverage two landmark resources to infer the cell types involved in the etiology of schizophrenia, other psychiatric disorders and informative comparison of brain phenotypes. We found both cortical and subcortical neuronal associations for schizophrenia, bipolar disorder and depression. These cell types included somatostatin interneurons, excitatory neurons from the retrosplenial cortex and eccentric medium spiny-like neurons from the amygdala. In contrast we found T cell and B cell associations with multiple sclerosis and microglial associations with Alzheimer's disease. We provide a framework for a cell-type-based classification system that can lead to drug repurposing or development opportunities and personalized treatments. This work formalizes a data-driven, cellular and molecular model of complex brain disorders.
Collapse
Affiliation(s)
- Laramie E Duncan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| | - Tayden Li
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Madeleine Salem
- Vice Provost for Undergraduate Education, Stanford University, Stanford, CA, USA
| | - Will Li
- Vice Provost for Undergraduate Education, Stanford University, Stanford, CA, USA
| | - Leili Mortazavi
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Hazal Senturk
- Department of Computer Science, University of San Francisco, San Francisco, CA, USA
| | - Naghmeh Shahverdizadeh
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sam Vesuna
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Hanyang Shen
- Department of Epidemiology, Stanford University, Stanford, CA, USA
| | - Jong Yoon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Gordon Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jacob Ballon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Longzhi Tan
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | | | - Brian Knutson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - William J Giardino
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
4
|
Bogdańska-Chomczyk E, Wojtacha P, Tsai ML, Huang ACW, Kozłowska A. Alterations in Striatal Architecture and Biochemical Markers' Levels During Postnatal Development in the Rat Model of an Attention Deficit/Hyperactivity Disorder (ADHD). Int J Mol Sci 2024; 25:13652. [PMID: 39769412 PMCID: PMC11680085 DOI: 10.3390/ijms252413652] [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] [Received: 11/06/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is defined as a neurodevelopmental condition. The precise underlying mechanisms remain incompletely elucidated. A body of research suggests disruptions in both the cellular architecture and neuronal function within the brain regions of individuals with ADHD, coupled with disturbances in the biochemical parameters. This study seeks to evaluate the morphological characteristics with a volume measurement of the striatal regions and a neuron density assessment within the studied areas across different developmental stages in Spontaneously Hypertensive Rats (SHRs) and Wistar Kyoto Rats (WKYs). Furthermore, the investigation aims to scrutinize the levels and activities of specific markers related to immune function, oxidative stress, and metabolism within the striatum of juvenile and maturing SHRs compared to WKYs. The findings reveal that the most pronounced reductions in striatal volume occur during the juvenile stage in SHRs, alongside alterations in neuronal density within these brain regions compared to WKYs. Additionally, SHRs exhibit heightened levels and activities of various markers, including RAC-alpha serine/threonine-protein kinase (AKT-1), glucocorticoid receptor (GCsRβ), malondialdehyde (MDA), sulfhydryl groups (-SH), glucose (G), iron (Fe), lactate dehydrogenase (LDH). alanine transaminase (ALT), and aspartate transaminase (AST). In summary, notable changes in striatal morphology and elevated levels of inflammatory, oxidative, and metabolic markers within the striatum may be linked to the disrupted brain development and maturation observed in ADHD.
Collapse
Affiliation(s)
- Ewelina Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Paweł Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Meng-Li Tsai
- Department of Biomechatronic Engineering, National Ilan University, Ylan 26047, Taiwan;
| | | | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| |
Collapse
|
5
|
Bocarsly ME, Shaw MJ, Ventriglia E, Anderson LG, Goldbach HC, Teresi CE, Bravo M, Bock R, Hong P, Kwon HB, Khawaja IM, Raman R, Murray EM, Bonaventura J, Burke DA, Michaelides M, Alvarez VA. Preexisting risk-avoidance and enhanced alcohol relief are driven by imbalance of the striatal dopamine receptors in mice. Nat Commun 2024; 15:9093. [PMID: 39438478 PMCID: PMC11496688 DOI: 10.1038/s41467-024-53414-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
Alcohol use disorder (AUD) is frequently comorbid with anxiety disorders, yet whether alcohol abuse precedes or follows the expression of anxiety remains unclear. Rodents offer control over the first drink, an advantage when testing the causal link between anxiety and AUD. Here, we utilized a risk-avoidance task to determine anxiety-like behaviors before and after alcohol exposure. We found that alcohol's anxiolytic efficacy varied among inbred mice and mice with high risk-avoidance showed heightened alcohol relief. While dopamine D1 receptors in the striatum are required for alcohol's relief, their levels alone were not correlated with relief. Rather, the ratio between striatal D1 and D2 receptors was a determinant factor for risk-avoidance and alcohol relief. We show that increasing striatal D1 to D2 receptor ratio was sufficient to promote risk-avoidance and enhance alcohol relief, even at initial exposure. Mice with high D1 to D2 receptor ratio were more prone to continue drinking despite adverse effects, a hallmark of AUD. These findings suggest that an anxiety phenotype may be a predisposing factor for AUD.
Collapse
Affiliation(s)
- Miriam E Bocarsly
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA.
- Department of Pharmacology, Physiology and Neuroscience, Brain Health Institute, Rutgers New Jersey Medical School, Newark, NJ, USA.
| | - Marlisa J Shaw
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- NIH Academy Enrichment Program, Office of OITE, NIH, Bethesda, MD, USA
| | - Emilya Ventriglia
- National Institute on Drug Abuse, Intramural Research Program, NIH, Baltimore, MD, USA
| | | | - Hannah C Goldbach
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- National Institute on Mental Health, NIH, Bethesda, MD, USA
| | - Catherine E Teresi
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Center on Compulsive Behaviors, NIH, Bethesda, MD, USA
| | - Marilyn Bravo
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Roland Bock
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- National Institute on Mental Health, NIH, Bethesda, MD, USA
| | - Patrick Hong
- Department of Pharmacology, Physiology and Neuroscience, Brain Health Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Han Bin Kwon
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Imran M Khawaja
- Department of Pharmacology, Physiology and Neuroscience, Brain Health Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Rishi Raman
- Department of Pharmacology, Physiology and Neuroscience, Brain Health Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Erin M Murray
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Jordi Bonaventura
- National Institute on Drug Abuse, Intramural Research Program, NIH, Baltimore, MD, USA
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Dennis A Burke
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Michael Michaelides
- National Institute on Drug Abuse, Intramural Research Program, NIH, Baltimore, MD, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA.
- National Institute on Drug Abuse, Intramural Research Program, NIH, Baltimore, MD, USA.
- National Institute on Mental Health, NIH, Bethesda, MD, USA.
- Center on Compulsive Behaviors, NIH, Bethesda, MD, USA.
| |
Collapse
|
6
|
Wei R, Zong F, Dong J, Zhao W, Zhang F, Wang W, Zhao S, Wang Z, Zhang F, Zhang HT. Identification of Phosphodiesterase-7A (PDE7A) as a Novel Target for Reducing Ethanol Consumption in Mice. Int J Neuropsychopharmacol 2024; 27:pyae032. [PMID: 39099166 PMCID: PMC11348009 DOI: 10.1093/ijnp/pyae032] [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: 04/09/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Ethanol elicits a rapid stimulatory effect and a subsequent, prolonged sedative response, which are potential predictors of EtOH consumption by decreasing adenosine signaling; this phenomenon also reflects the obvious sex difference. cAMP (cyclic Adenosine Monophosphate)-PKA (Protein Kinase A) signaling pathway modulation can influence the stimulatory and sedative effects induced by EtOH in mice. This study's objective is to clarify the role of phosphodiesterase (PDE) in mediating the observed sex differences in EtOH responsiveness between male and female animals. METHODS EtOH was administered i.p. for 7 days to identify the changes in PDE isoforms in response to EtOH treatment. Additionally, EtOH consumption and preference of male and female C57BL/6J mice were assessed using the drinking-in-the-dark and 2-bottle choice tests. Further, pharmacological inhibition of PDE7A heterozygote knockout mice was performed to investigate its effects on EtOH-induced stimulation and sedation in both male and female mice. Finally, Western blotting analysis was performed to evaluate the alterations in cAMP-PKA/Epac2 pathways. RESULTS EtOH administration resulted in an immediate upregulation in PDE7A expression in female mice, indicating a strong association between PDE7A and EtOH stimulation. Through the pharmacological inhibition of PDE7A KD mice, we have demonstrated for the first time, to our knowledge, that PDE7A selectively attenuates EtOH responsiveness and consumption exclusively in female mice, whichmay be associated with the cAMP-PKA/Epac2 pathway and downstream phosphorylation of CREB and ERK1/2. CONCLUSIONS Inhibition or knockdown of PDE7A attenuates EtOH responsivenessand consumption exclusively in female mice, which is associated with alterations in the cAMP-PKA/Epac2 signaling pathways, thereby highlighting its potential as a novel therapeutic target for alcohol use disorder.
Collapse
Affiliation(s)
- Ran Wei
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
- Weifang Chinese Medical Hospital, Shandong Second Medical University, Weifang, China
| | - Fangjiao Zong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Jiahao Dong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
- Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
| | - Wei Zhao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Fangfang Zhang
- Institude of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Wei Wang
- Institude of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Shuang Zhao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Ziqi Wang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Fang Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Han-Ting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| |
Collapse
|
7
|
Riveros ME, Leibold NK, Retamal MA, Ezquer F. Role of histaminergic regulation of astrocytes in alcohol use disorder. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111009. [PMID: 38653364 DOI: 10.1016/j.pnpbp.2024.111009] [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: 12/13/2023] [Revised: 02/26/2024] [Accepted: 04/14/2024] [Indexed: 04/25/2024]
Abstract
Alcohol use disorder (AUD) is a severe, yet not fully understood, mental health problem. It is associated with liver, pancreatic, and gastrointestinal diseases, thereby highly increasing the morbidity and mortality of these individuals. Currently, there is no effective and safe pharmacological therapy for AUD. Therefore, there is an urgent need to increase our knowledge about its neurophysiological etiology to develop new treatments specifically targeted at this health condition. Recent findings have shown an upregulation in the histaminergic system both in alcohol dependent individuals and in animals with high alcohol preference. The use of H3 histaminergic receptor antagonists has given promising therapeutic results in animal models of AUD. Interestingly, astrocytes, which are ubiquitously present in the brain, express the three main histamine receptors (H1, H2 and H3), and in the last few years, several studies have shown that astrocytes could play an important role in the development and maintenance of AUD. Accordingly, alterations in the density of astrocytes in brain areas such as the prefrontal cortex, ventral striatum, and hippocampus that are critical for AUD-related characteristics have been observed. These characteristics include addiction, impulsivity, motor function, and aggression. In this work, we review the current state of knowledge on the relationship between the histaminergic system and astrocytes in AUD and propose that histamine could increase alcohol tolerance by protecting astrocytes from ethanol-induced oxidative stress. This increased tolerance could lead to high levels of alcohol intake and therefore could be a key factor in the development of alcohol dependence.
Collapse
Affiliation(s)
- María Eugenia Riveros
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.
| | - Nicole K Leibold
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile; Programa de Comunicación Celular en Cáncer, Instituto de Ciencia e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Instituto de Ciencia e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago. Chile; Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago, Chile
| |
Collapse
|
8
|
Pati D, Taxier LR, Xia M, Lee SI, Conley SY, Sides T, Boyt KM, Hunker AC, Zweifel LS, Kash TL. Dopamine D2 receptors in the bed nucleus of the stria terminalis modulate alcohol-related behaviors. ADDICTION NEUROSCIENCE 2024; 11:100157. [PMID: 39741698 PMCID: PMC11687822 DOI: 10.1016/j.addicn.2024.100157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Dysregulation of the dopamine (DA) system is a hallmark of substance use disorders, including alcohol use disorder (AUD). Of the DA receptor subtypes, the DA D2 receptors (D2Rs) play a key role in the reinforcing effects of alcohol. D2Rs are expressed in numerous brain regions associated with the regulation of appetitive behaviors. One such region is the bed nucleus of the stria terminalis (BNST), which has been linked to the development and maintenance of AUD. Recently, we identified alcohol withdrawal-related neuroadaptations in the periaqueductal gray/dorsal raphe to BNST DA circuit in male mice. However, the role of D2R-expressing BNST neurons in voluntary alcohol consumption is not well characterized. In this study, we used a CRISPR-Cas9-based viral approach, to selectively reduce the expression of D2Rs in BNST GABA neurons (BNST vgat Drd2) and interrogated the impact on alcohol-related behaviors. In male mice, reduced BNST vgat Drd2 expression potentiated the stimulatory effects of alcohol and increased voluntary consumption of 20% w/v alcohol in a two-bottle choice intermittent access paradigm. This effect was not specific to alcohol, as BNST vgat Drd2 knockdown also increased sucrose intake in male mice. Interestingly, reduction in BNST vgat Drd2 expression in female mice did not alter alcohol-related behaviors but lowered the threshold for mechanical pain sensitivity. Collectively, our findings suggest a role for postsynaptic BNST D2Rs in the modulation of sex-specific behavioral responses to alcohol and sucrose.
Collapse
Affiliation(s)
- Dipanwita Pati
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lisa R. Taxier
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mengfan Xia
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sophia I. Lee
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sara Y. Conley
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum of Neuroscience, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Tori Sides
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kristen M. Boyt
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Avery C. Hunker
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Larry S. Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA, USA
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
9
|
Dilly GA, Blednov YA, Warden AS, Ezerskiy L, Fleischer C, Plotkin JD, Patil S, Osterndorff-Kahanek EA, Mayfield J, Mayfield RD, Homanics GE, Messing RO. Knockdown of Tlr3 in dorsal striatum reduces ethanol consumption and acute functional tolerance in male mice. Brain Behav Immun 2024; 118:437-448. [PMID: 38499210 PMCID: PMC11007683 DOI: 10.1016/j.bbi.2024.03.021] [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: 08/16/2023] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Systemic activation of toll-like receptor 3 (TLR3) signaling using poly(I:C), a TLR3 agonist, drives ethanol consumption in several rodent models, while global knockout of Tlr3 reduces drinking in C57BL/6J male mice. To determine if brain TLR3 pathways are involved in drinking behavior, we used CRISPR/Cas9 genome editing to generate a Tlr3 floxed (Tlr3F/F) mouse line. After sequence confirmation and functional validation of Tlr3 brain transcripts, we injected Tlr3F/F male mice with an adeno-associated virus expressing Cre recombinase (AAV5-CMV-Cre-GFP) to knockdown Tlr3 in the medial prefrontal cortex, nucleus accumbens, or dorsal striatum (DS). Only Tlr3 knockdown in the DS decreased two-bottle choice, every-other-day (2BC-EOD) ethanol consumption. DS-specific deletion of Tlr3 also increased intoxication and prevented acute functional tolerance to ethanol. In contrast, poly(I:C)-induced activation of TLR3 signaling decreased intoxication in male C57BL/6J mice, consistent with its ability to increase 2BC-EOD ethanol consumption in these mice. We also found that TLR3 was highly colocalized with DS neurons. AAV5-Cre transfection occurred predominantly in neurons, but there was minimal transfection in astrocytes and microglia. Collectively, our previous and current studies show that activating or inhibiting TLR3 signaling produces opposite effects on acute responses to ethanol and on ethanol consumption. While previous studies, however, used global knockout or systemic TLR3 activation (which alter peripheral and brain innate immune responses), the current results provide new evidence that brain TLR3 signaling regulates ethanol drinking. We propose that activation of TLR3 signaling in DS neurons increases ethanol consumption and that a striatal TLR3 pathway is a potential target to reduce excessive drinking.
Collapse
Affiliation(s)
- Geoffrey A Dilly
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Lubov Ezerskiy
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Caleb Fleischer
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Jesse D Plotkin
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Shruti Patil
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | | | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Gregg E Homanics
- Departments of Anesthesiology & Perioperative Medicine, Neurobiology, and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Robert O Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
10
|
da Silva D, Matsui A, Murray EM, Mamais A, Authement ME, Shin JH, Shaw M, Ron D, Cookson MR, Alvarez VA. Leucine-rich repeat kinase 2 limits dopamine D1 receptor signaling in striatum and biases against heavy persistent alcohol drinking. Neuropsychopharmacology 2024; 49:824-836. [PMID: 37684522 PMCID: PMC10948780 DOI: 10.1038/s41386-023-01731-z] [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: 03/30/2023] [Revised: 07/25/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023]
Abstract
The transition from hedonic alcohol drinking to problematic drinking is a hallmark of alcohol use disorder that occurs only in a subset of drinkers. This transition requires long-lasting changes in the synaptic drive and the activity of striatal neurons expressing dopamine D1 receptor (D1R). The molecular mechanisms that generate vulnerability in some individuals to undergo the transition are less understood. Here, we report that the Parkinson's-related protein leucine-rich repeat kinase 2 (LRRK2) modulates striatal D1R function to affect the behavioral response to alcohol and the likelihood that mice transition to heavy, persistent alcohol drinking. Constitutive deletion of the Lrrk2 gene specifically from D1R-expressing neurons potentiated D1R signaling at the cellular and synaptic level and enhanced alcohol-related behaviors and drinking. Mice with cell-specific deletion of Lrrk2 were more prone to heavy alcohol drinking, and consumption was insensitive to punishment. These findings identify a potential novel role for LRRK2 function in the striatum in promoting resilience against heavy and persistent alcohol drinking.
Collapse
Affiliation(s)
- Daniel da Silva
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Aya Matsui
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Erin M Murray
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD, 20892, USA
| | - Michael E Authement
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Jung Hoon Shin
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Marlisa Shaw
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD, 20892, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA.
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, 20892, USA.
- NIMH, National Institutes of Health, Bethesda, USA.
| |
Collapse
|
11
|
Khikhmetova K, Semenova Y, Bjørklund G. The Roles of Endogenous D2R Dopamine and μ-opioid Receptors of the Brain in Alcohol use Disorder. Curr Med Chem 2024; 31:6393-6406. [PMID: 37921171 DOI: 10.2174/0109298673248999231013043249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 09/02/2023] [Accepted: 09/15/2023] [Indexed: 11/04/2023]
Abstract
Alcohol use disorder (AUD) affects millions of people worldwide. It is characterized by a strong physiological and psychological craving to consume large amounts of alcohol despite adverse consequences. Alcohol use disorder carries a large health and economic burden on society. Despite its prevalence, AUD is still severely undertreated. The precise molecular mechanisms of how alcohol addiction forms are yet unknown. However, previous studies on animal models show that along with the μ-opioid receptors, the D2R dopamine receptors may also be involved in alcohol craving and reward pathways. Currently, there is a limited number of treatment strategies for alcohol use disorder, which include several medications and therapy. By understanding the limitations of current treatment options and exploring new potential targets, it could be possible to find more effective ways of treating AUD in the future.
Collapse
Affiliation(s)
| | - Yuliya Semenova
- School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| |
Collapse
|
12
|
Cavallaro J, Yeisley J, Akdoǧan B, Salazar RE, Floeder JR, Balsam PD, Gallo EF. Dopamine D2 receptors in nucleus accumbens cholinergic interneurons increase impulsive choice. Neuropsychopharmacology 2023; 48:1309-1317. [PMID: 37221325 PMCID: PMC10354036 DOI: 10.1038/s41386-023-01608-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/25/2023]
Abstract
Impulsive choice, often characterized by excessive preference for small, short-term rewards over larger, long-term rewards, is a prominent feature of substance use and other neuropsychiatric disorders. The neural mechanisms underlying impulsive choice are not well understood, but growing evidence implicates nucleus accumbens (NAc) dopamine and its actions on dopamine D2 receptors (D2Rs). Because several NAc cell types and afferents express D2Rs, it has been difficult to determine the specific neural mechanisms linking NAc D2Rs to impulsive choice. Of these cell types, cholinergic interneurons (CINs) of the NAc, which express D2Rs, have emerged as key regulators of striatal output and local dopamine release. Despite these relevant functions, whether D2Rs expressed specifically in these neurons contribute to impulsive choice behavior is unknown. Here, we show that D2R upregulation in CINs of the mouse NAc increases impulsive choice as measured in a delay discounting task without affecting reward magnitude sensitivity or interval timing. Conversely, mice lacking D2Rs in CINs showed decreased delay discounting. Furthermore, CIN D2R manipulations did not affect probabilistic discounting, which measures a different form of impulsive choice. Together, these findings suggest that CIN D2Rs regulate impulsive decision-making involving delay costs, providing new insight into the mechanisms by which NAc dopamine influences impulsive behavior.
Collapse
Affiliation(s)
| | - Jenna Yeisley
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Başak Akdoǧan
- Department of Psychology, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Ronald E Salazar
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Joseph R Floeder
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Peter D Balsam
- Department of Psychology, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
- Department of Neuroscience and Behavior, Barnard College, New York, NY, USA
| | - Eduardo F Gallo
- Department of Biological Sciences, Fordham University, Bronx, NY, USA.
| |
Collapse
|
13
|
Pati D, Lee SI, Conley SY, Sides T, Boyt KM, Hunker AC, Zweifel LS, Kash TL. Dopamine D2 receptors in the bed nucleus of the stria terminalis modulate alcohol-related behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544820. [PMID: 37398115 PMCID: PMC10312666 DOI: 10.1101/2023.06.13.544820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Dysregulation of the dopamine (DA) system is a hallmark of substance abuse disorders, including alcohol use disorder (AUD). Of the DA receptor subtypes, the DA D2 receptors (D2Rs) play a key role in the reinforcing effects of alcohol. D2Rs are expressed in numerous brain regions associated with the regulation of appetitive behaviors. One such region is the bed nucleus of the stria terminalis (BNST), which has been linked to the development and maintenance of AUD. Recently, we identified alcohol withdrawal-related neuroadaptations in the periaqueductal gray/dorsal raphe to BNST DA circuit in male mice. However, the role of D2R-expressing BNST neurons in voluntary alcohol consumption is not well characterized. In this study, we used a CRISPR-Cas9-based viral approach, to selectively reduce the expression of D2Rs in BNST VGAT neurons and interrogated the impact of BNST D2Rs in alcohol-related behaviors. In male mice, reduced D2R expression potentiated the stimulatory effects of alcohol and increased voluntary consumption of 20% w/v alcohol in a two-bottle choice intermittent access paradigm. This effect was not specific to alcohol, as D2R deletion also increased sucrose intake in male mice. Interestingly, cell-specific deletion of BNST D2Rs in female mice did not alter alcohol-related behaviors but lowered the threshold for mechanical pain sensitivity. Collectively, our findings suggest a role for postsynaptic BNST D2Rs in the modulation of sex-specific behavioral responses to alcohol and sucrose.
Collapse
Affiliation(s)
- Dipanwita Pati
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sophia I. Lee
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sara Y. Conley
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum of Neuroscience, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Tori Sides
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristen M. Boyt
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Avery C. Hunker
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Larry S. Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA, USA
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
14
|
Harmata GIS, Chan AC, Merfeld MJ, Taugher-Hebl RJ, Harijan AK, Hardie JB, Fan R, Long JD, Wang GZ, Dlouhy BJ, Bera AK, Narayanan NS, Wemmie JA. Intoxicating effects of alcohol depend on acid-sensing ion channels. Neuropsychopharmacology 2023; 48:806-815. [PMID: 36243771 PMCID: PMC10066229 DOI: 10.1038/s41386-022-01473-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/06/2022] [Accepted: 09/23/2022] [Indexed: 11/09/2022]
Abstract
Persons at risk for developing alcohol use disorder (AUD) differ in their sensitivity to acute alcohol intoxication. Alcohol effects are complex and thought to depend on multiple mechanisms. Here, we explored whether acid-sensing ion channels (ASICs) might play a role. We tested ASIC function in transfected CHO cells and amygdala principal neurons, and found alcohol potentiated currents mediated by ASIC1A homomeric channels, but not ASIC1A/2 A heteromeric channels. Supporting a role for ASIC1A in the intoxicating effects of alcohol in vivo, we observed marked alcohol-induced changes on local field potentials in basolateral amygdala, which differed significantly in Asic1a-/- mice, particularly in the gamma, delta, and theta frequency ranges. Altered electrophysiological responses to alcohol in mice lacking ASIC1A, were accompanied by changes in multiple behavioral measures. Alcohol administration during amygdala-dependent fear conditioning dramatically diminished context and cue-evoked memory on subsequent days after the alcohol had cleared. There was a significant alcohol by genotype interaction. Context- and cue-evoked memory were notably worse in Asic1a-/- mice. We further examined acute stimulating and sedating effects of alcohol on locomotor activity, loss of righting reflex, and in an acute intoxication severity scale. We found loss of ASIC1A increased the stimulating effects of alcohol and reduced the sedating effects compared to wild-type mice, despite similar blood alcohol levels. Together these observations suggest a novel role for ASIC1A in the acute intoxicating effects of alcohol in mice. They further suggest that ASICs might contribute to intoxicating effects of alcohol and AUD in humans.
Collapse
Affiliation(s)
- Gail I S Harmata
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
- Pharmacological Sciences Predoctoral Research Training Program, Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Aubrey C Chan
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Madison J Merfeld
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Rebecca J Taugher-Hebl
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Anjit K Harijan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Jason B Hardie
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Rong Fan
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | - Grace Z Wang
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Brian J Dlouhy
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Amal K Bera
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Nandakumar S Narayanan
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | - John A Wemmie
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA.
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA.
- Department of Veterans Affairs Medical Center, Iowa City, IA, USA.
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
- Roy J. Carver Chair of Psychiatry and Neuroscience, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
Moreira-Júnior RE, Guimarães MADF, Etcheverria da Silva M, Maioli TU, Faria AMC, Brunialti-Godard AL. Animal model for high consumption and preference of ethanol and its interplay with high sugar and butter diet, behavior, and neuroimmune system. Front Nutr 2023; 10:1141655. [PMID: 37063320 PMCID: PMC10097969 DOI: 10.3389/fnut.2023.1141655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/10/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction Mechanisms that dictate the preference for ethanol and its addiction are not only restricted to the central nervous system (CNS). An increasing body of evidence has suggested that abusive ethanol consumption directly affects the immune system, which in turn interacts with the CNS, triggering neuronal responses and changes, resulting in dependence on the drug. It is known that neuroinflammation and greater immune system reactivity are observed in behavioral disorders and that these can regulate gene transcription. However, there is little information about these findings of the transcriptional profile of reward system genes in high consumption and alcohol preference. In this regard, there is a belief that, in the striatum, an integrating region of the brain reward system, the interaction of the immune response and the transcriptional profile of the Lrrk2 gene that is associated with loss of control and addiction to ethanol may influence the alcohol consumption and preference. Given this information, this study aimed to assess whether problematic alcohol consumption affects the transcriptional profile of the Lrrk2 gene, neuroinflammation, and behavior and whether these changes are interconnected. Methods An animal model developed by our research group has been used in which male C57BL/6 mice and knockouts for the Il6 and Nfat genes were subjected to a protocol of high fat and sugar diet intake and free choice of ethanol in the following stages: Stage 1 (T1)-Dietary treatment, for 8 weeks, in which the animals receive high-calorie diet, High Sugar and Butter (HSB group), or standard diet, American Institute of Nutrition 93-Growth (AIN93G group); and Stage 2 (T2)-Ethanol consumption, in which the animals are submitted, for 4 weeks, to alcohol within the free choice paradigm, being each of them divided into 10 groups, four groups continued with the same diet and in the other six the HSB diet is substituted by the AIN93G diet. Five groups had access to only water, while the five others had a free choice between water and a 10% ethanol solution. The weight of the animals was evaluated weekly and the consumption of water and ethanol daily. At the end of the 12-week experiment, anxiety-like behavior was evaluated by the light/dark box test; compulsive-like behavior by Marble burying, transcriptional regulation of genes Lrrk2, Tlr4, Nfat, Drd1, Drd2, Il6, Il1β, Il10, and iNOS by RT-qPCR; and inflammatory markers by flow cytometry. Animals that the diet was replaced had an ethanol high preference and consumption. Results and discussion We observed that high consumption and preference for ethanol resulted in (1) elevation of inflammatory cells in the brain, (2) upregulation of genes associated with cytokines (Il6 and Il1β) and pro-inflammatory signals (iNOS and Nfat), downregulation of anti-inflammatory cytokine (Il10), dopamine receptor (Drd2), and the Lrrk2 gene in the striatum, and (3) behavioral changes such as decreased anxiety-like behavior, and increased compulsive-like behavior. Our findings suggest that interactions between the immune system, behavior, and transcriptional profile of the Lrrk2 gene influence the ethanol preferential and abusive consumption.
Collapse
Affiliation(s)
- Renato Elias Moreira-Júnior
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Andrade de Freitas Guimarães
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Miguel Etcheverria da Silva
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lúcia Brunialti-Godard
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
16
|
Konar-Nié M, Guzman-Castillo A, Armijo-Weingart L, Aguayo LG. Aging in nucleus accumbens and its impact on alcohol use disorders. Alcohol 2023; 107:73-90. [PMID: 36087859 DOI: 10.1016/j.alcohol.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 02/06/2023]
Abstract
Ethanol is one of the most widely consumed drugs in the world and prolonged excessive ethanol intake might lead to alcohol use disorders (AUDs), which are characterized by neuroadaptations in different brain regions, such as in the reward circuitry. In addition, the global population is aging, and it appears that they are increasing their ethanol consumption. Although research involving the effects of alcohol in aging subjects is limited, differential effects have been described. For example, studies in human subjects show that older adults perform worse in tests assessing working memory, attention, and cognition as compared to younger adults. Interestingly, in the field of the neurobiological basis of ethanol actions, there is a significant dichotomy between what we know about the effects of ethanol on neurochemical targets in young animals and how it might affect them in the aging brain. To be able to understand the distinct effects of ethanol in the aging brain, the following questions need to be answered: (1) How does physiological aging impact the function of an ethanol-relevant region (e.g., the nucleus accumbens)? and (2) How does ethanol affect these neurobiological systems in the aged brain? This review discusses the available data to try to understand how aging affects the nucleus accumbens (nAc) and its neurochemical response to alcohol. The data show that there is little information on the effects of ethanol in aged mice and rats, and that many studies had considered 2-3-month-old mice as adults, which needs to be reconsidered since more recent literature defines 6 months as young adults and >18 months as an older mouse. Considering the actual relevance of an aged worldwide population and that this segment is drinking more frequently, it appears at least reasonable to explore how ethanol affects the brain in adult and aged models.
Collapse
Affiliation(s)
- Macarena Konar-Nié
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile.
| | - Alejandra Guzman-Castillo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile; Programa en Neurociencia, Psiquiatría y Salud Mental, Universidad de Concepción, Concepcion, Chile.
| | - Lorena Armijo-Weingart
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile; Programa en Neurociencia, Psiquiatría y Salud Mental, Universidad de Concepción, Concepcion, Chile.
| | - Luis Gerardo Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile; Programa en Neurociencia, Psiquiatría y Salud Mental, Universidad de Concepción, Concepcion, Chile.
| |
Collapse
|
17
|
Sitzia G, Lovinger DM. Circuit dysfunctions of associative and sensorimotor basal ganglia loops in alcohol use disorder: insights from animal models. ADDICTION NEUROSCIENCE 2023; 5:100056. [PMID: 36567745 PMCID: PMC9788651 DOI: 10.1016/j.addicn.2022.100056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Persons that develop Alcohol Use Disorder (AUD) experience behavioral changes that include compulsion to seek and take alcohol despite its negative consequences on the person's psychosocial, health and economic spheres, inability to limit alcohol intake and a negative emotional/ motivational state that emerges during withdrawal. During all the stages of AUD executive functions, i.e. the person's ability to direct their behavior towards a goal, working memory and cognitive flexibility are eroded. Animal models of AUD recapitulate aspects of action selection impairment and offer the opportunity to benchmark the underlying circuit mechanisms. Here we propose a circuit-based approach to AUD research focusing on recent advances in behavioral analysis, neuroanatomy, genetics, and physiology to guide future research in the field.
Collapse
Affiliation(s)
- Giacomo Sitzia
- Current Address: Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, USA
- Molecular Neurophysiology Laboratory, Department of Physiology and Pharmacology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - David M. Lovinger
- Current Address: Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, USA
| |
Collapse
|
18
|
Hamel R, Demers O, Boileau C, Roy ML, Théoret H, Bernier PM, Lepage JF. The neurobiological markers of acute alcohol's subjective effects in humans. Neuropsychopharmacology 2022; 47:2101-2110. [PMID: 35701548 PMCID: PMC9556716 DOI: 10.1038/s41386-022-01354-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 12/21/2022]
Abstract
The ingestion of alcohol yields acute biphasic subjective effects: stimulation before sedation. Despite their predictive relevance to the development of alcohol use disorders (AUD), the neurobiological markers accounting for the biphasic effects of alcohol remain poorly understood in humans. Informed by converging lines of evidence, this study tested the hypothesis that alcohol ingestion acutely increases gamma-aminobutyric acid (GABA)-mediated inhibition, which would positively and negatively predict the feeling of stimulation and sedation, respectively. To do so, healthy participants (n = 20) ingested a single dose of 94% ABV alcohol (males: 1.0 ml/kg; females: 0.85 ml/kg) in a randomized placebo-controlled cross-over design. The alcohol's biphasic effects were assessed with the Brief-Biphasic Alcohol Effects Scale, and non-invasive neurobiological markers were measured with transcranial magnetic stimulation, before and every 30 min (up to 120 min) after the complete ingestion of the beverage. Results showed that acute alcohol ingestion selectively increased the duration of the cortical silent period (CSP) as compared to placebo, suggesting that alcohol increases non-specific GABAergic inhibition. Importantly, CSP duration positively and negatively predicted increases in the feeling of stimulation and sedation, respectively, suggesting that stimulation emerges as GABAergic inhibition increases and that sedation emerges as GABAergic inhibition returns to baseline values. Overall, these results suggest that modulations of GABAergic inhibition are central to the acute biphasic subjective effects of alcohol, providing a potential preventive target to curb the progression of at-risk individuals to AUD.
Collapse
Affiliation(s)
- Raphael Hamel
- Département de kinanthropologie, Faculté des sciences de l'activité physique, Université de Sherbrooke, Sherbrooke, QC, Canada
- Département de pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Demers
- Département de pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Camille Boileau
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Marie-Laurence Roy
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Théoret
- Département de psychologie, Faculté des arts et sciences, Université de Montréal, Montreal, QC, Canada
| | - Pierre-Michel Bernier
- Département de kinanthropologie, Faculté des sciences de l'activité physique, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Francois Lepage
- Département de pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
19
|
Simpson EH, Gallo EF, Balsam PD, Javitch JA, Kellendonk C. How changes in dopamine D2 receptor levels alter striatal circuit function and motivation. Mol Psychiatry 2022; 27:436-444. [PMID: 34385603 PMCID: PMC8837728 DOI: 10.1038/s41380-021-01253-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023]
Abstract
It was first posited, more than five decades ago, that the etiology of schizophrenia involves overstimulation of dopamine receptors. Since then, advanced clinical research methods, including brain imaging, have refined our understanding of the relationship between striatal dopamine and clinical phenotypes as well as disease trajectory. These studies point to striatal dopamine D2 receptors, the main target for all current antipsychotic medications, as being involved in both positive and negative symptoms. Simultaneously, animal models have been central to investigating causal relationships between striatal dopamine D2 receptors and behavioral phenotypes relevant to schizophrenia. We begin this article by reviewing the circuit, cell-type and subcellular locations of dopamine D2 receptors and their downstream signaling pathways. We then summarize results from several mouse models in which D2 receptor levels were altered in various brain regions, cell-types and developmental periods. Behavioral, electrophysiological and anatomical consequences of these D2 receptor perturbations are reviewed with a selective focus on striatal circuit function and alterations in motivated behavior, a core negative symptom of schizophrenia. These studies show that D2 receptors serve distinct physiological roles in different cell types and at different developmental time points, regulating motivated behaviors in sometimes opposing ways. We conclude by considering the clinical implications of this complex regulation of striatal circuit function by D2 receptors.
Collapse
Affiliation(s)
- Eleanor H. Simpson
- Division of Developmental Neuroscience, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States
| | - Eduardo F. Gallo
- Department of Biological Sciences, Fordham University, 441 East Fordham Road, Bronx, NY 10458
| | - Peter D. Balsam
- Division of Developmental Neuroscience, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States,Department of Psychology, Barnard College, 3009 Broadway, New York, NY 10027,Department of Psychology, Columbia University, 1190 Amsterdam Ave, New York, NY 10027
| | - Jonathan A. Javitch
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States,Division of Molecular Therapeutics, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032,Department of Molecular Pharmacology and Therapeutics, Columbia University, 1051 Riverside Drive, New York, NY 10032
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University, New York, NY, USA. .,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA. .,Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA.
| |
Collapse
|
20
|
Egervari G, Siciliano CA, Whiteley EL, Ron D. Alcohol and the brain: from genes to circuits. Trends Neurosci 2021; 44:1004-1015. [PMID: 34702580 PMCID: PMC8616825 DOI: 10.1016/j.tins.2021.09.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 01/27/2023]
Abstract
Alcohol use produces wide-ranging and diverse effects on the central nervous system. It influences intracellular signaling mechanisms, leading to changes in gene expression, chromatin remodeling, and translation. As a result of these molecular alterations, alcohol affects the activity of neuronal circuits. Together, these mechanisms produce long-lasting cellular adaptations in the brain that in turn can drive the development and maintenance of alcohol use disorder (AUD). We provide an update on alcohol research, focusing on multiple levels of alcohol-induced adaptations, from intracellular changes to changes in neural circuits. A better understanding of how alcohol affects these diverse and interlinked mechanisms may lead to the identification of novel therapeutic targets and to the development of much-needed novel and efficacious treatment options.
Collapse
Affiliation(s)
- Gabor Egervari
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37203, USA.
| | - Ellanor L Whiteley
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
21
|
Gong S, Fayette N, Heinsbroek JA, Ford CP. Cocaine shifts dopamine D2 receptor sensitivity to gate conditioned behaviors. Neuron 2021; 109:3421-3435.e5. [PMID: 34506723 PMCID: PMC8571051 DOI: 10.1016/j.neuron.2021.08.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/16/2021] [Accepted: 08/11/2021] [Indexed: 12/21/2022]
Abstract
Cocaine addiction is a chronic, relapsing disorder characterized by maladaptation in the brain mesolimbic and nigrostriatal dopamine system. Although changes in the properties of D2-receptor-expressing medium spiny neurons (D2-MSNs) and connected striatal circuits following cocaine treatment are known, the contributions of altered D2-receptor (D2R) function in mediating the rewarding properties of cocaine remain unclear. Here, we describe how a 7-day exposure to cocaine alters dopamine signaling by selectively reducing the sensitivity, but not the expression, of nucleus accumbens D2-MSN D2Rs via an alteration in the relative expression and coupling of G protein subunits. This cocaine-induced reduction of D2R sensitivity facilitated the development of the rewarding effects of cocaine as blocking the reduction in G protein expression was sufficient to prevent cocaine-induced behavioral adaptations. These findings identify an initial maladaptive change in sensitivity by which mesolimbic dopamine signals are encoded by D2Rs following cocaine exposure.
Collapse
Affiliation(s)
- Sheng Gong
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicholas Fayette
- Department of Anesthesiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
22
|
Hong SI, Kang S, Baker M, Choi DS. Astrocyte-neuron interaction in the dorsal striatum-pallidal circuits and alcohol-seeking behaviors. Neuropharmacology 2021; 198:108759. [PMID: 34433087 DOI: 10.1016/j.neuropharm.2021.108759] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 12/31/2022]
Abstract
In the striatum, two main types of GABAergic medium spiny neurons (MSNs), denoted striatonigral (or direct-pathway MSNs, dMSNs) and striatopallidal neurons (indirect-pathway MSNs, iMSNs), form circuits with distinct pallidal nuclei, which sends "GO" or "NO-GO" signals through the thalamus. These striatopallidal circuits evaluate and execute reward-seeking and taking behaviors. Especially, the dorsal striatum can be further divided into the dorsomedial striatum (DMS, equivalent to caudate in primates and humans) and dorsolateral striatum (DLS, equivalent to putamen), which orchestrates goal-directed and habitual reward-seeking and taking behaviors, respectively. Using optogenetics, chemogenetics and in vivo calcium imaging technologies combined with electrophysiology and digitalized behavior phenotyping, recent studies have revealed cell-, circuit- and context-specific functions of these microcircuits in addictive behaviors. Also, region-specific astrocytes regulate the homeostatic activities of the dMSNs and iMSNs as well as the downstream circuits, which determine the net balance of cortico-striato-pallidal activities to the thalamic neurons. This review will summarize the recent progress of striatopallidal circuits focusing on astrocyte-neuron interaction and, reward- and alcohol-seeking behaviors. Our review will also discuss the translational and clinical implications of these microcircuit studies. This article is part of the special Issue on "Neurocircuitry Modulating Drug and Alcohol Abuse".
Collapse
Affiliation(s)
- Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA
| | - Seungwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA
| | - Matthew Baker
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
| |
Collapse
|
23
|
García-García F, Priego-Fernández S, López-Muciño LA, Acosta-Hernández ME, Peña-Escudero C. Increased alcohol consumption in sleep-restricted rats is mediated by delta FosB induction. Alcohol 2021; 93:63-70. [PMID: 33662520 DOI: 10.1016/j.alcohol.2021.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
The reduction of sleep hours is a public health problem in contemporary society. It is estimated that humans sleep between 1.5 and 2 h less, per night, than 100 years ago. The reduction of sleep hours is a risk factor for developing cardiovascular, metabolic, and psychiatric problems. Previous studies have shown that low sleep quality is a factor that favors relapse in addicted patients. In rodents, sleep deprivation increases the preference for methylphenidate and the self-administration of cocaine. However, it is unknown whether chronic sleep restriction induces voluntary alcohol consumption in rats and whether alcohol intake is associated with delta FosB expression in the brain reward circuit. Potentially, chronic sleep restriction could make the brain vulnerable and consequently promote addictive behavior. Therefore, the present study's objective was to evaluate alcohol consumption in a chronic sleep restriction model and determine the expression of delta FosB in brains of adult rats. For this purpose, male Wistar rats (300-350 g body weight) were divided into four experimental groups (n = 6 each group): control (without manipulation), sleep restriction (SR) for 7 days, SR and ethanol exposure (Ethanol + SR), and a group with just ethanol exposure (Ethanol). At the end of the management, rats were sacrificed, and the brains were dissected and processed for immunohistochemical detection of delta FosB. The results showed that SR stimulates alcohol consumption compared to unrestricted-sleep rats and induces a significant increase in the number of delta FosB-positive cells in brain nuclei within the motivation/brain reward circuit. These results suggest that chronic reduction of sleep hours is a risk factor for developing a preference for alcohol consumption.
Collapse
Affiliation(s)
- Fabio García-García
- Biomedicine Department, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico.
| | - Sergio Priego-Fernández
- Health Sciences Program, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico
| | - Luis Angel López-Muciño
- Health Sciences Program, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico
| | | | - Carolina Peña-Escudero
- Health Sciences Program, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico
| |
Collapse
|
24
|
Scaplen KM, Petruccelli E. Receptors and Channels Associated with Alcohol Use: Contributions from Drosophila. Neurosci Insights 2021; 16:26331055211007441. [PMID: 33870197 PMCID: PMC8020223 DOI: 10.1177/26331055211007441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022] Open
Abstract
Alcohol Use Disorder (AUD) is a debilitating disorder that manifests as problematic patterns of alcohol use. At the core of AUD's behavioral manifestations are the profound structural, physiological, cellular, and molecular effects of alcohol on the brain. While the field has made considerable progress in understanding the neuromolecular targets of alcohol we still lack a comprehensive understanding of alcohol's actions and effective treatment strategies. Drosophila melanogaster is a powerful model for investigating the neuromolecular targets of alcohol because flies model many of the core behavioral elements of AUD and offer a rich genetic toolkit to precisely reveal the in vivo molecular actions of alcohol. In this review, we focus on receptors and channels that are often targeted by alcohol within the brain. We discuss the general roles of these proteins, their role in alcohol-associated behaviors across species, and propose ways in which Drosophila models can help advance the field.
Collapse
Affiliation(s)
- Kristin M Scaplen
- Department of Psychology, Bryant University, Smithfield, RI, USA
- Center for Health and Behavioral Studies, Bryant University, Smithfield, RI, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Emily Petruccelli
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
25
|
Bauer MR, McVey MM, Boehm SL. Three Weeks of Binge Alcohol Drinking Generates Increased Alcohol Front-Loading and Robust Compulsive-Like Alcohol Drinking in Male and Female C57BL/6J Mice. Alcohol Clin Exp Res 2021; 45:650-660. [PMID: 33496972 DOI: 10.1111/acer.14563] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Current models of compulsive-like quinine-adulterated alcohol (QuA) drinking in mice, if improved, could be more useful for uncovering the neural mechanisms of compulsive-like alcohol drinking. The purpose of these experiments was to further characterize and improve the validity of a model of compulsive-like QuA drinking in C57BL/6J mice. We sought to determine whether compulsive-like alcohol drinking could be achieved following 2 or 3 weeks of Drinking-in-the-Dark (DID), whether it provides evidence for a robust model of compulsive-like alcohol drinking by inclusion of a water control group and use of a highly concentrated QuA solution, whether repeated QuA exposures alter compulsive-like drinking, and whether there are sex differences in compulsive-like alcohol drinking. METHODS Male and Female C57BL/6J mice were allowed free access to either 20% alcohol or tap water for 2 hours each day for approximately 3 weeks. After 2 or 3 weeks, the mice were given QuA (500 μM) and the effect of repeated QuA drinking sessions on compulsive-like alcohol drinking was assessed. 3-minute front-loading, 2 hour binge-drinking, and blood alcohol concentrations were determined. RESULTS Compulsive-like QuA drinking was achieved after 3 weeks, but not 2 weeks, of daily alcohol access as determined by alcohol history mice consuming significantly more QuA than water history mice and drinking statistically nondifferent amounts of QuA than nonadulterated alcohol at baseline. Thirty-minute front-loading of QuA revealed that alcohol history mice front-loaded significantly more QuA than water history mice, but still found the QuA solution aversive. Repeated QuA exposures did not alter these patterns, compulsive-like drinking did not differ by sex, and BACs for QuA drinking were at the level of a binge. CONCLUSIONS These data suggest that compulsive-like QuA drinking can be robustly achieved following 3 weeks of DID and male and female C57BL/6J mice do not differ in compulsive-like alcohol drinking.
Collapse
Affiliation(s)
- Meredith R Bauer
- Department of Psychology, Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Megan M McVey
- Department of Psychology, Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Stephen L Boehm
- Department of Psychology, Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
26
|
Ehinger Y, Morisot N, Phamluong K, Sakhai SA, Soneja D, Adrover MF, Alvarez VA, Ron D. cAMP-Fyn signaling in the dorsomedial striatum direct pathway drives excessive alcohol use. Neuropsychopharmacology 2021; 46:334-342. [PMID: 32417851 PMCID: PMC7852539 DOI: 10.1038/s41386-020-0712-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
Fyn kinase in the dorsomedial striatum (DMS) of rodents plays a central role in mechanisms underlying excessive alcohol intake. The DMS is comprised of medium spiny neurons (MSNs) that project directly (dMSNs) or indirectly (iMSNs) to the substantia nigra. Here, we examined the cell-type specificity of Fyn's actions in alcohol use. First, we knocked down Fyn selectively in DMS dMSNs or iMSNs of mice and measured the level of alcohol consumption. We found that downregulation of Fyn in dMSNs, but not in iMSNs, reduces excessive alcohol but not saccharin intake. D1Rs are coupled to Gαs/olf, which activate cAMP signaling. To examine whether Fyn's actions are mediated through cAMP signaling, DMS dMSNs were infected with GαsDREADD, and the activation of Fyn signaling was measured following CNO treatment. We found that remote stimulation of cAMP signaling in DMS dMSNs activates Fyn and promotes the phosphorylation of the Fyn substrate, GluN2B. In contract, remote activation of GαsDREADD in DLS dMSNs did not alter Fyn signaling. We then tested whether activation of GαsDREADD in DMS dMSNs or iMSNs alters alcohol intake and observed that CNO-dependent activation of GαsDREADD in DMS dMSNs but not iMSNs increases alcohol but not saccharin intake. Finally, we examined the contribution of Fyn to GαsDREADD-dependent increase in alcohol intake, and found that systemic administration of the Fyn inhibitor, AZD0503 blocks GαsDREADD-dependent increase in alcohol consumption. Our results suggest that the cAMP-Fyn axis in the DMS dMSNs is a molecular transducer of mechanisms underlying the development of excessive alcohol consumption.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Nadege Morisot
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Nkarta Therapeutics, San Francisco, CA, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Sage Therapeutics, San Francisco, CA, USA
| | - Drishti Soneja
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Martin F Adrover
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- INGEBI, CONICET, Buenos Aires, Argentina
| | - Veronica A Alvarez
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health, Bethesda, MD, 20892, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA.
| |
Collapse
|
27
|
Region-dependent regulation of acute ethanol on γ oscillation in the rat hippocampal slices. Psychopharmacology (Berl) 2020; 237:2959-2966. [PMID: 32700022 DOI: 10.1007/s00213-020-05584-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/10/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Ethanol use disorders are a serious medical and public health problem in the world today. Acute ethanol intoxication can lead to cognitive dysfunction such as learning and memory impairment. Gamma oscillations (γ, 30-80 Hz) are synchronized rhythmic activity generated by population of neurons within local network, and closely related to learning and memory function. The hippocampus is a critical anatomic structure that supports learning and memory. On the grounds of structure and function, hippocampus can be divided into the intermediate (IH), the dorsal (DH), and ventral hippocampus (VH). The current study is the first to investigate the effects of acute ethanol on γ oscillations in these sub-regions of rat hippocampal slices. METHODS The sustained γ oscillations were induced by 200 nM kainate (KA) in the CA3c of IH, DH, and VH. When KA-induced γ oscillation reached the steady state, ethanol (50 mM or 100 mM) was applied and the effects of ethanol on γ oscillation power was measured in the slices sequentially sectioned from ventral to dorsal hippocampus of adult rats. RESULTS In the intermediate hippocampal slices, compared with control (KA only), ethanol (50 mM) caused 36.1 ± 3.9% decrease in γ power (p < 0.05, n = 10), while ethanol (100 mM) caused 55.3 ± 5.5% decrease in γ power (p < 0.001, n = 14). In the dorsal hippocampus, only ethanol (100 mM) caused 18.1 ± 8.6% decrease in γ power (p < 0.05, n = 12). However, in the ventral hippocampus, neither 50 mM nor 100 mM ethanol affected γ oscillation. CONCLUSIONS Our results demonstrate that ethanol may produce the differential suppression of γ oscillations in a dose-dependent manner in different sub-regions of hippocampus, suggesting that the modulation of ethanol on hippocampal γ oscillation is region-dependent.
Collapse
|