1
|
Mitten EH, Souders A, Marron Fernandez de Velasco E, Aguado C, Luján R, Wickman K. Chronic ethanol exposure in mice evokes pre- and postsynaptic deficits in GABAergic transmission in ventral tegmental area GABA neurons. Br J Pharmacol 2024. [PMID: 39358985 DOI: 10.1111/bph.17335] [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: 03/14/2024] [Revised: 06/26/2024] [Accepted: 07/18/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND AND PURPOSE GABAergic neurons in mouse ventral tegmental area (VTA) exhibit elevated activity during withdrawal following chronic ethanol exposure. While increased glutamatergic input and decreased GABAA receptor sensitivity have been implicated, the impact of inhibitory signaling in VTA GABA neurons has not been fully addressed. EXPERIMENTAL APPROACH We used electrophysiological and ultrastructural approaches to assess the impact of chronic intermittent ethanol vapour exposure in mice on GABAergic transmission in VTA GABA neurons during withdrawal. We used CRISPR/Cas9 ablation to mimic a somatodendritic adaptation involving the GABAB receptor (GABABR) in ethanol-naïve mice to investigate its impact on anxiety-related behaviour. KEY RESULTS The frequency of spontaneous inhibitory postsynaptic currents was reduced in VTA GABA neurons following chronic ethanol treatment and this was reversed by GABABR inhibition, suggesting chronic ethanol strengthens the GABABR-dependent suppression of GABAergic input to VTA GABA neurons. Similarly, paired-pulse depression of GABAA receptor-dependent responses evoked by optogenetic stimulation of nucleus accumbens inputs from ethanol-treated mice was reversed by GABABR inhibition. Somatodendritic currents evoked in VTA GABA neurons by GABABR activation were reduced following ethanol exposure, attributable to the suppression of GIRK (Kir3) channel activity. Mimicking this adaptation enhanced anxiety-related behaviour in ethanol-naïve mice. CONCLUSIONS AND IMPLICATIONS Chronic ethanol weakens the GABAergic regulation of VTA GABA neurons in mice via pre- and postsynaptic mechanisms, likely contributing to their elevated activity during withdrawal and expression of anxiety-related behaviour. As anxiety can promote relapse during abstinence, interventions targeting VTA GABA neuron excitability could represent new therapeutic strategies for treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Eric H Mitten
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anna Souders
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Carolina Aguado
- Instituto de Biomedicina de la UCLM (IB-UCLM), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, Albacete, Spain
| | - Rafael Luján
- Instituto de Biomedicina de la UCLM (IB-UCLM), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, Albacete, Spain
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
2
|
Lyu Z, Gong Z, Huang M, Xin S, Zou M, Ding Y. Benefits of exercise on cognitive impairment in alcohol use disorder following alcohol withdrawal. FEBS Open Bio 2024; 14:1540-1558. [PMID: 39054261 PMCID: PMC11492329 DOI: 10.1002/2211-5463.13865] [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: 10/10/2023] [Revised: 06/21/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Although most cognitive impairments induced by prolonged alcohol consumption tend to improve within the initial months of abstinence, there is evidence suggesting certain cognitive deficits may persist. This study aimed to investigate the impact of aerobic exercise on learning and memory in alcohol use disorder (AUD) mice following a period of abstinence from alcohol. We also sought to assess the levels of monoamine neurotransmitters in the hippocampus. To this end, we established an AUD mouse model through a two-bottle choice (sucrose fading mode and normal mode) and chronic intermittent alcohol vapor (combined with intraperitoneal injection) and randomly allocated mice into exercise groups to undergo treadmill training. Learning and memory abilities were assessed through the Morris water maze test and spontaneous activity was evaluated using the open field test. The levels of dopamine, norepinephrine, serotonin, and brain-derived neurotrophic factor in the hippocampus were quantified using enzyme-linked immunoassay (ELISA) kits. The findings reveal that after cessation of alcohol consumption, learning and memory abilities in AUD mice did not completely return to normal levels. The observed enhancement of cognitive functions in AUD mice through aerobic exercise may be attributed to restoring levels of monoamine neurotransmitters in the hippocampus, boosting brain-derived neurotrophic factor (BDNF) concentrations, and facilitating an increase in hippocampal mass. These results offer empirical evidence to support aerobic exercise as a viable therapeutic strategy to alleviate cognitive deficits associated with AUD.
Collapse
Affiliation(s)
- Zhen Lyu
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
- School of PsychologyShanghai University of SportChina
| | - Zhi‐Gang Gong
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| | - Min‐Xia Huang
- Science and Technology College of Nanchang Hangkong UniversityJiujiangChina
| | - Si‐Ping Xin
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| | - Mao‐Zhong Zou
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| | - Yu‐Quan Ding
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| |
Collapse
|
3
|
Lapish CC. Understanding How Acute Alcohol Impacts Neural Encoding in the Rodent Brain. Curr Top Behav Neurosci 2024. [PMID: 38858298 DOI: 10.1007/7854_2024_479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Alcohol impacts neural circuitry throughout the brain and has wide-ranging effects on the biophysical properties of neurons in these circuits. Articulating how these wide-ranging effects might eventually result in altered computational properties has the potential to provide a tractable working model of how alcohol alters neural encoding. This chapter reviews what is currently known about how acute alcohol influences neural activity in cortical, hippocampal, and dopaminergic circuits as these have been the primary focus of understanding how alcohol alters neural computation. While other neural systems have been the focus of exhaustive work on this topic, these brain regions are the ones where in vivo neural recordings are available, thus optimally suited to make the link between changes in neural activity and behavior. Rodent models have been key in developing an understanding of how alcohol impacts the function of these circuits, and this chapter therefore focuses on work from mice and rats. While progress has been made, it is critical to understand the challenges and caveats associated with experimental procedures, especially when performed in vivo, which are designed to answer this question and if/how to translate these data to humans. The hypothesis is discussed that alcohol impairs the ability of neural circuits to acquire states of neural activity that are transiently elevated and characterized by increased complexity. It is hypothesized that these changes are distinct from the traditional view of alcohol being a depressant of neural activity in the forebrain.
Collapse
Affiliation(s)
- Christopher C Lapish
- Department of Anatomy, Cell Biology, and Physiology, Stark Neuroscience Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
4
|
Rahamim N, Liran M, Aronovici C, Flumin H, Gordon T, Urshansky N, Barak S. Inhibition of ERK1/2 or CRMP2 Disrupts Alcohol Memory Reconsolidation and Prevents Relapse in Rats. Int J Mol Sci 2024; 25:5478. [PMID: 38791516 PMCID: PMC11122309 DOI: 10.3390/ijms25105478] [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: 02/22/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Relapse to alcohol abuse, often caused by cue-induced alcohol craving, is a major challenge in alcohol addiction treatment. Therefore, disrupting the cue-alcohol memories can suppress relapse. Upon retrieval, memories transiently destabilize before they reconsolidate in a process that requires protein synthesis. Evidence suggests that the mammalian target of rapamycin complex 1 (mTORC1), governing the translation of a subset of dendritic proteins, is crucial for memory reconsolidation. Here, we explored the involvement of two regulatory pathways of mTORC1, phosphoinositide 3-kinase (PI3K)-AKT and extracellular regulated kinase 1/2 (ERK1/2), in the reconsolidation process in a rat (Wistar) model of alcohol self-administration. We found that retrieval of alcohol memories using an odor-taste cue increased ERK1/2 activation in the amygdala, while the PI3K-AKT pathway remained unaffected. Importantly, ERK1/2 inhibition after alcohol memory retrieval impaired alcohol-memory reconsolidation and led to long-lasting relapse suppression. Attenuation of relapse was also induced by post-retrieval administration of lacosamide, an inhibitor of collapsin response mediator protein-2 (CRMP2)-a translational product of mTORC1. Together, our findings indicate the crucial role of ERK1/2 and CRMP2 in the reconsolidation of alcohol memories, with their inhibition as potential treatment targets for relapse prevention.
Collapse
Affiliation(s)
- Nofar Rahamim
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Mirit Liran
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
- Faculty of Life Sciences, Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Coral Aronovici
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Hila Flumin
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Tamar Gordon
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Nataly Urshansky
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Segev Barak
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
- Faculty of Life Sciences, Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
5
|
Montgomery SE, Li L, Russo SJ, Calipari ES, Nestler EJ, Morel C, Han MH. Mesolimbic Neural Response Dynamics Predict Future Individual Alcohol Drinking in Mice. Biol Psychiatry 2024; 95:951-962. [PMID: 38061466 DOI: 10.1016/j.biopsych.2023.11.019] [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: 03/22/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Individual variability in response to rewarding stimuli is a striking but understudied phenomenon. The mesolimbic dopamine system is critical in encoding the reinforcing properties of both natural reward and alcohol; however, how innate or baseline differences in the response dynamics of this circuit define individual behavior and shape future vulnerability to alcohol remain unknown. METHODS Using naturalistic behavioral assays, a voluntary alcohol drinking paradigm, in vivo fiber photometry, in vivo electrophysiology, and chemogenetics, we investigated how differences in mesolimbic neural circuit activity contribute to the individual variability seen in reward processing and, by proxy, alcohol drinking. RESULTS We first characterized heterogeneous behavioral and neural responses to natural reward and defined how these baseline responses predicted future individual alcohol-drinking phenotypes in male mice. We then determined spontaneous ventral tegmental area dopamine neuron firing profiles associated with responses to natural reward that predicted alcohol drinking. Using a dual chemogenetic approach, we mimicked specific mesolimbic dopamine neuron firing activity before or during voluntary alcohol drinking to link unique neurophysiological profiles to individual phenotype. We show that hyperdopaminergic individuals exhibit a lower neuronal response to both natural reward and alcohol that predicts lower levels of alcohol consumption in the future. CONCLUSIONS These findings reveal unique, circuit-specific neural signatures that predict future individual vulnerability or resistance to alcohol and expand the current knowledge base on how some individuals are able to titrate their alcohol consumption whereas others go on to engage in unhealthy alcohol-drinking behaviors.
Collapse
Affiliation(s)
- Sarah E Montgomery
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Long Li
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erin S Calipari
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Departments of Pharmacology, Molecular Physiology and Biophysics, and Psychiatry and Behavioral Sciences, Vanderbilt Center for Addiction Research, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carole Morel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Ming-Hu Han
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
6
|
Fu Y, Li W, Mai Y, Guan J, Ding R, Hou J, Chen B, Cao G, Sun S, Tang Y, Fu R. Association between RMTg Neuropeptide Genes and Negative Effect during Alcohol Withdrawal in Mice. Int J Mol Sci 2024; 25:2933. [PMID: 38474180 DOI: 10.3390/ijms25052933] [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: 12/18/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Alcohol use disorders (AUDs) frequently co-occur with negative mood disorders, such as anxiety and depression, exacerbating relapse through dopaminergic dysfunction. Stress-related neuropeptides play a crucial role in AUD pathophysiology by modulating dopamine (DA) function. The rostromedial tegmental nucleus (RMTg), which inhibits midbrain dopamine neurons and signals aversion, has been shown to increase ethanol consumption and negative emotional states during abstinence. Despite some stress-related neuropeptides acting through the RMTg to affect addiction behaviors, their specific roles in alcohol-induced contexts remain underexplored. This study utilized an intermittent voluntary drinking model in mice to induce negative effect behavior 24 h into ethanol (EtOH) abstinence (post-EtOH). It examined changes in pro-stress (Pnoc, Oxt, Npy) and anti-stress (Crf, Pomc, Avp, Orx, Pdyn) neuropeptide-coding genes and analyzed their correlations with aversive behaviors. We observed that adult male C57BL/6J mice displayed evident anxiety, anhedonia, and depression-like symptoms at 24 h post-EtOH. The laser-capture microdissection technique, coupled with or without retrograde tracing, was used to harvest total ventral tegmental area (VTA)-projecting neurons or the intact RMTg area. The findings revealed that post-EtOH consistently reduced Pnoc and Orx levels while elevating Crf levels in these neuronal populations. Notably, RMTg Pnoc and Npy levels counteracted ethanol consumption and depression severity, while Crf levels were indicative of the mice's anxiety levels. Together, these results underscore the potential role of stress-related neuropeptides in the RMTg in regulating the negative emotions related to AUDs, offering novel insights for future research.
Collapse
Affiliation(s)
- Yixin Fu
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Yunlin Mai
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Junhao Guan
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Ruxuan Ding
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Jiawei Hou
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Bingqing Chen
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Guoxin Cao
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Shizhu Sun
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Ying Tang
- Clinical Skills Training Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| |
Collapse
|
7
|
Becker HC, Lopez MF. Animal Models of Excessive Alcohol Consumption in Rodents. Curr Top Behav Neurosci 2024. [PMID: 38340255 DOI: 10.1007/7854_2024_461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
The development of animal models that demonstrate excessive levels of alcohol consumption has played an important role in advancing our knowledge about neurobiological underpinnings and environmental circumstances that engender such maladaptive behavior. The use of these preclinical models has also provided valuable opportunities for discovering new and novel therapeutic targets that may be useful in the treatment of alcohol use disorder (AUD). While no single model can fully capture the complexities of AUD, the goal is to develop animal models that closely approximate characteristics of heavy alcohol drinking in humans to enhance their translational value and utility. A variety of experimental approaches have been employed to produce the desired phenotype of interest-robust and reliable excessive levels of alcohol drinking. Here we provide an updated review of five animal models that are commonly used. The models entail procedural manipulations of scheduled access to alcohol (time of day, duration, frequency), periods of time when access to alcohol is withheld, and history of alcohol exposure. Specially, the models involve (a) scheduled access to alcohol, (b) scheduled periods of alcohol deprivation, (c) scheduled intermittent access to alcohol, (d) scheduled-induced polydipsia, and (e) chronic alcohol (dependence) and withdrawal experience. Each of the animal models possesses unique experimental features that engender excessive levels of alcohol consumption. Both advantages and disadvantages of each model are described along with discussion of future work to be considered in developing more optimal models. Ultimately, the validity and utility of these models will lie in their ability to aid in the discovery of new and novel potential therapeutic targets as well as serve as a platform to evaluate treatment strategies that effectively reduce excessive levels of alcohol consumption associated with AUD.
Collapse
Affiliation(s)
- Howard C Becker
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, USA.
- Departments of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
- Departments of Psychiatry and Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- RHJ Veterans Administration Health Care System, Medical University of South Carolina, Charleston, SC, USA.
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, USA
- Departments of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
8
|
Healey K, Waters RC, Knight SG, Wandling GM, Hall NI, Jones BN, Shobande MJ, Melton JG, Pandey SC, Scott Swartzwelder H, Maldonado-Devincci AM. Adolescent intermittent ethanol exposure alters adult exploratory and affective behaviors, and cerebellar Grin2b expression in C57BL/6J mice. Drug Alcohol Depend 2023; 253:111026. [PMID: 38006668 PMCID: PMC10990063 DOI: 10.1016/j.drugalcdep.2023.111026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 09/17/2023] [Accepted: 11/01/2023] [Indexed: 11/27/2023]
Abstract
Binge drinking is one of the most common patterns (more than 90%) of alcohol consumption by young people. During adolescence, the brain undergoes maturational changes that influence behavioral control and affective behaviors, such as cerebellar brain volume and function in adulthood. We investigated long-term impacts of adolescent binge ethanol exposure on affective and exploratory behaviors and cerebellar gene expression in adult male and female mice. Further, the cerebellum is increasingly recognized as a brain region integrating a multitude of behaviors that span from the traditional primary sensory-motor to affective functions, such as anxiety and stress reactivity. Therefore, we investigated the persistent effects of adolescent intermittent ethanol (AIE) on exploratory and affective behaviors and began to elucidate the role of the cerebellum in these behaviors through excitatory signaling gene expression. We exposed C57BL/6J mice to AIE or air (control) vapor inhalation from postnatal day 28-42. After prolonged abstinence (>34 days), in young adulthood (PND 77+) we assessed behavior in the open field, light/dark, tail suspension, and forced swim stress tests to determine changes in affective behaviors including anxiety-like, depressive-like, and stress reactivity behavior. Excitatory signaling gene mRNA levels of fragile X messenger ribonucleoprotein (FMR1), glutamate receptors (Grin2a, Grin2b and Grm5) and excitatory synaptic markers (PSD-95 and Eaat1) were measured in the cerebellum of adult control and AIE-exposed mice. AIE-exposed mice showed decreased exploratory behaviors in the open field test (OFT) where both sexes show reduced ambulation, however only females exhibited a reduction in rearing. Additionally, in the OFT, AIE-exposed females also exhibited increased anxiety-like behavior (entries to center zone). In the forced swim stress test, AIE-exposed male mice, but not females, spent less time immobile compared to their same-sex controls, indicative of sex-specific changes in stress reactivity. Male and female AIE-exposed mice showed increased Grin2b (Glutamate Ionotropic Receptor NMDA Type Subunit 2B) mRNA levels in the cerebellum compared to their same-sex controls. Together, these data show that adolescent binge-like ethanol exposure altered both exploratory and affective behaviors in a sex-specific manner and modified cerebellar Grin2b expression in adult mice. This indicates the cerebellum may serve as an important brain region that is susceptible to long-term molecular changes after AIE.
Collapse
Affiliation(s)
- Kati Healey
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, 323 Foster St., Durham, NC 27701, United States
| | - Renee C Waters
- Department of Psychology, Hairston College of Health and Human Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States; Department of Psychology, Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540, United States
| | - Sherilynn G Knight
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Gabriela M Wandling
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois, Chicago, IL, United States
| | - Nzia I Hall
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States; University of North Carolina at Chapel Hill School of Medicine, NC 27516, United States
| | - Brooke N Jones
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Mariah J Shobande
- Department of Chemical, Biological and Bioengineering, College of Engineering, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Jaela G Melton
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois, Chicago, IL, United States; Jesse Brown VA Medical Center, Chicago, IL, United States
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, 323 Foster St., Durham, NC 27701, United States
| | - Antoniette M Maldonado-Devincci
- Department of Psychology, Hairston College of Health and Human Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States.
| |
Collapse
|
9
|
Baker EJ, Moore S, Gonzales SW, Grant KA. Long-term drinking stability in the open-access self-administration monkey model. Alcohol 2023; 113:41-48. [PMID: 37516372 PMCID: PMC10818025 DOI: 10.1016/j.alcohol.2023.07.002] [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: 03/30/2023] [Revised: 06/14/2023] [Accepted: 07/05/2023] [Indexed: 07/31/2023]
Abstract
The Non-Human Primate (NHP) model for the study of Alcohol Use Disorders (AUD) as developed in our laboratories is critical to our understanding of the pathophysiology of voluntary, chronic, ethanol consumption. Previous work in this model established categories of ethanol consumption that parallel reported categories of human consumption across a spectrum spanning low drinking, binge drinking, heavy drinking, and very heavy drinking, albeit at generally higher daily intakes across categories than documented in people. Original categories assigned to ethanol consumption patterns were established using a limited cohort of rhesus macaques. This study revisits the validity of categorical drinking using an additional 28 monkeys. In addition to finding categorical representations consistent with the original 2014 report, our findings demonstrate that drinking categories remain stable across the observed 12 months of nearly consistent access to ethanol (22 h/day), termed "open access". Animals occupying the two ends of the spectrum, "low" and "very heavy" drinkers, exhibit the largest stability. The findings also indicate a slight escalatory drift over time, with very heavy drinking animals experiencing fatigue near the end of open access.
Collapse
Affiliation(s)
- Erich J Baker
- Department of Computer Science, Baylor University, Waco, TX, USA.
| | - Sharon Moore
- Department of Computer Science, Baylor University, Waco, TX, USA
| | - Steven W Gonzales
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
10
|
Healey K, Waters RC, Knight SG, Wandling GM, Hall NI, Jones BN, Shobande MJ, Melton JG, Pandey SC, Scott Swartzwelder H, Maldonado-Devincci AM. Adolescent Intermittent Ethanol Exposure Alters Adult Exploratory and Affective Behaviors, and Cerebellar Grin2B Expression in C57BL/6J Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528396. [PMID: 36824954 PMCID: PMC9949091 DOI: 10.1101/2023.02.13.528396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Binge drinking is one of the most common patterns (more than 90%) of alcohol consumption by young people. During adolescence, the brain undergoes maturational changes that influence behavioral control and affective behaviors, such as cerebellar brain volume and function in adulthood. We investigated long-term impacts of adolescent binge ethanol exposure on affective and exploratory behaviors and cerebellar gene expression in adult male and female mice. Further, the cerebellum is increasingly recognized as a brain region integrating a multitude of behaviors that span from the traditional primary sensory-motor to affective functions, such as anxiety and stress reactivity. Therefore, we investigated the persistent effects of adolescent intermittent ethanol (AIE) on exploratory and affective behaviors and began to elucidate the role of the cerebellum in these behaviors through excitatory signaling gene expression. We exposed C57BL/6J mice to AIE or air (control) vapor inhalation from postnatal day 28-42. After prolonged abstinence (>34 days), in young adulthood (PND 77+) we assessed behavior in the open field, light/dark, tail suspension, and forced swim stress tests to determine changes in affective behaviors including anxiety-like, depressive-like, and stress reactivity behavior. Excitatory signaling gene mRNA levels of fragile X messenger ribonucleoprotein ( FMR1) , glutamate receptors ( Grin2a , Grin2B and Grm5 ) and excitatory synaptic markers (PSD-95 and Eaat1) were measured in the cerebellum of adult control and AIE-exposed mice. AIE-exposed mice showed decreased exploratory behaviors in the open field test (OFT) where both sexes show reduced ambulation, however only females exhibited a reduction in rearing. Additionally, in the OFT, AIE-exposed females also exhibited increased anxiety-like behavior (entries to center zone). In the forced swim stress test, AIE-exposed male mice, but not females, spent less time immobile compared to their same-sex controls, indicative of sex-specific changes in stress reactivity. Male and female AIE-exposed mice showed increased Grin2B (Glutamate Ionotropic Receptor NMDA Type Subunit 2B) mRNA levels in the cerebellum compared to their same-sex controls. Together, these data show that adolescent binge-like ethanol exposure altered both exploratory and affective behaviors in a sex-specific manner and modified cerebellar Grin2B expression in adult mice. This indicates the cerebellum may serve as an important brain region that is susceptible to long-term molecular changes after AIE. Highlights Adolescent intermittent ethanol (AIE) exposure decreased exploratory behavior in adult male and female mice.In females, but not males, AIE increased anxiety-like behavior.In males, but not females, AIE reduced stress reactivity in adulthood.These findings indicate sex differences in the enduring effects of AIE on exploratory and affective behaviors. Cerebellar Grin2B mRNA levels were increased in adulthood in both male and female AIE-exposed mice. These findings add to the small, but growing literature on behavioral AIE effects in mice, and establish cerebellar excitatory synaptic gene expression as an enduring effect of adolescent ethanol exposure.
Collapse
|
11
|
Wills TA. Unveiling New Brain Circuits and Network Activity in Alcohol Use Disorder: Insights From Innovative Technologies. Biol Psychiatry 2023; 94:365-366. [PMID: 37558313 DOI: 10.1016/j.biopsych.2023.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 08/11/2023]
Affiliation(s)
- Tiffany A Wills
- Department of Anatomy and Cell Biology, Neuroscience Center of Excellence and Alcohol and Drug Abuse Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana.
| |
Collapse
|
12
|
Borgonetti V, Roberts AJ, Bajo M, Galeotti N, Roberto M. Chronic alcohol induced mechanical allodynia by promoting neuroinflammation: A mouse model of alcohol-evoked neuropathic pain. Br J Pharmacol 2023; 180:2377-2392. [PMID: 37050867 PMCID: PMC10898491 DOI: 10.1111/bph.16091] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic pain is considered a key factor contributing to alcohol use disorder (AUD). The mechanisms responsible for chronic pain associated with chronic alcohol consumption are unknown. We evaluated the development of chronic pain in a mouse model of alcohol dependence and investigate the role of neuroinflammation. EXPERIMENTAL APPROACH The chronic-intermittent ethanol two-bottle choice CIE-2BC paradigm generates three groups: alcohol-dependent with escalating alcohol intake, nondependent (moderate drinking) and alcohol-naïve control male and female mice. We measured mechanical allodynia during withdrawal and after the last voluntary drinking. Immunoblotting was used to evaluate the protein levels of IBA-1, CSFR, IL-6, p38 and ERK2/1 in spinal cord tissue of dependent and non-dependent animals. KEY RESULTS We found significant escalation of drinking in the dependent group in male and female compared with the non-dependent group. The dependent group developed mechanical allodynia during 72 h of withdrawal, which was completely reversed after voluntary drinking. We observed an increased pain hypersensitivity compared with the naïve in 50% of non-dependent group. Increased IBA-1 and CSFR expression was observed in spinal cord tissue of both hypersensitivity-abstinence related and neuropathy-alcohol mice, and increased IL-6 expression and ERK1/2 activation in mice with hypersensitivity-related to abstinence, but not in mice with alcohol-evoked neuropathic pain. CONCLUSIONS AND IMPLICATIONS The CIE-2BC model induces two distinct pain conditions specific to the type of ethanol exposure: abstinence-related hypersensitivity in dependent mice and alcohol-evoked neuropathic pain in about a half of the non-dependent mice.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, Florence, 50139, Italy
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Amanda J. Roberts
- Animal Models Core, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Michal Bajo
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, Florence, 50139, Italy
| | - Marisa Roberto
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
13
|
Xiao T, Chen Y, Boisvert A, Cole M, Kimbrough A. Chronic Intermittent Ethanol Vapor Exposure Paired with Two-Bottle Choice to Model Alcohol Use Disorder. J Vis Exp 2023:10.3791/65320. [PMID: 37427930 PMCID: PMC11164185 DOI: 10.3791/65320] [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] [Indexed: 07/11/2023] Open
Abstract
Alcohol use disorder (AUD) is a chronic alcohol-related disorder that typically presents as uncontrolled drinking and preoccupation with alcohol. A key component of AUD research is using translationally relevant preclinical models. Over the past several decades, a variety of animal models have been used to study AUD. One prominent model of AUD is the chronic intermittent ethanol vapor exposure (CIE) model, which is a well-established approach for inducing alcohol dependence in rodents through repeated cycles of ethanol exposure via inhalation. To model AUD in mice, the CIE exposure is paired with a voluntary two-bottle choice (2BC) of alcohol drinking and water to measure the escalation of alcohol drinking. The 2BC/CIE procedure involves alternating weeks of 2BC drinking and CIE, which repeat until the escalation of alcohol drinking is achieved. In the present study, we outline the procedures for performing 2BC/CIE, including the daily use of the CIE vapor chamber, and provide an example of escalated alcohol drinking in C57BL/6J mice using this approach.
Collapse
Affiliation(s)
- Tiange Xiao
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University
| | - Yueyi Chen
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University
| | - Alyssa Boisvert
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University
| | | | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University; Purdue Institute for Integrative Neuroscience; Weldon School of Biomedical Engineering, Purdue University; Purdue Institute of Inflammation, Immunology, and Infectious Disease;
| |
Collapse
|
14
|
Brewton HW, Robinson SL, Thiele TE. Astrocyte expression in the extended amygdala of C57BL/6J mice is sex-dependently affected by chronic intermittent and binge-like ethanol exposure. Alcohol 2023; 108:55-64. [PMID: 36539069 PMCID: PMC10033386 DOI: 10.1016/j.alcohol.2022.12.001] [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: 09/07/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Excessive ethanol drinking is a major problem within the United States, causing alterations in brain plasticity and neurocognitive function. Astrocytes are glial cells that regulate neurosynaptic plasticity, modulate neurochemicals, and monitor other homeostatic roles. Astrocytes have been found to play a part in modulating excessive ethanol consumption. The basolateral amygdala (BLA), central amygdala (CeA), and bed nucleus of the stria terminalis (BNST) are brain regions that process stress, anxiety, and reward; they are also implicated in modulating ethanol intake. Little is understood, however, about how astrocyte expression in each region is modulated by chronic and binge-like ethanol drinking patterns. In the present report, we utilized two separate animal models of excessive drinking: chronic intermittent ethanol (CIE) and "Drinking-in-the-dark" (DID). Following these paradigms, animal brains were processed through immunohistochemistry (IHC) and stained for glial fibrillary acidic protein (GFAP). Collected data illustrated a sex-dependent relationship between ethanol intake and GFAP immunoreactivity (IR) in the BLA and BNST, but not in the CeA. Specifically, CIE and DID ethanol drinking resulted in blunted GFAP-IR (specifically via GFAP-positive cell count) in the BLA and BNST, particularly in males. These findings may implicate sex-dependent ethanol-induced changes in BLA and BNST astrocytes, providing a potential therapeutic target for anxiety and stress disorders.
Collapse
Affiliation(s)
- Honoreé W Brewton
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, North Carolina, 27599-3270, United States
| | - Stacey L Robinson
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, North Carolina, 27599-3270, United States; The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599-3270, United States
| | - Todd E Thiele
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, North Carolina, 27599-3270, United States; The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599-3270, United States.
| |
Collapse
|
15
|
Griffin WC, Lopez MF, Woodward JJ, Becker HC. Alcohol dependence and the ventral hippocampal influence on alcohol drinking in male mice. Alcohol 2023; 106:44-54. [PMID: 36328184 PMCID: PMC9868110 DOI: 10.1016/j.alcohol.2022.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/26/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2022]
Abstract
Examining neural circuits underlying persistent, heavy drinking provides insight into the neurobiological mechanisms driving alcohol use disorder. Facilitated by its connectivity with other parts of the brain such as the nucleus accumbens (NAc), the ventral hippocampus (vHC) supports many behaviors, including those related to reward seeking and addiction. These studies used a well-established mouse model of alcohol (ethanol) dependence. After surgery to infuse DREADD-expressing viruses (hM4Di, hM3Dq, or mCherry-only) into the vHC and position guide cannula above the NAc, male C57BL/6J mice were treated in the CIE drinking model that involved repeated cycles of chronic intermittent alcohol (CIE) vapor or air (CTL) exposure alternating with weekly test drinking cycles in which mice were offered alcohol (15% v/v) 2 h/day. Additionally, smaller groups of mice were evaluated for either cFos expression or glutamate release using microdialysis procedures. In CIE mice expressing inhibitory (hM4Di) DREADDs in the vHC, drinking increased as expected, but CNO (3 mg/kg intraperitoneally [i.p.]) given 30 min before testing did not alter alcohol intake. However, in CTL mice expressing hM4Di, CNO significantly increased alcohol drinking (∼30%; p < 0.05) to levels similar to the CIE mice. The vHC-NAc pathway was targeted by infusing CNO into the NAc (3 or 10 μM/side) 30 min before testing. CNO activation of the pathway in mice expressing excitatory (hM3Dq) DREADDs selectively reduced consumption in CIE mice back to CTL levels (∼35-45%; p < 0.05) without affecting CTL alcohol intake. Lastly, activating the vHC-NAc pathway increased cFos expression and evoked significant glutamate release from the vHC terminals in the NAc. These data indicate that reduced activity of the vHC increases alcohol consumption and that targeted, increased activity of the vHC-NAc pathway attenuates excessive drinking associated with alcohol dependence. Thus, these findings indicate that the vHC and its glutamatergic projections to the NAc are involved in excessive alcohol drinking.
Collapse
Affiliation(s)
- William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States.
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States
| | - John J Woodward
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC 29425-0742, United States
| |
Collapse
|
16
|
Domi E, Barchiesi R, Barbier E. Epigenetic Dysregulation in Alcohol-Associated Behaviors: Preclinical and Clinical Evidence. Curr Top Behav Neurosci 2023. [PMID: 36717533 DOI: 10.1007/7854_2022_410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Alcohol use disorder (AUD) is characterized by loss of control over intake and drinking despite harmful consequences. At a molecular level, AUD is associated with long-term neuroadaptations in key brain regions that are involved in reward processing and decision-making. Over the last decades, a great effort has been made to understand the neurobiological basis underlying AUD. Epigenetic mechanisms have emerged as an important mechanism in the regulation of long-term alcohol-induced gene expression changes. Here, we review the literature supporting a role for epigenetic processes in AUD. We particularly focused on the three most studied epigenetic mechanisms: DNA methylation, Histone modification and non-coding RNAs. Clinical studies indicate an association between AUD and DNA methylation both at the gene and global levels. Using behavioral paradigms that mimic some of the characteristics of AUD, preclinical studies demonstrate that changes in epigenetic mechanisms can functionally impact alcohol-associated behaviors. While many studies support a therapeutic potential for targeting epigenetic enzymes, more research is needed to fully understand their role in AUD. Identification of brain circuits underlying alcohol-associated behaviors has made major advances in recent years. However, there are very few studies that investigate how epigenetic mechanisms can affect these circuits or impact the neuronal ensembles that promote alcohol-associated behaviors. Studies that focus on the role of circuit-specific and cell-specific epigenetic changes for clinically relevant alcohol behaviors may provide new insights on the functional role of epigenetic processes in AUD.
Collapse
Affiliation(s)
- Esi Domi
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- School of Pharmacy, Pharmacology Unit, Center for Neuroscience, University of Camerino, Camerino, Italy
| | - Riccardo Barchiesi
- Department of Neuroscience, Waggoner Center for Alcohol and Alcohol Addiction Research, University of Texas at Austin, Austin, TX, USA
| | - Estelle Barbier
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden.
| |
Collapse
|
17
|
Althobaiti YS. Investigating the potential of mirtazapine to induce drug-seeking behavior in free-choice drinking mouse model. Saudi Pharm J 2022; 30:1809-1815. [PMID: 36601513 PMCID: PMC9805978 DOI: 10.1016/j.jsps.2022.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/26/2022] [Indexed: 11/05/2022] Open
Abstract
Addiction to various drugs and chemicals is a significant public health concern worldwide. Addiction to prescription medications has increased due to the psychoactive effects of these medications, their availability, low price, and the lack of legal consequences for abusers. One of such prescription medication is mirtazapine (MIRT). MIRT is an antidepressant that has recently been reported to be abused and could induce withdrawal symptoms in different case studies. No previous study has investigated its abuse potential in animal models of drug addiction. Here, we conducted a free-choice drinking paradigm to investigate voluntary drinking of MIRT at two different concentrations. Male BALB/c mice were given unlimited access to two water bottles for five days before being divided into three groups: the first group had free access to two water bottles. The second group (MIRT10) and the third group (MIRT20) was allowed unlimited choice to one bottle of water and one bottle of MIRT at concentrations of 0.03 and 0.06 mg/mL, respectively. The average daily MIRT intake in the MIRT20 group was significantly higher on all tested days than that in the MIRT10 group. Moreover, mice in the MIRT20 group preferred to self-administer MIRT over water, indicating that MIRT can induce drug-seeking behavior. To further investigate the addictive potential of MIRT and its possible deterioration of memory and recognition, as reported with several known drugs of abuse, animals underwent a novel object recognition test. Mice in the MIRT20 group demonstrated significant deterioration in memory and recognition, indicating its effects on different brain regions involved in recognition, similar to other known drugs of abuse. The forced swimming test and tail suspension test were used to test MIRT-induced withdrawal symptoms after forced abstinence. After eight days of abstinence, mice in the MIRT20 group demonstrated significant depression-like symptoms in both the TST and FST, manifested by a significant increase in immobility time. MIRT was shown to induce drug-seeking behavior, deteriorate recognition, and cause withdrawal symptoms. This might confirm that MIRT has the potential to induce drug dependence and further studies are warranted to explore the neurobiological basis of MIRT-induced drug-seeking behavior.
Collapse
Affiliation(s)
- Yusuf S. Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia,Addiction and Neuroscience Research Unit, Taif University, Taif, Saudi Arabia
| |
Collapse
|
18
|
Bloch S, Holleran KM, Kash TL, Vazey EM, Rinker JA, Lebonville CL, O'Hara K, Lopez MF, Jones SR, Grant KA, Becker HC, Mulholland PJ. Assessing negative affect in mice during abstinence from alcohol drinking: Limitations and future challenges. Alcohol 2022; 100:41-56. [PMID: 35181404 PMCID: PMC8983487 DOI: 10.1016/j.alcohol.2022.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/09/2023]
Abstract
Alcohol use disorder (AUD) is frequently comorbid with mood disorders, and these co-occurring neuropsychiatric disorders contribute to the development and maintenance of alcohol dependence and relapse. In preclinical models, mice chronically exposed to alcohol display anxiety-like and depressive-like behaviors during acute withdrawal and protracted abstinence. However, in total, results from studies using voluntary alcohol-drinking paradigms show variable behavioral outcomes in assays measuring negative affective behaviors. Thus, the main objective of this review is to summarize the literature on the variability of negative affective behaviors in mice after chronic alcohol exposure. We compare the behavioral phenotypes that emerge during abstinence across different exposure models, including models of alcohol and stress interactions. The complicated outcomes from these studies highlight the difficulties of assessing negative affective behaviors in mouse models designed for the study of AUD. We discuss new behavioral assays, comprehensive platforms, and unbiased machine-learning algorithms as promising approaches to better understand the interaction between alcohol and negative affect in mice. New data-driven approaches in the understanding of mouse behavior hold promise for improving the identification of mechanisms, cell subtypes, and neurocircuits that mediate negative affect. In turn, improving our understanding of the neurobehavioral basis of alcohol-associated negative affect will provide a platform to test hypotheses in mouse models that aim to improve the development of more effective strategies for treating individuals with AUD and co-occurring mood disorders.
Collapse
Affiliation(s)
- Solal Bloch
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Jennifer A Rinker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Christina L Lebonville
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Krysten O'Hara
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Marcelo F Lopez
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States
| | - Howard C Becker
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States.
| |
Collapse
|
19
|
de Biedma-Elduayen LG, Giménez-Gómez P, Morales-Puerto N, Vidal R, de la Calle CN, Gutiérrez-López MD, O'Shea E, Colado MI. Influx of kynurenine into the brain is involved in the reduction of ethanol consumption induced by Ro 61-8048 after chronic intermittent ethanol in mice. Br J Pharmacol 2022; 179:3711-3726. [PMID: 35189673 PMCID: PMC9314579 DOI: 10.1111/bph.15825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/07/2022] [Accepted: 02/10/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The kynurenine pathway has been proposed as a new target for modulating drug abuse. We previously demonstrated that inhibition of kynurenine 3-monooxygenase (KMO) using Ro 61-8048 reduces ethanol consumption in a binge drinking model. Here we investigate the effect of the kynurenine pathway modulation in ethanol -dependent mice. EXPERIMENTAL APPROACH Adult male and female mice were subjected to the Chronic Intermittent Ethanol (CIE) paradigm. On the last day of CIE, mice were treated with Ro 61-8048, Ro 61-8048 + PNU-120596, a positive allosteric modulator of α7nAChR, and Ro 61-8048 + L-leucine or probenecid, which block the influx or efflux of kynurenine from the brain, respectively. Ethanol, water consumption and preference were measured and kynurenine levels in plasma and limbic forebrain were determined. KEY RESULTS Ro 61-8048 decreases consumption and preference for ethanol in both sexes exposed to the CIE model, an effect that is prevented by PNU-120596. The Ro 61-8048-induced decrease in ethanol consumption depends on the influx of kynurenine into the brain. CONCLUSION AND IMPLICATIONS Inhibition of KMO reduces ethanol consumption and preference in both male and female mice subjected to CIE model by a mechanism involving α7nAChR. Moreover, the effect which is mediated centrally depends on the influx of peripheral kynurenine to the brain and can be prolonged by blocking the efflux of kynurenine from the brain. Here, for the first time we demonstrate that the modulation of the kynurenine pathway is a valid strategy for the treatment of ethanol dependence in both sexes.
Collapse
Affiliation(s)
- Leticia Gil de Biedma-Elduayen
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Current address: University of Massachusetts Chan Medical School, The Brudnick Neuropsychiatric Research Institute, Worcester, MA
| | - Nuria Morales-Puerto
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Rebeca Vidal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Carlos Núñez de la Calle
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María Dolores Gutiérrez-López
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
20
|
Simmons KE, Healey KL, Li Q, Moore SD, Klein RC. Effects of sex and genotype in human APOE-targeted replacement mice on alcohol self-administration measured with the automated IntelliCage system before and after repeated mild traumatic brain injury. Alcohol Clin Exp Res 2021; 45:2231-2245. [PMID: 34585391 DOI: 10.1111/acer.14717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Few studies have examined the association between APOE genotype and alcohol use. Although some of these studies have reported outcomes associated with a history of drinking, none have examined alcohol-seeking behavior. In addition, no preclinical studies have examined alcohol use as a function of APOE genotype with or without traumatic brain injury. METHODS Male and female human APOE3- and APOE4-targeted replacement (TR) mice were used to assess voluntary alcohol seeking longitudinally using a 2-bottle choice paradigm conducted within the automated IntelliCage system prior to and following repeated mild TBI (rmTBI). Following an acquisition phase in which the concentration of ethanol (EtOH) was increased to 12%, a variety of drinking paradigms that included extended alcohol access (EAA1 and EAA2), alcohol deprivation effect (ADE), limited access drinking in the dark (DID), and progressive ratio (PR) were used to assess alcohol-seeking behavior. Additional behavioral tasks were performed to measure cognitive function and anxiety-like behavior. RESULTS All groups readily consumed increasing concentrations of EtOH (4-12%) during the acquisition phase. During the EAA1 period (12% EtOH), there was a significant genotype effect in both males and females for EtOH preference. Following a 3-week abstinence period, mice received sham or rmTBI resulting in a genotype- and sex-independent main effect of rmTBI on the recovery of righting reflex and a main effect of rmTBI on spontaneous home-cage activity in females only. Reintroduction of 12% EtOH (EAA2) resulted in a significant effect genotype for alcohol preference in males with APOE4 mice displaying increased preference and motivation for alcohol compared with APOE3 mice independent of TBI while in females, there was a significant genotype × TBI interaction under the ADE and DID paradigms. Finally, there was a main effect of rmTBI on increased risk-seeking behavior in both sexes, but no effect on spatial learning or cognitive flexibility. CONCLUSION These results suggest that sex and APOE genotype play a significant role in alcohol consumption and may subsequently influence long-term recovery following traumatic brain insults.
Collapse
Affiliation(s)
- Kathryn E Simmons
- Trinity College of Arts and Sciences, Duke University, Durham, North Carolina, USA
| | - Kati L Healey
- Department of Psychiatry, Duke University Medical Center, Durham, North Carolina, USA
| | - Qiang Li
- Department of Psychiatry, Duke University Medical Center, Durham, North Carolina, USA
| | - Scott D Moore
- Department of Psychiatry, Duke University Medical Center, Durham, North Carolina, USA.,Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Rebecca C Klein
- Department of Psychiatry, Duke University Medical Center, Durham, North Carolina, USA.,Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| |
Collapse
|
21
|
Effects of ceftriaxone on ethanol drinking and GLT-1 expression in ethanol dependence and relapse drinking. Alcohol 2021; 92:1-9. [PMID: 33465464 DOI: 10.1016/j.alcohol.2021.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/11/2020] [Accepted: 01/12/2021] [Indexed: 12/26/2022]
Abstract
Repeated cycles of chronic intermittent ethanol (CIE) exposure increase voluntary consumption of alcohol (ethanol) in mice. Previous reports from our laboratory show that CIE increases extracellular glutamate in the nucleus accumbens (NAc) and that manipulating accumbal glutamate concentrations will alter ethanol drinking, indicating that glutamate homeostasis plays a crucial role in ethanol drinking in this model. A number of studies have shown that ceftriaxone increases GLT-1 expression, the major glutamate transporter, and that treatment with this antibiotic reduces ethanol drinking. The present studies examined the effects of ceftriaxone on ethanol drinking and GLT-1 in a mouse model of ethanol dependence and relapse drinking. The results show that ceftriaxone did not influence drinking at any dose in either ethanol-dependent or non-dependent mice. Further, ceftriaxone did not increase GLT-1 expression in the accumbens core or shell, with the exception of the ethanol-dependent mice receiving the highest dose of ceftriaxone. Interestingly, ethanol-dependent mice treated with only vehicle displayed reduced expression of GLT-1 in the accumbens shell and of the presynaptic mGlu2 receptor in the accumbens core. The reduced expression of the major glutamate transporter (GLT-1), as well as a receptor that regulates glutamate release (mGlu2), may help explain, at least in part, increased glutamatergic transmission in this model of ethanol dependence and relapse drinking.
Collapse
|
22
|
Sharma L, Sharma A, Dash AK, Bisht GS, Gupta GL. A standardized polyherbal preparation POL-6 diminishes alcohol withdrawal anxiety by regulating Gabra1, Gabra2, Gabra3, Gabra4, Gabra5 gene expression of GABA A receptor signaling pathway in rats. BMC Complement Med Ther 2021; 21:13. [PMID: 33407346 PMCID: PMC7789136 DOI: 10.1186/s12906-020-03181-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/07/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Alcohol abuse is a major problem worldwide and it affects people's health and economy. There is a relapse in alcohol intake due to alcohol withdrawal. Alcohol withdrawal anxiety-like behavior is a symptom that appears 6-24 h after the last alcohol ingestion. METHODS The present study was designed to explore the protective effect of a standardized polyherbal preparation POL-6 in ethanol withdrawal anxiety in Wistar rats. POL-6 was prepared by mixing the dried extracts of six plants Bacopa monnieri, Hypericum perforatum, Centella asiatica, Withania somnifera, Camellia sinesis, and Ocimum sanctum in the proportion 2:1:2:2:1:2 respectively. POL-6 was subjected to phytochemical profiling through LC-MS, HPLC, and HPTLC. The effect of POL-6 on alcohol withdrawal anxiety was tested using a two-bottle choice drinking paradigm model giving animals' free choice between alcohol and water for 15 days. Alcohol was withdrawn on the 16th day and POL-6 (20, 50, and 100 mg/kg, oral), diazepam (2 mg/kg) treatment was given on the withdrawal days. Behavioral parameters were tested using EPM and LDT. On the 18th day blood was collected from the retro-orbital sinus of the rats and alcohol markers ALT, AST, ALP, and GGT were studied. At end of the study, animals were sacrificed and the brain was isolated for exploring the influences of POL-6 on the mRNA expression of GABAA receptor subunits in the amygdala and hippocampus. RESULTS Phytochemical profiling showed that POL-6 contains major phytoconstituents like withaferin A, quercetin, catechin, rutin, caeffic acid, and β-sitosterol. In-vivo studies showed that POL-6 possesses an antianxiety effect in alcohol withdrawal. Gene expression studies on the isolated brain tissues showed that POL-6 normalizes the GABAergic transmission in the amygdala and hippocampus of the rats. CONCLUSION The study concludes that POL-6 may have therapeutic potential for treating ethanol-type dependence.
Collapse
Affiliation(s)
- Lalit Sharma
- Department of Pharmacy, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Ashutosh Kumar Dash
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Gopal Singh Bisht
- Department of BT/BI, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Girdhari Lal Gupta
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, Shri Vile Parle Kelavani Mandal's Narsee Monjee Institute of Management Studies University, Mumbai, Maharashtra, 400056, India.
- School of Pharmacy & Technology Management, Shri Vile Parle Kelavani Mandal's Narsee Monjee Institute of Management Studies, Shirpur Campus, Shirpur, Maharashtra, 425405, India.
| |
Collapse
|
23
|
Somkuwar SS, Villalpando EG, Quach LW, Head BP, McKenna BS, Scadeng M, Mandyam CD. Abstinence from ethanol dependence produces concomitant cortical gray matter abnormalities, microstructural deficits and cognitive dysfunction. Eur Neuropsychopharmacol 2021; 42:22-34. [PMID: 33279357 PMCID: PMC7797163 DOI: 10.1016/j.euroneuro.2020.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/12/2020] [Accepted: 11/07/2020] [Indexed: 12/20/2022]
Abstract
Previous studies demonstrate that ethanol dependence induced by repeating cycles of chronic intermittent ethanol vapor exposure (CIE) followed by protracted abstinence (CIE-PA) produces significant alterations in oligodendrogenesis in the rodent medial prefrontal cortex (mPFC). Specifically, CIE-PA produced an unprecedented increase in premyelinating oligodendroglial progenitor cells and myelin, which have been associated with persistent elevated drinking behaviors during abstinence. The current study used neuroimaging and electron microscopy to evaluate the integrity of enhanced myelin and microstructural deficits underlying enhanced myelination in the mPFC in male rats experiencing forced abstinence for 1 day (D), 7D, 21D and 42D following seven weeks of CIE. In vivo diffusion tensor imaging (DTI) detected altered microstructural integrity in the mPFC and corpus callosum (CC). Altered integrity was characterized as reduced fractional anisotropy (FA) in the CC, and enhanced mean diffusivity (MD) in the mPFC in 7D abstinent rats. Increased MD occurred concomitantly with increases in myelin associated proteins, flayed myelin and enhanced mitochondrial stress in the mPFC in 7D abstinent rats, suggesting that the increases in myelination during abstinence was aberrant. Evaluation of cognitive performance via Pavlovian conditioning in 7D abstinent rats revealed reduced retrieval and recall of fear memories dependent on the mPFC. These findings indicate that forced abstinence from moderate to severe alcohol use disorder produces gray matter damage via myelin dysfunction in the mPFC and that these microstructural changes were associated with deficits in PFC dependent behaviors.
Collapse
Affiliation(s)
| | | | - Leon W Quach
- VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Brian P Head
- VA San Diego Healthcare System, San Diego, CA 92161, USA; Departments of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Benjamin S McKenna
- Departments of Psychiatry, University of California San Diego, San Diego, CA 92161, USA
| | - Miriam Scadeng
- Departments of Radiology, University of California San Diego, San Diego, CA 92161, USA
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA 92161, USA; Departments of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA.
| |
Collapse
|
24
|
Smith RJ, Anderson RI, Haun HL, Mulholland PJ, Griffin WC, Lopez MF, Becker HC. Dynamic c-Fos changes in mouse brain during acute and protracted withdrawal from chronic intermittent ethanol exposure and relapse drinking. Addict Biol 2020; 25:e12804. [PMID: 31288295 PMCID: PMC7579841 DOI: 10.1111/adb.12804] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 01/05/2023]
Abstract
Alcohol dependence promotes neuroadaptations in numerous brain areas, leading to escalated drinking and enhanced relapse vulnerability. We previously developed a mouse model of ethanol dependence and relapse drinking in which repeated cycles of chronic intermittent ethanol (CIE) vapor exposure drive a significant escalation of voluntary ethanol drinking. In the current study, we used this model to evaluate changes in neuronal activity (as indexed by c‐Fos expression) throughout acute and protracted withdrawal from CIE (combined with or without a history of ethanol drinking). We analyzed c‐Fos protein expression in 29 brain regions in mice sacrificed 2, 10, 26, and 74 hours or 7 days after withdrawal from 5 cycles of CIE. Results revealed dynamic time‐ and brain region‐dependent changes in c‐Fos activity over the time course of withdrawal from CIE exposure, as compared with nondependent air‐exposed control mice, beginning with markedly low expression levels upon removal from the ethanol vapor chambers (2 hours), reflecting intoxication. c‐Fos expression was enhanced during acute CIE withdrawal (10 and 26 hours), followed by widespread reductions at the beginning of protracted withdrawal (74 hours) in several brain areas. Persistent reductions in c‐Fos expression were observed during prolonged withdrawal (7 days) in prelimbic cortex, nucleus accumbens shell, dorsomedial striatum, paraventricular nucleus of thalamus, and ventral subiculum. A history of ethanol drinking altered acute CIE withdrawal effects and caused widespread reductions in c‐Fos that persisted during extended abstinence even without CIE exposure. These data indicate that ethanol dependence and relapse drinking drive long‐lasting neuroadaptations in several brain regions.
Collapse
Affiliation(s)
- Rachel J. Smith
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
| | - Rachel I. Anderson
- Department of Psychiatry and Behavioral Sciences Medical University of South Carolina Charleston SC USA
| | - Harold L. Haun
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
| | - Patrick J. Mulholland
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
- Department of Psychiatry and Behavioral Sciences Medical University of South Carolina Charleston SC USA
- Charleston Alcohol Research Center Medical University of South Carolina Charleston SC USA
| | - William C. Griffin
- Department of Psychiatry and Behavioral Sciences Medical University of South Carolina Charleston SC USA
- Charleston Alcohol Research Center Medical University of South Carolina Charleston SC USA
| | - Marcelo F. Lopez
- Department of Psychiatry and Behavioral Sciences Medical University of South Carolina Charleston SC USA
- Charleston Alcohol Research Center Medical University of South Carolina Charleston SC USA
| | - Howard C. Becker
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
- Department of Psychiatry and Behavioral Sciences Medical University of South Carolina Charleston SC USA
- Charleston Alcohol Research Center Medical University of South Carolina Charleston SC USA
- Ralph H. Johnson Veteran Affairs Medical Center Medical University of South Carolina Charleston SC USA
| |
Collapse
|
25
|
The infralimbic cortex and mGlu5 mediate the effects of chronic intermittent ethanol exposure on fear learning and memory. Psychopharmacology (Berl) 2020; 237:3417-3433. [PMID: 32767063 PMCID: PMC7572878 DOI: 10.1007/s00213-020-05622-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/27/2020] [Indexed: 01/19/2023]
Abstract
RATIONALE AND OBJECTIVES Alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD) often occur comorbidly. While the incidence of these disorders is increasing, there is little investigation into the interacting neural mechanisms between these disorders. These studies aim to identify cognitive deficits that occur as a consequence of fear and ethanol exposure, implement a novel pharmaceutical intervention, and determine relevant underlying neurocircuitry. Additionally, due to clinical sex differences in PTSD prevalence and alcohol abuse, these studies examine the nature of this relationship in rodent models. METHODS Animals were exposed to a model of PTSD+AUD using auditory fear conditioning followed by chronic intermittent ethanol exposure (CIE). Then, rats received extinction training consisting of multiple conditioned stimulus presentations in absence of the shock. Extinction recall and context-induced freezing were measured in subsequent tests. CDPPB, a metabotropic glutamate receptor 5 (mGlu5) positive allosteric modulator, was used to treat these deficits, and region-specific effects were determined using microinjections. RESULTS These studies determined that CIE exposure led to deficits in fear extinction learning and heightened context-induced freezing while sex differences emerged in fear conditioning and extinction cue recall tests. Furthermore, using CDPPB, these studies found that enhancement of infralimbic (IfL) mGlu5 activity was able to recover CIE-induced deficits in both males and females. CONCLUSIONS These studies show that CIE induces deficits in fear-related behaviors and that enhancement of IfL glutamatergic activity can facilitate learning during extinction. Additionally, we identify novel pharmacological targets for the treatment of individuals who suffer from PTSD and AUD.
Collapse
|
26
|
Johnson KA, Lovinger DM. Allosteric modulation of metabotropic glutamate receptors in alcohol use disorder: Insights from preclinical investigations. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 88:193-232. [PMID: 32416868 DOI: 10.1016/bs.apha.2020.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are family C G protein-coupled receptors (GPCRs) that modulate neuronal excitability and synaptic transmission throughout the nervous system. Owing to recent advances in development of subtype-selective allosteric modulators of mGlu receptors, individual members of the mGlu receptor family have been proposed as targets for treating a variety of neurological and psychiatric disorders, including substance use disorders. In this chapter, we highlight preclinical evidence that allosteric modulators of mGlu receptors could be useful for reducing alcohol consumption and preventing relapse in alcohol use disorder (AUD). We begin with an overview of the preclinical models that are used to study mGlu receptor involvement in alcohol-related behaviors. Alcohol exposure causes adaptations in both expression and function of various mGlu receptor subtypes, and pharmacotherapies aimed at reversing these adaptations have the potential to reduce alcohol consumption and seeking. Positive allosteric modulators (PAMs) of mGlu2 and negative allosteric modulators of mGlu5 show particular promise for reducing alcohol intake and/or preventing relapse. Finally, this chapter discusses important considerations for translating preclinical findings toward the development of clinically useful drugs, including the potential for PAMs to avoid tolerance issues that are frequently observed with repeated administration of GPCR agonists.
Collapse
Affiliation(s)
- Kari A Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
27
|
Morisot N, Phamluong K, Ehinger Y, Berger AL, Moffat JJ, Ron D. mTORC1 in the orbitofrontal cortex promotes habitual alcohol seeking. eLife 2019; 8:51333. [PMID: 31820733 PMCID: PMC6959998 DOI: 10.7554/elife.51333] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) plays an important role in dendritic translation and in learning and memory. We previously showed that heavy alcohol use activates mTORC1 in the orbitofrontal cortex (OFC) of rodents (Laguesse et al., 2017a). Here, we set out to determine the consequences of alcohol-dependent mTORC1 activation in the OFC. We found that inhibition of mTORC1 activity in the OFC attenuates alcohol seeking and restores sensitivity to outcome devaluation in rats that habitually seek alcohol. In contrast, habitual responding for sucrose was unaltered by mTORC1 inhibition, suggesting that mTORC1’s role in habitual behavior is specific to alcohol. We further show that inhibition of GluN2B in the OFC attenuates alcohol-dependent mTORC1 activation, alcohol seeking and habitual responding for alcohol. Together, these data suggest that the GluN2B/mTORC1 axis in the OFC drives alcohol seeking and habit.
Collapse
Affiliation(s)
- Nadege Morisot
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Khanhky Phamluong
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Yann Ehinger
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Anthony L Berger
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Jeffrey J Moffat
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
28
|
Kuhn BN, Kalivas PW, Bobadilla AC. Understanding Addiction Using Animal Models. Front Behav Neurosci 2019; 13:262. [PMID: 31849622 PMCID: PMC6895146 DOI: 10.3389/fnbeh.2019.00262] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/13/2022] Open
Abstract
Drug addiction is a neuropsychiatric disorder with grave personal consequences that has an extraordinary global economic impact. Despite decades of research, the options available to treat addiction are often ineffective because our rudimentary understanding of drug-induced pathology in brain circuits and synaptic physiology inhibits the rational design of successful therapies. This understanding will arise first from animal models of addiction where experimentation at the level of circuits and molecular biology is possible. We will review the most common preclinical models of addictive behavior and discuss the advantages and disadvantages of each. This includes non-contingent models in which animals are passively exposed to rewarding substances, as well as widely used contingent models such as drug self-administration and relapse. For the latter, we elaborate on the different ways of mimicking craving and relapse, which include using acute stress, drug administration or exposure to cues and contexts previously paired with drug self-administration. We further describe paradigms where drug-taking is challenged by alternative rewards, such as appetitive foods or social interaction. In an attempt to better model the individual vulnerability to drug abuse that characterizes human addiction, the field has also established preclinical paradigms in which drug-induced behaviors are ranked by various criteria of drug use in the presence of negative consequences. Separation of more vulnerable animals according to these criteria, along with other innate predispositions including goal- or sign-tracking, sensation-seeking behavior or impulsivity, has established individual genetic susceptibilities to developing drug addiction and relapse vulnerability. We further examine current models of behavioral addictions such as gambling, a disorder included in the DSM-5, and exercise, mentioned in the DSM-5 but not included yet due to insufficient peer-reviewed evidence. Finally, after reviewing the face validity of the aforementioned models, we consider the most common standardized tests used by pharmaceutical companies to assess the addictive potential of a drug during clinical trials.
Collapse
Affiliation(s)
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Ana-Clara Bobadilla
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
29
|
Bosse KE, Chiu VM, Lloyd SC, Conti AC. Neonatal alcohol exposure augments voluntary ethanol intake in the absence of potentiated anxiety-like behavior induced by chronic intermittent ethanol vapor exposure. Alcohol 2019; 79:17-24. [PMID: 30385201 DOI: 10.1016/j.alcohol.2018.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/12/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022]
Abstract
Individuals fetally exposed to alcohol have a disproportionate risk for developing lifetime alcohol dependence, an association that may be confounded by the presence of comorbid conditions, such as anxiety. Anxiety is also observed following fetal alcohol exposure and is known to exacerbate ethanol consumption, highlighting the utility of animal models to assess this relationship. The present study evaluated the impact of third-trimester equivalent ethanol exposure on ethanol consumption and anxiety-like, marble burying behavior in adult, male C57BL/6 mice following exposure to chronic intermittent ethanol vapor, proposed to model dependence. Neonatal mice (P5-6, 2.5-3.0 g) were administered one injection of saline or ethanol (2.5 g/kg, subcutaneously [s.c.]). Pre-vapor marble burying and limited-access two-bottle choice ethanol intake (15% v/v, 2 h) were comparable in adults (8 weeks of age) across neonatal treatment groups. Five consecutive drinking sessions were repeated 72 h after each weekly ethanol vapor exposure procedure for a total of five vapor/drinking cycles. Consistent with prior research, an increase in voluntary ethanol drinking was observed in vapor-exposed, neonatal saline-treated mice throughout the study starting after the second vapor cycle compared to both air-exposed control groups. In neonatal ethanol-treated mice, this increase in ethanol intake and preference following vapor exposure was accelerated, being observed after the first vapor cycle, and observed at an augmented level compared to vapor-exposed, neonatal saline-treated mice and air controls for both neonatal conditions. Conversely, marble burying was enhanced equivalently in vapor-exposed mice from either neonatal treatment group relative to their respective air-exposed controls. These data recapitulate clinical observations of enhanced sensitivity for alcohol dependence following developmental alcohol exposure, which may reflect enhanced motivational drive rather than potentiated negative affect. The present model will facilitate the future exploration of mechanisms that underlie increased risk for alcohol use after early developmental exposure.
Collapse
Affiliation(s)
- K E Bosse
- Research & Development Service, John D. Dingell VA Medical Center, 4646 John R St., Detroit, MI, 48201, United States; Department of Neurosurgery, Wayne State University School of Medicine, 4160 John R St., Detroit, MI, 48201, United States
| | - V M Chiu
- Research & Development Service, John D. Dingell VA Medical Center, 4646 John R St., Detroit, MI, 48201, United States; Department of Neurosurgery, Wayne State University School of Medicine, 4160 John R St., Detroit, MI, 48201, United States
| | - S C Lloyd
- Research & Development Service, John D. Dingell VA Medical Center, 4646 John R St., Detroit, MI, 48201, United States; Department of Neurosurgery, Wayne State University School of Medicine, 4160 John R St., Detroit, MI, 48201, United States
| | - A C Conti
- Research & Development Service, John D. Dingell VA Medical Center, 4646 John R St., Detroit, MI, 48201, United States; Department of Neurosurgery, Wayne State University School of Medicine, 4160 John R St., Detroit, MI, 48201, United States.
| |
Collapse
|
30
|
Erickson EK, Blednov YA, Harris RA, Mayfield RD. Glial gene networks associated with alcohol dependence. Sci Rep 2019; 9:10949. [PMID: 31358844 PMCID: PMC6662804 DOI: 10.1038/s41598-019-47454-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic alcohol abuse alters the molecular structure and function of brain cells. Recent work suggests adaptations made by glial cells, such as astrocytes and microglia, regulate physiological and behavioral changes associated with addiction. Defining how alcohol dependence alters the transcriptome of different cell types is critical for developing the mechanistic hypotheses necessary for a nuanced understanding of cellular signaling in the alcohol-dependent brain. We performed RNA-sequencing on total homogenate and glial cell populations isolated from mouse prefrontal cortex (PFC) following chronic intermittent ethanol vapor exposure (CIE). Compared with total homogenate, we observed unique and robust gene expression changes in astrocytes and microglia in response to CIE. Gene co-expression network analysis revealed biological pathways and hub genes associated with CIE in astrocytes and microglia that may regulate alcohol-dependent phenotypes. Astrocyte identity and synaptic calcium signaling genes were enriched in alcohol-associated astrocyte networks, while TGF-β signaling and inflammatory response genes were disrupted by CIE treatment in microglia gene networks. Genes related to innate immune signaling, specifically interferon pathways, were consistently up-regulated across CIE-exposed astrocytes, microglia, and total homogenate PFC tissue. This study illuminates the cell-specific effects of chronic alcohol exposure and provides novel molecular targets for studying alcohol dependence.
Collapse
Affiliation(s)
- Emma K Erickson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA.
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| |
Collapse
|
31
|
Yu W, Hwa LS, Makhijani VH, Besheer J, Kash TL. Chronic inflammatory pain drives alcohol drinking in a sex-dependent manner for C57BL/6J mice. Alcohol 2019; 77:135-145. [PMID: 30300665 DOI: 10.1016/j.alcohol.2018.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/01/2018] [Accepted: 10/01/2018] [Indexed: 12/31/2022]
Abstract
Sex differences in chronic pain and alcohol abuse are not well understood. The development of rodent models is imperative for investigating the underlying changes behind these pathological states. In the present study, we investigated whether hind paw treatment with the inflammatory agent Complete Freund's Adjuvant (CFA) could generate hyperalgesia and alter alcohol consumption in male and female C57BL/6J mice. CFA treatment led to greater nociceptive sensitivity for both sexes in the Hargreaves test, and increased alcohol drinking for males in a continuous-access two-bottle choice (CA2BC) paradigm. Regardless of treatment, female mice exhibited greater alcohol drinking than males. Following a 2-h terminal drinking session, CFA treatment failed to produce changes in alcohol drinking, blood ethanol concentration (BEC), and plasma corticosterone (CORT) for both sexes. Two-hour alcohol consumption and CORT was higher in females than males, regardless of CFA treatment. Taken together, these findings have established that male mice are more susceptible to escalations in alcohol drinking when undergoing pain, despite higher levels of total alcohol drinking and CORT in females. Furthermore, the exposure of CFA-treated C57BL/6J mice to the CA2BC drinking paradigm has proven to be a useful model for studying the relationship between chronic pain and alcohol abuse. Future applications of the CFA/CA2BC model should incorporate manipulations of stress signaling and other related biological systems to improve our mechanistic understanding of pain and alcohol interactions.
Collapse
|
32
|
Ethanol Experience Enhances Glutamatergic Ventral Hippocampal Inputs to D1 Receptor-Expressing Medium Spiny Neurons in the Nucleus Accumbens Shell. J Neurosci 2019; 39:2459-2469. [PMID: 30692226 DOI: 10.1523/jneurosci.3051-18.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/11/2019] [Accepted: 01/15/2019] [Indexed: 02/05/2023] Open
Abstract
A growing number of studies implicate alterations in glutamatergic signaling within the reward circuitry of the brain during alcohol abuse and dependence. A key integrator of glutamatergic signaling in the reward circuit is the nucleus accumbens, more specifically, the dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) within this region, which have been implicated in the formation of dependence to many drugs of abuse including alcohol. D1-MSNs receive glutamatergic input from several brain regions; however, it is not currently known how individual inputs onto D1-MSNs are altered by alcohol experience. Here, we investigate input-specific adaptations in glutamatergic transmission in response to varying levels of alcohol experience. Virally mediated expression of Channelrhodopsin in ventral hippocampal (vHipp) glutamate neurons of male mice allowed for selective activation of vHipp to D1-MSN synapses. Therefore, we were able to compare synaptic adaptations in response to low and high alcohol experience in vitro and in vivo Alcohol experience enhanced glutamatergic activity and abolished LTD at vHipp to D1-MSN synapses. Following chronic alcohol experience, GluA2-lacking AMPARs, which are Ca permeable, were inserted into vHipp to D1-MSN synapses. These findings support the reversal of alcohol-induced insertion of Ca-permeable AMPARs and the enhancement of glutamatergic activity at vHipp to D1-MSNs as potential targets for intervention during early exposure to alcohol.SIGNIFICANCE STATEMENT Given the roles of the nucleus accumbens (NAc) in integrating cortical and allocortical information and in reward learning, it is vital to understand how inputs to this region are altered by drugs of abuse such as alcohol. The strength of excitatory inputs from the ventral hippocampus (vHipp) to the NAc has been positively associated with reward-related behaviors, but it is unclear whether or how ethanol affects these inputs. Here we show that vHipp-NAc synapses indeed are altered by ethanol exposure, with vHipp glutamatergic input to the NAc being enhanced following chronic ethanol experience. This work provides insight into ethanol-induced alterations of vHipp-NAc synapses and suggests that, similarly to drugs such as cocaine, the strengthening of these synapses promotes reward behavior.
Collapse
|
33
|
Ziv Y, Rahamim N, Lezmy N, Even-Chen O, Shaham O, Malishkevich A, Giladi E, Elkon R, Gozes I, Barak S. Activity-dependent neuroprotective protein (ADNP) is an alcohol-responsive gene and negative regulator of alcohol consumption in female mice. Neuropsychopharmacology 2019; 44:415-424. [PMID: 30008470 PMCID: PMC6300527 DOI: 10.1038/s41386-018-0132-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 12/16/2022]
Abstract
Neuroadaptations in the brain reward system caused by excessive alcohol intake, lead to drinking escalation and alcohol use disorder phenotypes. Activity-dependent neuroprotective protein (ADNP) is crucial for brain development, and is implicated in neural plasticity in adulthood. Here, we discovered that alcohol exposure regulates Adnp expression in the mesolimbic system, and that Adnp keeps alcohol drinking in moderation, in a sex-dependent manner. Specifically, Sub-chronic alcohol treatment (2.5 g/kg/day for 7 days) increased Adnp mRNA levels in the dorsal hippocampus in both sexes, and in the nucleus accumbens of female mice, 24 h after the last alcohol injection. Long-term voluntary consumption of excessive alcohol quantities (~10-15 g/kg/24 h, 5 weeks) increased Adnp mRNA in the hippocampus of male mice immediately after an alcohol-drinking session, but the level returned to baseline after 24 h of withdrawal. In contrast, excessive alcohol consumption in females led to long-lasting reduction in hippocampal Adnp expression. We further tested the regulatory role of Adnp in alcohol consumption, using the Adnp haploinsufficient mouse model. We found that Adnp haploinsufficient female mice showed higher alcohol consumption and preference, compared to Adnp intact females, whereas no genotype difference was observed in males. Importantly, daily intranasal administration of the ADNP-snippet drug candidate NAP normalized alcohol consumption in Adnp haploinsufficient females. Finally, female Adnp haploinsufficient mice showed a sharp increase in alcohol intake after abstinence, suggesting that Adnp protects against relapse in females. The current data suggest that ADNP is a potential novel biomarker and negative regulator of alcohol-drinking behaviors.
Collapse
Affiliation(s)
- Yarden Ziv
- 0000 0004 1937 0546grid.12136.37Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel ,0000 0004 1937 0546grid.12136.37Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Nofar Rahamim
- 0000 0004 1937 0546grid.12136.37Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978 Tel Aviv, Israel ,0000 0004 1937 0546grid.12136.37School of Psychological Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Noa Lezmy
- 0000 0004 1937 0546grid.12136.37Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978 Tel Aviv, Israel ,0000 0004 1937 0546grid.12136.37School of Psychological Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Oren Even-Chen
- 0000 0004 1937 0546grid.12136.37School of Psychological Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ohad Shaham
- 0000 0004 1937 0546grid.12136.37School of Psychological Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Anna Malishkevich
- 0000 0004 1937 0546grid.12136.37Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Eliezer Giladi
- 0000 0004 1937 0546grid.12136.37Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ran Elkon
- 0000 0004 1937 0546grid.12136.37Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel ,0000 0004 1937 0546grid.12136.37Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Illana Gozes
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel. .,Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978, Tel Aviv, Israel.
| | - Segev Barak
- Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978, Tel Aviv, Israel. .,School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
34
|
Moorman DE. The role of the orbitofrontal cortex in alcohol use, abuse, and dependence. Prog Neuropsychopharmacol Biol Psychiatry 2018; 87:85-107. [PMID: 29355587 PMCID: PMC6072631 DOI: 10.1016/j.pnpbp.2018.01.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/22/2017] [Accepted: 01/13/2018] [Indexed: 12/21/2022]
Abstract
One of the major functions of the orbitofrontal cortex (OFC) is to promote flexible motivated behavior. It is no surprise, therefore, that recent work has demonstrated a prominent impact of chronic drug use on the OFC and a potential role for OFC disruption in drug abuse and addiction. Among drugs of abuse, the use of alcohol is particularly salient with respect to OFC function. Although a number of studies in humans have implicated OFC dysregulation in alcohol use disorders, animal models investigating the association between OFC and alcohol use are only beginning to be developed, and there is still a great deal to be revealed. The goal of this review is to consider what is currently known regarding the role of the OFC in alcohol use and dependence. I will first provide a brief, general overview of current views of OFC function and its contributions to drug seeking and addiction. I will then discuss research to date related to the OFC and alcohol use, both in human clinical populations and in non-human models. Finally I will consider issues and strategies to guide future study that may identify this brain region as a key player in the transition from moderated to problematic alcohol use and dependence.
Collapse
Affiliation(s)
- David E. Moorman
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst MA 01003 USA
| |
Collapse
|
35
|
Joffe ME, Centanni SW, Jaramillo AA, Winder DG, Conn PJ. Metabotropic Glutamate Receptors in Alcohol Use Disorder: Physiology, Plasticity, and Promising Pharmacotherapies. ACS Chem Neurosci 2018; 9:2188-2204. [PMID: 29792024 PMCID: PMC6192262 DOI: 10.1021/acschemneuro.8b00200] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Developing efficacious treatments for alcohol use disorder (AUD) has proven difficult. The insidious nature of the disease necessitates a deep understanding of its underlying biology as well as innovative approaches to ameliorate ethanol-related pathophysiology. Excessive ethanol seeking and relapse are generated by long-term changes to membrane properties, synaptic physiology, and plasticity throughout the limbic system and associated brain structures. Each of these factors can be modulated by metabotropic glutamate (mGlu) receptors, a diverse set of G protein-coupled receptors highly expressed throughout the central nervous system. Here, we discuss how different components of the mGlu receptor family modulate neurotransmission in the limbic system and other brain regions involved in AUD etiology. We then describe how these processes are dysregulated following ethanol exposure and speculate about how mGlu receptor modulation might restore such pathophysiological changes. To that end, we detail the current understanding of the behavioral pharmacology of mGlu receptor-directed drug-like molecules in animal models of AUD. Together, this review highlights the prominent position of the mGlu receptor system in the pathophysiology of AUD and provides encouragement that several classes of mGlu receptor modulators may be translated as viable treatment options.
Collapse
Affiliation(s)
- Max E. Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| | - Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Anel A. Jaramillo
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Danny G. Winder
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| |
Collapse
|
36
|
Yawalkar R, Changotra H, Gupta GL. Protective influences of N-acetylcysteine against alcohol abstinence-induced depression by regulating biochemical and GRIN2A, GRIN2B gene expression of NMDA receptor signaling pathway in rats. Neurochem Int 2018; 118:73-81. [DOI: 10.1016/j.neuint.2018.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 10/17/2022]
|
37
|
Renteria R, Buske TR, Morrisett RA. Long-term subregion-specific encoding of enhanced ethanol intake by D1DR medium spiny neurons of the nucleus accumbens. Addict Biol 2018; 23:689-698. [PMID: 28656742 DOI: 10.1111/adb.12526] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/10/2017] [Accepted: 05/03/2017] [Indexed: 12/22/2022]
Abstract
The nucleus accumbens (NAc) is a critical component of the mesocorticolimbic system and is involved in mediating the motivational and reinforcing aspects of ethanol consumption. Chronic intermittent ethanol (CIE) exposure is a reliable model to induce ethanol dependence and increase volitional ethanol consumption in mice. Following a CIE-induced escalation of ethanol consumption, NMDAR (N-methyl-D-aspartate receptor)-dependent long-term depression in D1 dopamine receptor expressing medium spiny neurons of the NAc shell was markedly altered with no changes in plasticity in D1 dopamine receptor medium spiny neurons from the NAc core. This disruption of plasticity persisted for up to 2 weeks after cessation of ethanol access. To determine if changes in AMPA receptor (AMPAR) composition contribute to this ethanol-induced neuroadaptation, we monitored the rectification of AMPAR excitatory postsynaptic currents (EPSCs). We observed a marked decrease in the rectification index in the NAc shell, suggesting the presence of GluA2-lacking AMPARs. There was no change in the amplitude of spontaneous EPSCs (sEPSCs), but there was a transient increase in sEPSC frequency in the NAc shell. Using the paired pulse ratio, we detected a similar transient increase in the probability of neurotransmitter release. With no change in sEPSC amplitude, the change in the rectification index suggests that GluA2-containing AMPARs are removed and replaced with GluA2-lacking AMPARs in the NAc shell. This CIE-induced alteration in AMPAR subunit composition may contribute to the loss of NMDAR-dependent long-term depression in the NAc shell and therefore may constitute a critical neuroadaptive response underlying the escalation of ethanol intake in the CIE model.
Collapse
Affiliation(s)
- Rafael Renteria
- Institute for Neuroscience; The University of Texas at Austin; Austin Texas USA
| | - Tavanna R. Buske
- The College of Pharmacy; The University of Texas at Austin; Austin Texas USA
| | - Richard A. Morrisett
- Institute for Neuroscience; The University of Texas at Austin; Austin Texas USA
- The College of Pharmacy; The University of Texas at Austin; Austin Texas USA
- Waggoner Center for Alcohol and Addiction Research; The University of Texas at Austin; Austin Texas USA
| |
Collapse
|
38
|
Even-Chen O, Sadot-Sogrin Y, Shaham O, Barak S. Fibroblast Growth Factor 2 in the Dorsomedial Striatum Is a Novel Positive Regulator of Alcohol Consumption. J Neurosci 2017; 37:8742-8754. [PMID: 28821667 PMCID: PMC6596666 DOI: 10.1523/jneurosci.0890-17.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/16/2017] [Accepted: 07/31/2017] [Indexed: 12/13/2022] Open
Abstract
Repeated alcohol intake leads to mesostriatal neuroadaptations, resulting in drinking escalation and addiction phenotypes. Fibroblast growth factor 2 (FGF2) has been shown to interact with the mesostriatal dopaminergic system, and has been implicated in the actions of psychostimulants in the brain, and in several psychiatric disorders. Here, we report on a positive regulatory feedback loop of alcohol and FGF2 in rodent models. Specifically, we found that acute alcohol exposure (2.5 g/kg, i.p.) increased the mRNA expression of Fgf2 in the dorsal hippocampus, nucleus accumbens, and dorsal striatum. Longer alcohol exposure (7 d × 2.5 g/kg, i.p.) restricted these increases to the dorsal striatum, and the latter effect was blocked by the dopamine D2-like receptor antagonist haloperidol. Voluntary prolonged and excessive alcohol consumption in a 2-bottle choice procedure increased Fgf2 expression selectively in dorsomedial striatum (DMS) of both mice and rats. Importantly, we found that systemic administration of recombinant FGF2 (rFGF2) in mice, or rFGF2 infusion into the dorsal striatum or DMS of rats, increased alcohol consumption and preference, with no similar effects on saccharin or sucrose consumption. Finally, we found that inhibition of the endogenous FGF2 function in the DMS, by an anti-FGF2 neutralizing antibody, suppressed alcohol consumption and preference. Together, our results suggest that alcohol consumption increases the expression of Fgf2 in the DMS, and that striatal FGF2 promotes alcohol consumption, suggesting that FGF2 in the DMS is a positive regulator of alcohol drinking.SIGNIFICANCE STATEMENT Long-term alcohol intake may lead to neuroadaptations in the mesostriatal reward system, resulting in addiction phenotypes. Fibroblast growth factor 2 (FGF2) is crucial for the development and maintenance of the mesostriatal dopaminergic system. Here, we provide evidence for the involvement of FGF2 in alcohol-drinking behaviors. We show that alcohol increases Fgf2 expression in the dorsal striatum, an effect mediated via dopamine D2-like receptors. Importantly, we show that infusion of recombinant FGF2 into the dorsomedial striatum increases alcohol consumption, whereas inhibiting the endogenous FGF2 function suppresses consumption. Thus, FGF2 is an alcohol-responsive gene constituting a positive regulatory feedback loop with alcohol. This loop leads to facilitation of alcohol consumption, marking FGF2 as a potential new therapeutic target for alcohol addiction.
Collapse
Affiliation(s)
| | - Yossi Sadot-Sogrin
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | | | - Segev Barak
- School of Psychological Sciences and
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| |
Collapse
|
39
|
Bell RL, Hauser SR, Liang T, Sari Y, Maldonado-Devincci A, Rodd ZA. Rat animal models for screening medications to treat alcohol use disorders. Neuropharmacology 2017; 122:201-243. [PMID: 28215999 PMCID: PMC5659204 DOI: 10.1016/j.neuropharm.2017.02.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 01/21/2023]
Abstract
The purpose of this review is to present animal research models that can be used to screen and/or repurpose medications for the treatment of alcohol abuse and dependence. The focus will be on rats and in particular selectively bred rats. Brief introductions discuss various aspects of the clinical picture, which provide characteristics of individuals with alcohol use disorders (AUDs) to model in animals. Following this, multiple selectively bred rat lines will be described and evaluated in the context of animal models used to screen medications to treat AUDs. Next, common behavioral tests for drug efficacy will be discussed particularly as they relate to stages in the addiction cycle. Tables highlighting studies that have tested the effects of compounds using the respective techniques are included. Wherever possible the Tables are organized chronologically in ascending order to describe changes in the focus of research on AUDs over time. In general, high ethanol-consuming selectively bred rats have been used to test a wide range of compounds. Older studies usually followed neurobiological findings in the selected lines that supported an association with a propensity for high ethanol intake. Most of these tests evaluated the compound's effects on the maintenance of ethanol drinking. Very few compounds have been tested during ethanol-seeking and/or relapse and fewer still have assessed their effects during the acquisition of AUDs. Overall, while a substantial number of neurotransmitter and neuromodulatory system targets have been assessed; the roles of sex- and age-of-animal, as well as the acquisition of AUDs, ethanol-seeking and relapse continue to be factors and behaviors needing further study. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Richard L Bell
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA.
| | - Sheketha R Hauser
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| | - Tiebing Liang
- Indiana University School of Medicine, Department of Gastroenterology, Indianapolis, IN 46202, USA
| | - Youssef Sari
- University of Toledo, Department of Pharmacology, Toledo, OH 43614, USA
| | | | - Zachary A Rodd
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| |
Collapse
|
40
|
McGraw JJ, Zona LC, Cromwell HC. The effects of ethanol on diverse components of choice in the rat: reward discrimination, preference and relative valuation. Eur J Neurosci 2017. [PMID: 28639261 DOI: 10.1111/ejn.13627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alcohol consumption impairs judgment and choice. How alcohol alters these crucial processes is primarily unknown. Choice can be fractionated into different components including reward discrimination, preference and relative valuation that can function together or in isolation depending upon diverse factors including choice context. We examined the diverse components and contextual effects by analyzing the effects of alcohol drinking on choice behavior in a task with a reduced level of temporal and spatial constraints. Rats were trained to drink 10% ethanol during 6 weeks of behavior testing using a combined sucrose-fade and two-bottle free-choice procedure. Two different sucrose pellet outcomes (e.g., constant vs. variable) were presented each week to examine the impact of voluntary drinking on reward-based decision-making. Behavioral contexts of single option, free choice and extinction were examined for each outcome set. Comparisons were made between alcohol and control groups and within the alcohol group over time to inspect choice profiles. Between-group results showed alcohol drinking animals expressed altered place preference and modified sucrose reward approach latencies. The within-group profile showed that alcohol drinking animals can express adequate reward discrimination, preference and incentive contrast during free choice. All of these components were significantly reduced during the context of extinction. Control animals were also impacted by extinction but not as severely. The findings point to a need for a greater focus on the context and the diverse components of choice when examining external and internal factors influencing decision-making during alcohol or other substance of abuse exposure.
Collapse
Affiliation(s)
- Justin J McGraw
- J.P. Scott Center for Neuroscience, Mind and Behavior and Department of Psychology, Bowling Green State University, Bowling Green, OH, USA
| | - Luke C Zona
- J.P. Scott Center for Neuroscience, Mind and Behavior and Department of Psychology, Bowling Green State University, Bowling Green, OH, USA
| | - Howard C Cromwell
- J.P. Scott Center for Neuroscience, Mind and Behavior and Department of Psychology, Bowling Green State University, Bowling Green, OH, USA
| |
Collapse
|
41
|
Hwa L, Besheer J, Kash T. Glutamate plasticity woven through the progression to alcohol use disorder: a multi-circuit perspective. F1000Res 2017; 6:298. [PMID: 28413623 PMCID: PMC5365217 DOI: 10.12688/f1000research.9609.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2017] [Indexed: 12/18/2022] Open
Abstract
Glutamate signaling in the brain is one of the most studied targets in the alcohol research field. Here, we report the current understanding of how the excitatory neurotransmitter glutamate, its receptors, and its transporters are involved in low, episodic, and heavy alcohol use. Specific animal behavior protocols can be used to assess these different drinking levels, including two-bottle choice, operant self-administration, drinking in the dark, the alcohol deprivation effect, intermittent access to alcohol, and chronic intermittent ethanol vapor inhalation. Importantly, these methods are not limited to a specific category, since they can be interchanged to assess different states in the development from low to heavy drinking. We encourage a circuit-based perspective beyond the classic mesolimbic-centric view, as multiple structures are dynamically engaged during the transition from positive- to negative-related reinforcement to drive alcohol drinking. During this shift from lower-level alcohol drinking to heavy alcohol use, there appears to be a shift from metabotropic glutamate receptor-dependent behaviors to N-methyl-D-aspartate receptor-related processes. Despite high efficacy of the glutamate-related pharmaceutical acamprosate in animal models of drinking, it is ineffective as treatment in the clinic. Therefore, research needs to focus on other promising glutamatergic compounds to reduce heavy drinking or mediate withdrawal symptoms or both.
Collapse
Affiliation(s)
- Lara Hwa
- Department of Pharmacology, University of North Carolina School of Medicine, Bowles Center for Alcohol Studies, Chapel Hill, NC, 27599, USA
| | - Joyce Besheer
- Department of Psychiatry, University of North Carolina School of Medicine, Bowles Center for Alcohol Studies, Chapel Hill, NC, 27599, USA
| | - Thomas Kash
- Department of Pharmacology, University of North Carolina School of Medicine, Bowles Center for Alcohol Studies, Chapel Hill, NC, 27599, USA
| |
Collapse
|
42
|
Holgate JY, Shariff M, Mu EWH, Bartlett S. A Rat Drinking in the Dark Model for Studying Ethanol and Sucrose Consumption. Front Behav Neurosci 2017; 11:29. [PMID: 28275340 PMCID: PMC5319958 DOI: 10.3389/fnbeh.2017.00029] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/08/2017] [Indexed: 01/21/2023] Open
Abstract
Background: The intermittent access 2-bottle choice (IA2BC) and drinking in the dark (DID) models were developed for studying rodent binge-like consumption. Traditionally, IA2BC was used with rats and DID with mice. Recently, IA2BC was adapted to study mouse ethanol consumption. However, it is unknown whether DID is suitable for rats or if one rat model is more advantageous than another for studying binge-like consumption. Methods: Male Wistar rats consumed 20% ethanol or 5% sucrose using IA2BC or DID for 12 weeks. IA2BC drinking sessions occurred on alternate days (Mondays–Fridays) and lasted 24 h, whereas DID sessions ran 4 h/day, 5 days/week (Monday–Friday). Average consumption/session, week and hour was measured. To explore DID model suitability for screening novel compounds for controlling ethanol and sucrose intake, varenicline (2 mg/kg) or vehicle was administered to DID rats. Results: IA2BC rats consume more ethanol/session and similar amounts of ethanol/week than DID rats. While, IA2BC rats consume more sucrose/session and week than DID rats. Although IA2BC rats had more ethanol and sucrose access time, DID rats had greater ethanol and sucrose intake/hour. Varenicline significantly reduced ethanol and sucrose consumption in DID rats, consistent with previously published IA2BC studies. Conclusions: Despite the shorter access time, the rat DID model induced higher initial intake and greater consumption/hour for both ethanol and sucrose. The shorter duration of DID sessions did not prevent detection of varenicline-induced reductions in ethanol or sucrose consumption, suggesting the DID model may be suitable for studying binge-like ethanol and sucrose consumption.
Collapse
Affiliation(s)
- Joan Y Holgate
- Institute of Health and Medical Innovation, Translational Research Institute, Queensland University of Technology Woolloongabba, QLD, Australia
| | - Masroor Shariff
- Institute of Health and Medical Innovation, Translational Research Institute, Queensland University of Technology Woolloongabba, QLD, Australia
| | - Erica W H Mu
- Institute of Health and Medical Innovation, Translational Research Institute, Queensland University of Technology Woolloongabba, QLD, Australia
| | - Selena Bartlett
- Institute of Health and Medical Innovation, Translational Research Institute, Queensland University of Technology Woolloongabba, QLD, Australia
| |
Collapse
|
43
|
Abstract
The main characteristic of alcohol use disorder is the consumption of large quantities of alcohol despite the negative consequences. The transition from the moderate use of alcohol to excessive, uncontrolled alcohol consumption results from neuroadaptations that cause aberrant motivational learning and memory processes. Here, we examine studies that have combined molecular and behavioural approaches in rodents to elucidate the molecular mechanisms that keep the social intake of alcohol in check, which we term 'stop pathways', and the neuroadaptations that underlie the transition from moderate to uncontrolled, excessive alcohol intake, which we term 'go pathways'. We also discuss post-transcriptional, genetic and epigenetic alterations that underlie both types of pathways.
Collapse
Affiliation(s)
- Dorit Ron
- Corresponding author: Dorit Ron, 675 Nelson Rising Lane, BOX 0663, San Francisco, CA 94143-0663,
| | - Segev Barak
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
- School of Psychological Sciences and Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
44
|
Quadir SG, Santos JRBD, Campbell RR, Wroten MG, Singh N, Holloway JJ, Bal SK, Camarini R, Szumlinski KK. Homer2 regulates alcohol and stress cross-sensitization. Addict Biol 2016; 21:613-33. [PMID: 25916683 DOI: 10.1111/adb.12252] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
An interaction exists between stress and alcohol in the etiology and chronicity of alcohol use disorders, yet a knowledge gap exists regarding the neurobiological underpinnings of this interaction. In this regard, we employed an 11-day unpredictable, chronic, mild stress (UCMS) procedure to examine for stress-alcohol cross-sensitization of motor activity as well as alcohol consumption/preference and intoxication. We also employed immunoblotting to relate the expression of glutamate receptor-related proteins within subregions of the nucleus accumbens (NAC) to the manifestation of behavioral cross-sensitization. UCMS mice exhibited a greater locomotor response to an acute injection of 2 g/kg alcohol than unstressed controls and this cross-sensitization extended to alcohol intake (0-20 percent), as well as to the intoxicating and sedative properties of 3 and 5 g/kg alcohol, respectively. Regardless of prior alcohol injection (2 g/kg), UCMS mice exhibited elevated NAC shell levels of mGlu1α, GluN2b and Homer2, as well as lower phospholipase Cβ within this subregion. GluN2b levels were also lower within the NAC core of UCMS mice. The expression of stress-alcohol locomotor cross-sensitization was associated with lower mGlu1α within the NAC core and lower extracellular signal-regulated kinase activity within both NAC subregions. As Homer2 regulates alcohol sensitization, we assayed also for locomotor cross-sensitization in Homer2 wild-type (WT) and knock-out (KO) mice. WT mice exhibited a very robust cross-sensitization that was absent in KO animals. These results indicate that a history of mild stress renders an animal more sensitive to the psychomotor and rewarding properties of alcohol, which may depend on neuroplasticity within NAC glutamate transmission.
Collapse
Affiliation(s)
- Sema G. Quadir
- Department of Psychological and Brain Sciences; Neuroscience Research Institute; University of California Santa Barbara; Santa Barbara CA USA
| | | | - Rianne R. Campbell
- Department of Psychological and Brain Sciences; Neuroscience Research Institute; University of California Santa Barbara; Santa Barbara CA USA
| | - Melissa G. Wroten
- Department of Psychological and Brain Sciences; Neuroscience Research Institute; University of California Santa Barbara; Santa Barbara CA USA
| | - Nimrita Singh
- Department of Psychological and Brain Sciences; Neuroscience Research Institute; University of California Santa Barbara; Santa Barbara CA USA
| | - John J. Holloway
- Department of Psychological and Brain Sciences; Neuroscience Research Institute; University of California Santa Barbara; Santa Barbara CA USA
| | - Sukhmani K. Bal
- Department of Psychological and Brain Sciences; Neuroscience Research Institute; University of California Santa Barbara; Santa Barbara CA USA
| | - Rosana Camarini
- Department of Pharmacology; Institute of Biomedical Sciences; Universidade de São Paulo; São Paulo Brazil
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences; Neuroscience Research Institute; University of California Santa Barbara; Santa Barbara CA USA
| |
Collapse
|
45
|
Renteria R, Jeanes ZM, Mangieri RA, Maier EY, Kircher DM, Buske TR, Morrisett RA. Using In Vitro Electrophysiology to Screen Medications: Accumbal Plasticity as an Engram of Alcohol Dependence. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:441-65. [PMID: 27055622 DOI: 10.1016/bs.irn.2016.02.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The nucleus accumbens (NAc) is a central component of the mesocorticolimbic reward system. Increasing evidence strongly implicates long-term synaptic neuroadaptations in glutamatergic excitatory activity of the NAc shell and/or core medium spiny neurons in response to chronic drug and alcohol exposure. Such neuroadaptations likely play a critical role in the development and expression of drug-seeking behaviors. We have observed unique cell-type-specific bidirectional changes in NAc synaptic plasticity (metaplasticity) following acute and chronic intermittent ethanol exposure. Other investigators have also previously observed similar metaplasticity in the NAc following exposure to psychostimulants, opiates, and amazingly, even following an anhedonia-inducing experience. Considering that the proteome of the postsynaptic density likely contains hundreds of biochemicals, proteins and other components and regulators, we believe that there is a large number of potential molecular sites through which accumbal metaplasticity may be involved in chronic alcohol abuse. Many of our companion laboratories are now engaged in identifying and screening medications targeting candidate genes and its products previously linked to maladaptive alcohol phenotypes. We hypothesize that if manipulation of such target genes and their products change NAc plasticity, then that observation constitutes an important validation step for the development of novel therapeutics to treat alcohol dependence.
Collapse
Affiliation(s)
- R Renteria
- University of Texas at Austin, Austin, TX, United States
| | - Z M Jeanes
- University of Texas at Austin, Austin, TX, United States
| | - R A Mangieri
- University of Texas at Austin, Austin, TX, United States
| | - E Y Maier
- University of Texas at Austin, Austin, TX, United States
| | - D M Kircher
- University of Texas at Austin, Austin, TX, United States
| | - T R Buske
- University of Texas at Austin, Austin, TX, United States
| | - R A Morrisett
- University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
46
|
Becker HC, Lopez MF. An Animal Model of Alcohol Dependence to Screen Medications for Treating Alcoholism. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:157-77. [PMID: 27055614 DOI: 10.1016/bs.irn.2016.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite the high prevalence of alcohol use disorders in the United States, only a relatively small percentage of those afflicted seek treatment. This is further compounded by the fact that there are too few medications available to effectively treat this significant public health problem. The need for identifying and evaluating more effective treatments that aid in preventing relapse and/or tempering risky and harmful alcohol consumption cannot be overstated. Use of animal models represents a critical step in the process of screening, identifying, and informing plans for prioritizing the most promising candidate medications that can be advanced to the next stage of evaluation (clinical laboratory paradigms and controlled clinical trials). Numerous animal models have been developed to study excessive levels of alcohol self-administration. In recent years, a large literature has amassed of studies in which rodent models of dependence have been linked with alcohol self-administration procedures. This chapter focuses on studies employing a dependence model that involves chronic exposure to alcohol vapor by inhalation, which yields in both mice and rats significant escalation of voluntary alcohol consumption. These animal models of dependence and alcohol self-administration have revealed valuable insights about underlying mechanisms that drive excessive drinking. Additionally, this preclinical approach is useful in evaluating the effects of medications on escalated drinking associated with dependence vs more stable levels displayed by nondependent animals.
Collapse
Affiliation(s)
- H C Becker
- Charleston Alcohol Research Center, Charleston, SC, United States; Medical University of South Carolina, Charleston, SC, United States; RHJ Department of Veterans Affairs Medical Center, Charleston, SC, United States.
| | - M F Lopez
- Charleston Alcohol Research Center, Charleston, SC, United States
| |
Collapse
|
47
|
Renteria R, Maier EY, Buske TR, Morrisett RA. Selective alterations of NMDAR function and plasticity in D1 and D2 medium spiny neurons in the nucleus accumbens shell following chronic intermittent ethanol exposure. Neuropharmacology 2016; 112:164-171. [PMID: 26946430 DOI: 10.1016/j.neuropharm.2016.03.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 02/22/2016] [Accepted: 03/01/2016] [Indexed: 12/27/2022]
Abstract
A major mouse model widely adopted in recent years to induce pronounced ethanol intake is the ethanol vapor model known as "CIE" or "Chronic Intermittent Ethanol." One critical question concerning this model is whether the rapid induction of high blood ethanol levels for such short time periods is sufficient to induce alterations in N-methyl-d-aspartate receptor (NMDAR) function which may contribute to excessive ethanol intake. In this study, we determined whether such short term intermittent ethanol exposure modulates NMDAR function as well as other prominent electrophysiological properties and the expression of plasticity in both D1 (D1+) and D2 (D1-) dopamine receptor expressing medium spiny neurons (MSNs) in the nucleus accumbens (NAc) shell. To distinguish between the two subtypes of MSNs in the NAc we treated Drd1a-TdTomato transgenic mice with CIE vapor and electrophysiological recordings were conducted 24 h after the last vapor exposure. To investigate CIE induced alterations in plasticity, long-term depression (LTD) was induced by pairing low frequency stimulation (LFS) with post synaptic depolarization. In ethanol naïve mice, LFS induced synaptic depression (LTD) was apparent exclusively in D1+ MSNs. Whereas in slices prepared from CIE treated mice, LFS induced synaptic potentiation (LTP) in D1+ MSNs. Furthermore, following CIE exposure, LFS now produced LTD in D1- MSNs. We found that CIE exposure induced an increase in excitability in D1+ MSNs with no change in D1- MSNs. After CIE, we found a significant increase in spontaneous EPSCs (sEPSCs) frequency in D1+ but not D1- MSNs suggesting alterations in baseline α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) mediated signaling. CIE induced changes in NMDAR function were measured using the NMDA/AMPA ratio and input-output curves of isolated NMDAR currents. We observed a significant increase in NMDAR function in D1+ MSNs and a decrease in D1- MSNs after ethanol vapor exposure. The reversal of NMDAR function may account for the CIE induced alterations in the expression of plasticity. The cell type specific alterations in excitatory signaling in the NAc shell may constitute an important neuroadaptation necessary for the expression of increased ethanol consumption induced by intermittent ethanol vapor exposure. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.
Collapse
Affiliation(s)
- Rafael Renteria
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA.
| | - Esther Y Maier
- The Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX, USA.
| | - Tavanna R Buske
- The Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX, USA.
| | - Richard A Morrisett
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA; The Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX, USA; Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
48
|
Phillips TJ, Reed C, Pastor R. Preclinical evidence implicating corticotropin-releasing factor signaling in ethanol consumption and neuroadaptation. GENES BRAIN AND BEHAVIOR 2015; 14:98-135. [PMID: 25565358 DOI: 10.1111/gbb.12189] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/22/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022]
Abstract
The results of many studies support the influence of the corticotropin-releasing factor (CRF) system on ethanol (EtOH) consumption and EtOH-induced neuroadaptations that are critical in the addiction process. This review summarizes the preclinical data in this area after first providing an overview of the components of the CRF system. This complex system involves hypothalamic and extra-hypothalamic mechanisms that play a role in the central and peripheral consequences of stressors, including EtOH and other drugs of abuse. In addition, several endogenous ligands and targets make up this system and show differences in their involvement in EtOH drinking and in the effects of chronic or repeated EtOH treatment. In general, genetic and pharmacological approaches paint a consistent picture of the importance of CRF signaling via type 1 CRF receptors (CRF(1)) in EtOH-induced neuroadaptations that result in higher levels of intake, encourage alcohol seeking during abstinence and alter EtOH sensitivity. Furthermore, genetic findings in rodents, non-human primates and humans have provided some evidence of associations of genetic polymorphisms in CRF-related genes with EtOH drinking, although additional data are needed. These results suggest that CRF(1) antagonists have potential as pharmacotherapeutics for alcohol use disorders. However, given the broad and important role of these receptors in adaptation to environmental and other challenges, full antagonist effects may be too profound and consideration should be given to treatments with modulatory effects.
Collapse
Affiliation(s)
- T J Phillips
- VA Portland Health Care System, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA; Department of Behavioral Neuroscience, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | | |
Collapse
|
49
|
Pleil KE, Lowery-Gionta EG, Crowley NA, Li C, Marcinkiewcz CA, Rose JH, McCall NM, Maldonado-Devincci AM, Morrow AL, Jones SR, Kash TL. Effects of chronic ethanol exposure on neuronal function in the prefrontal cortex and extended amygdala. Neuropharmacology 2015; 99:735-49. [PMID: 26188147 DOI: 10.1016/j.neuropharm.2015.06.017] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/28/2015] [Accepted: 06/26/2015] [Indexed: 10/23/2022]
Abstract
Chronic alcohol consumption and withdrawal leads to anxiety, escalated alcohol drinking behavior, and alcohol dependence. Alterations in the function of key structures within the cortico-limbic neural circuit have been implicated in underlying the negative behavioral consequences of chronic alcohol exposure in both humans and rodents. Here, we used chronic intermittent ethanol vapor exposure (CIE) in male C57BL/6J mice to evaluate the effects of chronic alcohol exposure and withdrawal on anxiety-like behavior and basal synaptic function and neuronal excitability in prefrontal cortical and extended amygdala brain regions. Forty-eight hours after four cycles of CIE, mice were either assayed in the marble burying test (MBT) or their brains were harvested and whole-cell electrophysiological recordings were performed in the prelimbic and infralimbic medial prefrontal cortex (PLC and ILC), the lateral and medial central nucleus of the amygdala (lCeA and mCeA), and the dorsal and ventral bed nucleus of the stria terminalis (dBNST and vBNST). Ethanol-exposed mice displayed increased anxiety in the MBT compared to air-exposed controls, and alterations in neuronal function were observed in all brain structures examined, including several distinct differences between subregions within each structure. Chronic ethanol exposure induced hyperexcitability of the ILC, as well as a shift toward excitation in synaptic drive and hyperexcitability of vBNST neurons; in contrast, there was a net inhibition of the CeA. This study reveals extensive effects of chronic ethanol exposure on the basal function of cortico-limbic brain regions, suggests that there may be complex interactions between these regions in the regulation of ethanol-dependent alterations in anxiety state, and highlights the need for future examination of projection-specific effects of ethanol in cortico-limbic circuitry.
Collapse
Affiliation(s)
- Kristen E Pleil
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Emily G Lowery-Gionta
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nicole A Crowley
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chia Li
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Catherine A Marcinkiewcz
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jamie H Rose
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nora M McCall
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | - A Leslie Morrow
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Sara R Jones
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
50
|
Griffin WC, Ramachandra VS, Knackstedt LA, Becker HC. Repeated cycles of chronic intermittent ethanol exposure increases basal glutamate in the nucleus accumbens of mice without affecting glutamate transport. Front Pharmacol 2015; 6:27. [PMID: 25755641 PMCID: PMC4337330 DOI: 10.3389/fphar.2015.00027] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/02/2015] [Indexed: 11/28/2022] Open
Abstract
Repeated cycles of chronic intermittent ethanol (CIE) exposure increase voluntary consumption of ethanol in mice. Previous work has shown that extracellular glutamate in the nucleus accumbens (NAc) is significantly elevated in ethanol-dependent mice and that pharmacologically manipulating glutamate concentrations in the NAc will alter ethanol drinking, indicating that glutamate homeostasis plays a crucial role in ethanol drinking in this model. The present studies were designed to measure extracellular glutamate at a time point in which mice would ordinarily be allowed voluntary access to ethanol in the CIE model and, additionally, to measure glutamate transport capacity in the NAc at the same time point. Extracellular glutamate was measured using quantitative microdialysis procedures. Glutamate transport capacity was measured under Na+-dependent and Na+-independent conditions to determine whether the function of excitatory amino acid transporters (also known as system XAG) or of system Xc– (glial cysteine–glutamate exchanger) was influenced by CIE exposure. The results of the quantitative microdialysis experiment confirm increased extracellular glutamate (approximately twofold) in the NAc of CIE exposed mice (i.e., ethanol-dependent) compared to non-dependent mice in the NAc, consistent with earlier work. However, the increase in extracellular glutamate was not due to altered transporter function in the NAc of ethanol-dependent mice, because neither Na+-dependent nor Na+-independent glutamate transport was significantly altered by CIE exposure. These findings point to the possibility that hyperexcitability of cortical–striatal pathways underlies the increases in extracellular glutamate found in the ethanol-dependent mice.
Collapse
Affiliation(s)
- William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina , Charleston, SC, USA
| | - Vorani S Ramachandra
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina , Charleston, SC, USA
| | - Lori A Knackstedt
- Department of Psychology, University of Florida , Gainesville, FL, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina , Charleston, SC, USA ; Department of Neurosciences, Medical University of South Carolina , Charleston, SC, USA ; Ralph H. Johnson VA Medical Center , Charleston, SC, USA
| |
Collapse
|