1
|
Nippert KE, Rowland CP, Vazey EM, Moorman DE. Alcohol, flexible behavior, and the prefrontal cortex: Functional changes underlying impaired cognitive flexibility. Neuropharmacology 2024; 260:110114. [PMID: 39134298 PMCID: PMC11694314 DOI: 10.1016/j.neuropharm.2024.110114] [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: 02/15/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Cognitive flexibility enables individuals to alter their behavior in response to changing environmental demands, facilitating optimal behavior in a dynamic world. The inability to do this, called behavioral inflexibility, is a pervasive behavioral phenotype in alcohol use disorder (AUD), driven by disruptions in cognitive flexibility. Research has repeatedly shown that behavioral inflexibility not only results from alcohol exposure across species but can itself be predictive of future drinking. Like many high-level executive functions, flexible behavior requires healthy functioning of the prefrontal cortex (PFC). The scope of this review addresses two primary themes: first, we outline tasks that have been used to investigate flexibility in the context of AUD or AUD models. We characterize these based on the task features and underlying cognitive processes that differentiate them from one another. We highlight the neural basis of flexibility measures, focusing on the PFC, and how acute or chronic alcohol in humans and non-human animal models impacts flexibility. Second, we consolidate findings on the molecular, physiological and functional changes in the PFC elicited by alcohol, that may contribute to cognitive flexibility deficits seen in AUD. Collectively, this approach identifies several key avenues for future research that will facilitate effective treatments to promote flexible behavior in the context of AUD, to reduce the risk of alcohol related harm, and to improve outcomes following AUD. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Kathryn E Nippert
- Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Courtney P Rowland
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Elena M Vazey
- Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA; Department of Biology, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| | - David E Moorman
- Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA; Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
2
|
Shen G, Chen YH, Wu Y, Jiahui H, Fang J, Jiayi T, Yimin K, Wang W, Liu Y, Wang F, Chen L. Exploring core symptoms of alcohol withdrawal syndrome in alcohol use disorder patients: a network analysis approach. Front Psychiatry 2024; 15:1320248. [PMID: 39267702 PMCID: PMC11390437 DOI: 10.3389/fpsyt.2024.1320248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 08/07/2024] [Indexed: 09/15/2024] Open
Abstract
Background Understanding the interplay between psychopathology of alcohol withdrawal syndrome (AWS) in alcohol use disorder (AUD) patients may improve the effectiveness of relapse interventions for AUD. Network theory of mental disorders assumes that mental disorders persist not of a common functional disorder, but from a sustained feedback loop between symptoms, thereby explaining the persistence of AWS and the high relapse rate of AUD. The current study aims to establish a network of AWS, identify its core symptoms and find the bridges between the symptoms which are intervention target to relieve the AWS and break the self-maintaining cycle of AUD. Methods Graphical lasso network were constructed using psychological symptoms of 553 AUD patients. Global network structure, centrality indices, cluster coefficient, and bridge symptom were used to identify the core symptoms of the AWS network and the transmission pathways between different symptom clusters. Results The results revealed that: (1) AWS constitutes a stable symptom network with a stability coefficient (CS) of 0.21-0.75. (2) Anger (Strength = 1.52) and hostility (Strength = 0.84) emerged as the core symptom in the AWS network with the highest centrality and low clustering coefficient. (3) Hostility mediates aggression and anxiety; anger mediates aggression and impulsivity in AWS network respectively. Conclusions Anger and hostility may be considered the best intervention targets for researching and treating AWS. Hostility and anxiety, anger and impulsiveness are independent but related dimensions, suggesting that different neurobiological bases may be involved in withdrawal symptoms, which play a similar role in withdrawal syndrome.
Collapse
Affiliation(s)
- Guanghui Shen
- Department of Behavioral Medicine, Wenzhou Seventh People's Hospital, Wenzhou, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Yu-Hsin Chen
- The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuyu Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Huang Jiahui
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Juan Fang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Tang Jiayi
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Kang Yimin
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, China
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, China
| | - Li Chen
- The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, China
| |
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
|
Torres Irizarry VC, Feng B, Yang X, Patel N, Schaul S, Ibrahimi L, Ye H, Luo P, Carrillo-Sáenz L, Lai P, Kota M, Dixit D, Wang C, Lasek AW, He Y, Xu P. Estrogen signaling in the dorsal raphe regulates binge-like drinking in mice. Transl Psychiatry 2024; 14:122. [PMID: 38413577 PMCID: PMC10899193 DOI: 10.1038/s41398-024-02821-2] [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: 01/19/2024] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
Estrogens promote binge alcohol drinking and contribute to sex differences in alcohol use disorder. However, the mechanisms are largely unknown. This study aims to test if estrogens act on 5-hydroxytryptamine neurons in the dorsal raphe nucleus (5-HTDRN) to promote binge drinking. We found that female mice drank more alcohol than male mice in chronic drinking in the dark (DID) tests. This sex difference was associated with distinct alterations in mRNA expression of estrogen receptor α (ERα) and 5-HT-related genes in the DRN, suggesting a potential role of estrogen/ERs/5-HT signaling. In supporting this view, 5-HTDRN neurons from naïve male mice had lower baseline firing activity but higher sensitivity to alcohol-induced excitation compared to 5-HTDRN neurons from naïve female mice. Notably, this higher sensitivity was blunted by 17β-estradiol treatment in males, indicating an estrogen-dependent mechanism. We further showed that both ERα and ERβ are expressed in 5-HTDRN neurons, whereas ERα agonist depolarizes and ERβ agonist hyperpolarizes 5-HTDRN neurons. Notably, both treatments blocked the stimulatory effects of alcohol on 5-HTDRN neurons in males, even though they have antagonistic effects on the activity dynamics. These results suggest that ERs' inhibitory effects on ethanol-induced burst firing of 5-HTDRN neurons may contribute to higher levels of binge drinking in females. Consistently, chemogenetic activation of ERα- or ERβ-expressing neurons in the DRN reduced binge alcohol drinking. These results support a model in which estrogens act on ERα/β to prevent alcohol-induced activation of 5-HTDRN neurons, which in return leads to higher binge alcohol drinking.
Collapse
Affiliation(s)
- Valeria C Torres Irizarry
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bing Feng
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Xiaohua Yang
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, 510642, Guangzhou, Guangdong, China
| | - Nirali Patel
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sarah Schaul
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Lucas Ibrahimi
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Hui Ye
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Pei Luo
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, 510642, Guangzhou, Guangdong, China
| | - Leslie Carrillo-Sáenz
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Penghua Lai
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Maya Kota
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Devin Dixit
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VI, 23298, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, 70808, USA.
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA.
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
5
|
Bi-directional modulation of hyperpolarization-activated cation currents (I h) by ethanol in rat hippocampal CA3 pyramidal neurons. Neuropharmacology 2023; 227:109423. [PMID: 36690323 DOI: 10.1016/j.neuropharm.2023.109423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
It is widely acknowledged that ethanol (EtOH) can alter many neuronal functions, including synaptic signaling, firing discharge, and membrane excitability, through its interaction with multiple membrane proteins and intracellular pathways. Previous work has demonstrated that EtOH enhances the firing rate of hippocampal GABAergic interneurons and thus the presynaptic GABA release at CA1 and CA3 inhibitory synapses through a positive modulation of the hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels. Activation of HCN channels produce an inward current, commonly called Ih, which plays an essential role in generating/regulating specific neuronal activities in GABAergic interneurons and principal glutamatergic pyramidal neurons such as those in the CA3 subregion. Since the direct effect of EtOH on HCN channels expressed in CA3 pyramidal neurons was not thoroughly elucidated, we investigated the possible interaction between EtOH and HCN channels and the impact on excitability and postsynaptic integration of these neurons. Patch-clamp recordings were performed in single CA3 pyramidal neurons from acute male rat coronal hippocampal slices. Our results show that EtOH modulates HCN-mediated Ih in a concentration-dependent and bi-directional manner, with a positive modulation at lower (20 mM) and an inhibitory action at higher (60-80 mM) concentrations. The modulation of Ih by EtOH was mimicked by forskolin, antagonized by different drugs that selectively interfere with the AC/cAMP/PKA intracellular pathway, as well as by the selective HCN inhibitor ZD7288. Altogether, these data further support the evidence that HCN channels may represent an important molecular target through which EtOH may regulate neuronal activity.
Collapse
|
6
|
Hasegawa H, Tanaka T, Kondo M, Teramoto K, Nakayama K, Hwang GW. Blood vessel remodeling in the cerebral cortex induced by binge alcohol intake in mice. Toxicol Res 2023; 39:169-177. [PMID: 36726835 PMCID: PMC9839917 DOI: 10.1007/s43188-022-00164-y] [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: 09/29/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Ethanol is toxic to the brain and causes various neurological disorders. Although ethanol can directly exert toxicity on neurons, it also acts on other cell types in the central nervous system. Blood vessel endothelial cells interact with, and are affected by blood ethanol. However, the effects of ethanol on the vascular structures of the brain have not been well documented. In this study, we examined the effects of binge levels of ethanol on brain vasculature. Immunostaining analysis indicated structural alterations of blood vessels in the cerebral cortex, which became more tortuous than those in the control mice after ethanol administration. The interaction between the blood vessels and astrocytes decreased, especially in the upper layers of the cerebral cortex. Messenger RNA expression analysis revealed a unique downregulation of Vegfa mRNA encoding vascular endothelial growth factor (VEGF)-A among VEGF, angiopoietin, endothelin family angiogenic and blood vessel remodeling factors. The expression of three proteoglycan core proteins, glypican-5, neurocan, and serglycin, was also altered after ethanol administration. Thus, binge levels of ethanol affect the expression of VEGF-A and blood vessel-supporting proteoglycans, resulting in changes in the vascular structure of the cerebral cortex. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-022-00164-y.
Collapse
Affiliation(s)
- Hiroshi Hasegawa
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Toshiya Tanaka
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Mari Kondo
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Koji Teramoto
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Kei Nakayama
- Laboratory of Hygienic Sciences, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-Machi, Higashinada-Ku, Kobe, 6588558 Japan
| | - Gi-Wook Hwang
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 9818558 Japan
| |
Collapse
|
7
|
Nimitvilai-Roberts S, Woodward JJ. The ethanol inhibition of basolateral amygdala neuron spiking is mediated by a γ-aminobutyric acid type A-mediated tonic current. Alcohol Clin Exp Res 2022; 46:1665-1676. [PMID: 35915568 PMCID: PMC9509443 DOI: 10.1111/acer.14918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/19/2022] [Accepted: 07/28/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND The basolateral nucleus of the amygdala (BLA) plays an important role in the development of fear and anxiety-related behaviors. The BLA receives inputs from all sensory stimuli. After processing those stimuli, BLA neurons signal neurons within the central amygdala and other brain regions, including the ventral and dorsal striatum and frontal cortex. Studies suggest that the BLA is involved in drug dependence and in the reinforcing actions of ethanol. For example, acute exposure to ethanol reduces anxiety, while withdrawal from chronic ethanol exposure alters BLA synaptic transmission, which increases anxiety, a common underlying cause of relapse. Exposure to and withdrawal from chronic alcohol also disrupts many brain areas that connect with the BLA. Despite these important findings, the acute actions of alcohol on the intrinsic excitability of BLA neurons have not been fully characterized. METHODS Brain slices containing the BLA were prepared from adult C57BL/6J male mice. Whole-cell and sharp electrode electrophysiological recordings were performed to characterize the effects of acute ethanol on BLA neuronal and astrocyte function, respectively. RESULTS Ethanol inhibited action potential (AP) firing of BLA neurons but had no effect on BLA astrocyte resting membrane potential. The ethanol-induced inhibition of firing was concentration-dependent (11 to 66 mM) and accompanied by a reduction in the input resistance and an increase in the rheobase of BLA neurons. The inhibitory effect of ethanol was suppressed by picrotoxin, which blocks both γ-aminobutyric acid type A (GABAA ) and glycine receptors, but not by the selective glycine receptor antagonist strychnine, which suggests an involvement of GABAA receptors. Ethanol did not affect spontaneous inhibitory postsynaptic currents suggesting that the inhibition of BLA neuronal excitability by ethanol was not due to an increase in GABAA -mediated synaptic transmission. However, acute ethanol enhanced the amplitude of the holding current of BLA neurons, an effect that was prevented by picrotoxin, which by itself reduced the holding current. CONCLUSIONS These results suggest that BLA neurons express a GABA-mediated tonic current that is enhanced by acute ethanol, which leads to reduced excitability of BLA neurons.
Collapse
Affiliation(s)
| | - John J. Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
- Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
8
|
Fish KN, Joffe ME. Targeting prefrontal cortex GABAergic microcircuits for the treatment of alcohol use disorder. Front Synaptic Neurosci 2022; 14:936911. [PMID: 36105666 PMCID: PMC9465392 DOI: 10.3389/fnsyn.2022.936911] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Developing novel treatments for alcohol use disorders (AUDs) is of paramount importance for improving patient outcomes and alleviating the suffering related to the disease. A better understanding of the molecular and neurocircuit mechanisms through which alcohol alters brain function will be instrumental in the rational development of new efficacious treatments. Clinical studies have consistently associated the prefrontal cortex (PFC) function with symptoms of AUDs. Population-level analyses have linked the PFC structure and function with heavy drinking and/or AUD diagnosis. Thus, targeting specific PFC cell types and neural circuits holds promise for the development of new treatments. Here, we overview the tremendous diversity in the form and function of inhibitory neuron subtypes within PFC and describe their therapeutic potential. We then summarize AUD population genetics studies, clinical neurophysiology findings, and translational neuroscience discoveries. This study collectively suggests that changes in fast transmission through PFC inhibitory microcircuits are a central component of the neurobiological effects of ethanol and the core symptoms of AUDs. Finally, we submit that there is a significant and timely need to examine sex as a biological variable and human postmortem brain tissue to maximize the efforts in translating findings to new clinical treatments.
Collapse
Affiliation(s)
| | - Max E. Joffe
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Single-dose ethanol intoxication causes acute and lasting neuronal changes in the brain. Proc Natl Acad Sci U S A 2022; 119:e2122477119. [PMID: 35700362 DOI: 10.1073/pnas.2122477119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alcohol intoxication at early ages is a risk factor for the development of addictive behavior. To uncover neuronal molecular correlates of acute ethanol intoxication, we used stable-isotope-labeled mice combined with quantitative mass spectrometry to screen more than 2,000 hippocampal proteins, of which 72 changed synaptic abundance up to twofold after ethanol exposure. Among those were mitochondrial proteins and proteins important for neuronal morphology, including MAP6 and ankyrin-G. Based on these candidate proteins, we found acute and lasting molecular, cellular, and behavioral changes following a single intoxication in alcohol-naïve mice. Immunofluorescence analysis revealed a shortening of axon initial segments. Longitudinal two-photon in vivo imaging showed increased synaptic dynamics and mitochondrial trafficking in axons. Knockdown of mitochondrial trafficking in dopaminergic neurons abolished conditioned alcohol preference in Drosophila flies. This study introduces mitochondrial trafficking as a process implicated in reward learning and highlights the potential of high-resolution proteomics to identify cellular mechanisms relevant for addictive behavior.
Collapse
|
10
|
Richard Y, Tazi N, Frydecka D, Hamid MS, Moustafa AA. A systematic review of neural, cognitive, and clinical studies of anger and aggression. CURRENT PSYCHOLOGY 2022; 42:1-13. [PMID: 35693838 PMCID: PMC9174026 DOI: 10.1007/s12144-022-03143-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 01/23/2023]
Abstract
Anger and aggression have large impact on people's safety and the society at large. In order to provide an intervention to minimise aggressive behaviours, it is important to understand the neural and cognitive aspects of anger and aggression. In this systematic review, we investigate the cognitive and neural aspects of anger-related processes, including anger-related behaviours and anger reduction. Using this information, we then review prior existing methods on the treatment of anger-related disorders as well as anger management, including mindfulness and cognitive behavioural therapy. At the cognitive level, our review that anger is associated with excessive attention to anger-related stimuli and impulsivity. At the neural level, anger is associated with abnormal functioning of the amygdala and ventromedial prefrontal cortex. In conclusions, based on cognitive and neural studies, we here argue that mindfulness based cognitive behavioural therapy may be better at reducing anger and aggression than other behavioural treatments, such as cognitive behavioural therapy or mindfulness alone. We provide key information on future research work and best ways to manage anger and reduce aggression. Importantly, future research should investigate how anger related behaviours is acquired and how stress impacts the development of anger.
Collapse
Affiliation(s)
| | - Nadia Tazi
- Arabian Gulf University, Manama, Bahrain
- Universite Med 5th, Rabat, Morocco
| | - Dorota Frydecka
- Department of Psychiatry, Wroclaw Medical University, Pasteur Street 10, 50-367 Wroclaw, Poland
| | | | - Ahmed A. Moustafa
- Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, 2193 South Africa
- School of Psychology, Faculty of Society and Design, Bond University, Gold Coast, QLD Australia
| |
Collapse
|
11
|
Crofton EJ, Zhu M, Curtis KN, Nolan GW, O’Buckley TK, Morrow AL, Herman MA. Medial prefrontal cortex-basolateral amygdala circuit dysfunction in chronic alcohol-exposed male rats. Neuropharmacology 2022; 205:108912. [PMID: 34883134 PMCID: PMC8800149 DOI: 10.1016/j.neuropharm.2021.108912] [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: 08/18/2021] [Revised: 11/14/2021] [Accepted: 11/30/2021] [Indexed: 10/19/2022]
Abstract
Alcohol is a commonly used drug that can produce alcohol use disorders (AUDs). Few individuals with AUDs receive treatment and treatment options are complicated by issues with effectiveness and compliance. Alcohol has been shown to differentially affect specific brain regions and an improved understanding of circuit-specific dysregulation caused by alcohol is warranted. Previous work has implicated both the medial prefrontal cortex (mPFC) and basolateral amygdala (BLA) in alcohol-associated plasticity, however studies directly examining the impact of alcohol exposure on this circuit are lacking. The current study employed an optogenetic strategy to investigate the prelimbic mPFC to BLA circuit and changes in circuit activity following chronic intragastric ethanol exposure in male Sprague Dawley rats. We observed monosynaptic connections with light-evoked stimulation of mPFC terminals in the BLA with efficacy and short latency. We also found that mPFC-BLA projections are primarily glutamatergic under basal inhibitory control, with a lesser population of GABAergic projections. We examined optically-evoked glutamate currents in the BLA using repeated trains of stimulation that displayed accommodation, or a reduction in evoked current amplitude over repeated stimulations. We found that following chronic ethanol exposure mPFC-BLA glutamatergic connections were dysregulated such that there were decreases in overall function, notably in synaptic strength and accommodation, with no change in probability of evoked glutamate release. The lesser GABAergic component of the mPFC-BLA circuit was not altered by chronic ethanol exposure. Collectively these data indicate that mPFC-BLA circuitry is a significant target of alcohol-associated plasticity, which may contribute to pathological behavior associated with AUDs.
Collapse
Affiliation(s)
- Elizabeth J. Crofton
- Department of Psychiatry, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Department of Pharmacology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Department of Psychology and Neuroscience, Emmanuel College, Boston, MA, USA
| | - ManHua Zhu
- Department of Pharmacology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Neuroscience Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katelin N. Curtis
- Department of Psychology and Neuroscience, Emmanuel College, Boston, MA, USA
| | - Gavin W. Nolan
- Department of Psychology and Neuroscience, Emmanuel College, Boston, MA, USA
| | - Todd K. O’Buckley
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - A. Leslie Morrow
- Department of Psychiatry, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Department of Pharmacology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - Melissa A. Herman
- Department of Pharmacology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
12
|
León BE, Kang S, Franca-Solomon G, Shang P, Choi DS. Alcohol-Induced Neuroinflammatory Response and Mitochondrial Dysfunction on Aging and Alzheimer's Disease. Front Behav Neurosci 2022; 15:778456. [PMID: 35221939 PMCID: PMC8866940 DOI: 10.3389/fnbeh.2021.778456] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/07/2021] [Indexed: 12/27/2022] Open
Abstract
Mitochondria are essential organelles central to various cellular functions such as energy production, metabolic pathways, signaling transduction, lipid biogenesis, and apoptosis. In the central nervous system, neurons depend on mitochondria for energy homeostasis to maintain optimal synaptic transmission and integrity. Deficiencies in mitochondrial function, including perturbations in energy homeostasis and mitochondrial dynamics, contribute to aging, and Alzheimer's disease. Chronic and heavy alcohol use is associated with accelerated brain aging, and increased risk for dementia, especially Alzheimer's disease. Furthermore, through neuroimmune responses, including pro-inflammatory cytokines, excessive alcohol use induces mitochondrial dysfunction. The direct and indirect alcohol-induced neuroimmune responses, including pro-inflammatory cytokines, are critical for the relationship between alcohol-induced mitochondrial dysfunction. In the brain, alcohol activates microglia and increases inflammatory mediators that can impair mitochondrial energy production, dynamics, and initiate cell death pathways. Also, alcohol-induced cytokines in the peripheral organs indirectly, but synergistically exacerbate alcohol's effects on brain function. This review will provide recent and advanced findings focusing on how alcohol alters the aging process and aggravates Alzheimer's disease with a focus on mitochondrial function. Finally, we will contextualize these findings to inform clinical and therapeutic approaches towards Alzheimer's disease.
Collapse
Affiliation(s)
- Brandon Emanuel León
- Regenerative Sciences Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Gabriela Franca-Solomon
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Pei Shang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
13
|
Yildirim S, Ozkan A, Aytac G, Agar A, Tanriover G. Role of melatonin in TLR4-mediated inflammatory pathway in the MTPT-induced mouse model. Neurotoxicology 2021; 88:168-177. [PMID: 34808223 DOI: 10.1016/j.neuro.2021.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/08/2021] [Accepted: 11/17/2021] [Indexed: 12/21/2022]
Abstract
Neuroinflammation has an essential role in various neurodegenerative diseases including Parkinson's disease (PD). Microglial activation as a result of neuroinflammation exacerbates the pathological consequences of the disease. The toxic effect of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) causes alpha-synuclein (α-synuclein) accumulation, which leads to dopaminergic neuron death in the MPTP-induced mouse model. Toll-like receptor 4 (TLR4) stimulates release of cytokine through NF-kB by activating glial cells, thus resulting in the death of dopaminergic neurons. Melatonin has the ability to cross the blood-brain barrier and protect neurons through anti-inflammatory properties. We hypothesized that melatonin could suppress TLR4-mediated neuroinflammation, decrease cytokine release due to the inflammatory response, and reduce dopaminergic neuron loss in the MPTP-induced mouse model. In the MPTP-induced mouse model, we aimed to assess the neuroinflammatory responses caused by TLR4 activation as well as the effect of melatonin on these responses. Three-month-old male C57BL/6 mice were randomly divided into five groups; Control (Group-C), Sham (Group-S), Melatonin-treated (Group-M), MPTP-injected (Group-P), and MPTP + melatonin-injected (Group-P + M). MPTP toxin (20 mg/kg) was dissolved in saline and intraperitoneally (i.p.) injected to mice for two days with 12 h intervals. The total dose per mouse was 80 mg/kg. Melatonin was administered (20 mg/kg) intraperitoneally to Group-M and Group-P + M twice a day for five days. Eight days after starting the experiment, the motor activities of mice were evaluated by locomotor activity tests. The effects on dopamine neurons in the SNPc was determined by tyrosine hydroxylase (TH) immunohistochemistry. TLR4, α-synuclein, and p65 expression was evaluated by immunostaining as well. The amount of TNF-alpha in the total brain was evaluated by western blot analysis. In our results seen that locomotor activity was lower in Group-P compared to Group-C. However, melatonin administration was improved this impairment. MPTPcaused decrease in TH immuno-expression in dopaminergic neurons in Group-P. TLR4 (p < 0.001), α-synuclein (p < 0.001), and p65 (p < 0.01) immuno-expressions were also decreased in Group-P+M compared to Group-P (using MPTP). TNF-α expression was lower in Group-C, Group-S, Group-M, and Group-P+M, when compared to Group-P (p < 0.0001) due to the absence of inflammatory response. In conclusion, our study revealed that melatonin administration reduced α-synuclein aggregation and TLR4-mediated inflammatory response in the MPTP-induced mouse model.
Collapse
Affiliation(s)
- Sendegul Yildirim
- Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, Antalya, Turkey
| | - Ayse Ozkan
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| | - Gunes Aytac
- TOBB University of Economics & Technology, Faculty of Medicine, Department of Anatomy, Ankara, Turkey; University of Hawai'i at Mānoa, John A. Burns School of Medicine, Department of Anatomy, Biochemistry & Physiology, Hawaii, USA
| | - Aysel Agar
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| | - Gamze Tanriover
- Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, Antalya, Turkey; Akdeniz University, Faculty of Medicine, Department of Medical Biotechnology, Antalya, Turkey.
| |
Collapse
|
14
|
Morningstar MD, Barnett WH, Goodlett CR, Kuznetsov A, Lapish CC. Understanding ethanol's acute effects on medial prefrontal cortex neural activity using state-space approaches. Neuropharmacology 2021; 198:108780. [PMID: 34480911 PMCID: PMC8488975 DOI: 10.1016/j.neuropharm.2021.108780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/10/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022]
Abstract
Acute ethanol (EtOH) intoxication results in several maladaptive behaviors that may be attributable, in part, to the effects of EtOH on neural activity in medial prefrontal cortex (mPFC). The acute effects of EtOH on mPFC function have been largely described as inhibitory. However, translating these observations on function into a mechanism capable of delineating acute EtOH's effects on behavior has proven difficult. This review highlights the role of acute EtOH on electrophysiological measurements of mPFC function and proposes that interpreting these changes through the lens of dynamical systems theory is critical to understand the mechanisms that mediate the effects of EtOH intoxication on behavior. Specifically, the present review posits that the effects of EtOH on mPFC N-methyl-d-aspartate (NMDA) receptors are critical for the expression of impaired behavior following EtOH consumption. This hypothesis is based on the observation that recurrent activity in cortical networks is supported by NMDA receptors, and, when disrupted, may lead to impairments in cognitive function. To evaluate this hypothesis, we discuss the representation of mPFC neural activity in low-dimensional, dynamic state spaces. This approach has proven useful for identifying the underlying computations necessary for the production of behavior. Ultimately, we hypothesize that EtOH-related alterations to NMDA receptor function produces alterations that can be effectively conceptualized as impairments in attractor dynamics and provides insight into how acute EtOH disrupts forms of cognition that rely on mPFC function. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
Affiliation(s)
| | - William H Barnett
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA
| | - Charles R Goodlett
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| | - Alexey Kuznetsov
- Indiana University-Purdue University Indianapolis, Department of Mathematics, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| | - Christopher C Lapish
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| |
Collapse
|
15
|
Nunes-Freitas AL, Soni N, Polli FS, Kohlmeier KA. Prenatal exposure to nicotine in mice is associated with alterations in development and cellular and synaptic effects of alcohol in a brainstem arousal nucleus. Neurotoxicol Teratol 2021; 87:106980. [PMID: 33838245 DOI: 10.1016/j.ntt.2021.106980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023]
Abstract
Using drugs of abuse while pregnant has tremendous negative consequences for the offspring, including an enhanced risk for substance use disorder (SUD). This vulnerability suggests that gestational exposure to drugs alters the developmental trajectory of neurons important in SUD processes, which could lead to later life changes in responsiveness to motivationally salient stimuli. The laterodorsal tegmentum (LDT) gates the behaviorally relevant firing pattern signaling stimuli saliency in mesoaccumbal circuits. Accordingly, any alterations in LDT functionality could alter output, and play a role in negative outcomes on motivated behavior associated with early-life nicotine exposure. Therefore, we investigated whether prenatal exposure to nicotine (PNE), which is a known teratogen, altered responsiveness of LDT neurons to alcohol by conducting electrophysiology in brain slices. Alcohol induced an outward current in control LDT cells, which was not seen in PNE LDT neurons. The frequency of mEPSCs was significantly decreased by alcohol in LDT PNE cells and accompanied by a decrease in action potential frequency, which were actions not seen in controls. Changes in baseline activity of PNE LDT cells were also observed. In summary, PNE LDT neurons showed alterations in baseline activity and membrane and synaptic responses to postnatal exposures to alcohol. The differences in PNE baseline activity and alcohol responses likely lead to differential output from the LDT to mesoaccumbal targets that could play a role in biasing coding of relevant stimuli, which could participate in the enhanced proclivity for development of SUD in those exposed during gestation to nicotine.
Collapse
Affiliation(s)
- André Luiz Nunes-Freitas
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark; Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Neeraj Soni
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Filip S Polli
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kristi A Kohlmeier
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark.
| |
Collapse
|
16
|
Avchalumov Y, Oliver RJ, Trenet W, Heyer Osorno RE, Sibley BD, Purohit DC, Contet C, Roberto M, Woodward JJ, Mandyam CD. Chronic ethanol exposure differentially alters neuronal function in the medial prefrontal cortex and dentate gyrus. Neuropharmacology 2021; 185:108438. [PMID: 33333103 PMCID: PMC7927349 DOI: 10.1016/j.neuropharm.2020.108438] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 12/06/2020] [Accepted: 12/09/2020] [Indexed: 12/28/2022]
Abstract
Alterations in the function of prefrontal cortex (PFC) and hippocampus have been implicated in underlying the relapse to alcohol seeking behaviors in humans and animal models of moderate to severe alcohol use disorders (AUD). Here we used chronic intermittent ethanol vapor exposure (CIE), 21d protracted abstinence following CIE (21d AB), and re-exposure to one vapor session during protracted abstinence (re-exposure) to evaluate the effects of chronic ethanol exposure on basal synaptic function, neuronal excitability and expression of key synaptic proteins that play a role in neuronal excitability in the medial PFC (mPFC) and dentate gyrus (DG). CIE consistently enhanced excitability of layer 2/3 pyramidal neurons in the mPFC and granule cell neurons in the DG. In the DG, this effect persisted during 21d AB. Re-exposure did not enhance excitability, suggesting resistance to vapor-induced effects. Analysis of action potential kinetics revealed that altered afterhyperpolarization, rise time and decay time constants are associated with the altered excitability during CIE, 21d AB and re-exposure. Molecular adaptations that may underlie increases in neuronal excitability under these different conditions were identified. Quantitative polymerase chain reaction of large-conductance potassium (BK) channel subunit mRNA in PFC and DG tissue homogenates did not show altered expression patterns of BK subunits. Western blotting demonstrates enhanced phosphorylation of Ca2⁺/calmodulin-dependent protein kinase II (CaMKII), and reduced phosphorylation of glutamate receptor GluN2A/2B subunits. These results suggest a novel relationship between activity of CaMKII and GluN receptors in the mPFC and DG, and neuronal excitability in these brain regions in the context of moderate to severe AUD.
Collapse
Affiliation(s)
| | | | - Wulfran Trenet
- VA San Diego Healthcare System, San Diego, CA, 92161, USA
| | | | | | | | - Candice Contet
- Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, 92037, USA
| | - Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, 92037, USA
| | - John J Woodward
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC, USA
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, 92161, USA; Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, 92037, USA; Department of Anesthesiology, University of California San Diego, San Diego, CA, 92161, USA.
| |
Collapse
|
17
|
Li M, Cabrera-Garcia D, Salling MC, Au E, Yang G, Harrison NL. Alcohol reduces the activity of somatostatin interneurons in the mouse prefrontal cortex: A neural basis for its disinhibitory effect? Neuropharmacology 2021; 188:108501. [PMID: 33636191 DOI: 10.1016/j.neuropharm.2021.108501] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The prefrontal cortex (PFC) is involved in executive ("top-down") control of behavior and its function is especially susceptible to the effects of alcohol, leading to behavioral disinhibition that is associated with alterations in decision making, response inhibition, social anxiety and working memory. The circuitry of the PFC involves a complex interplay between pyramidal neurons (PNs) and several subclasses of inhibitory interneurons (INs), including somatostatin (SST)-expressing INs. Using in vivo calcium imaging, we showed that alcohol dose-dependently altered network activity in layers 2/3 of the prelimbic subregion of the mouse PFC. Low doses of alcohol (1 g/kg, intraperitoneal, i.p.) caused moderate activation of SST INs and weak inhibition of PNs. At moderate to high doses, alcohol (2-3 g/kg) strongly inhibited the activity of SST INs in vivo, and this effect may result in disinhibition, as the activity of a subpopulation of PNs was simultaneously enhanced. In contrast, recordings in brain slices using ex vivo electrophysiology revealed no direct effect of alcohol on the excitability of either SST INs or PNs over a range of concentrations (20 and 50 mM) consistent with the blood alcohol levels reached in the in vivo experiments. This dose-dependent effect of alcohol on SST INs in vivo may reveal a neural basis for the disinhibitory effect of alcohol in the PFC mediated by other neurons within or external to the PFC circuitry.
Collapse
Affiliation(s)
- Miao Li
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - David Cabrera-Garcia
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - Michael C Salling
- Louisiana State University, Department of Anatomy, New Orleans, LA, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Edmund Au
- Columbia University, Department of Pathology & Cell Biology and Rehabilitative Medicine and Regeneration, Columbia Translational Neuroscience Initiative Scholar, 630 West 168th Street, New York, NY, 10032, USA
| | - Guang Yang
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA.
| | - Neil L Harrison
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA; Columbia University, Department of Molecular Pharmacology and Therapeutics, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
18
|
Maldonado R, Calvé P, García-Blanco A, Domingo-Rodriguez L, Senabre E, Martín-García E. Vulnerability to addiction. Neuropharmacology 2021; 186:108466. [PMID: 33482225 DOI: 10.1016/j.neuropharm.2021.108466] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 01/14/2021] [Indexed: 12/22/2022]
Abstract
Addiction is a chronic brain disease that has dramatic health and socioeconomic consequences worldwide. Multiple approaches have been used for decades to clarify the neurobiological basis of this disease and to identify novel potential treatments. This review summarizes the main brain networks involved in the vulnerability to addiction and specific innovative technological approaches to investigate these neural circuits. First, the evolution of the definition of addiction across the Diagnostic and Statistical Manual of Mental Disorders (DSM) is revised. We next discuss several innovative experimental techniques that, combined with behavioral approaches, have allowed recent critical advances in understanding the neural circuits involved in addiction, including DREADDs, calcium imaging, and electrophysiology. All these techniques have been used to investigate specific neural circuits involved in vulnerability to addiction and have been extremely useful to clarify the neurobiological basis of each specific component of the addictive process. These novel tools targeting specific brain regions are of great interest to further understand the different aspects of this complex disease. This article is part of the special issue on 'Vulnerabilities to Substance Abuse.'.
Collapse
Affiliation(s)
- R Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain; Hospital Del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| | - P Calvé
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - A García-Blanco
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - L Domingo-Rodriguez
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - E Senabre
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - E Martín-García
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
19
|
Abstract
Alcohol is one of the oldest pharmacological agents used for its sedative/hypnotic effects, and alcohol abuse and alcohol use disorder (AUD) continues to be major public health issue. AUD is strongly indicated to be a brain disorder, and the molecular and cellular mechanism/s by which alcohol produces its effects in the brain are only now beginning to be understood. In the brain, synaptic plasticity or strengthening or weakening of synapses, can be enhanced or reduced by a variety of stimulation paradigms. Synaptic plasticity is thought to be responsible for important processes involved in the cellular mechanisms of learning and memory. Long-term potentiation (LTP) is a form of synaptic plasticity, and occurs via N-methyl-D-aspartate type glutamate receptor (NMDAR or GluN) dependent and independent mechanisms. In particular, NMDARs are a major target of alcohol, and are implicated in different types of learning and memory. Therefore, understanding the effect of alcohol on synaptic plasticity and transmission mediated by glutamatergic signaling is becoming important, and this will help us understand the significant contribution of the glutamatergic system in AUD. In the first part of this review, we will briefly discuss the mechanisms underlying long term synaptic plasticity in the dorsal striatum, neocortex and the hippocampus. In the second part we will discuss how alcohol (ethanol, EtOH) can modulate long term synaptic plasticity in these three brain regions, mainly from neurophysiological and electrophysiological studies. Taken together, understanding the mechanism(s) underlying alcohol induced changes in brain function may lead to the development of more effective therapeutic agents to reduce AUDs.
Collapse
Affiliation(s)
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
20
|
Morningstar MD, Linsenbardt DN, Lapish CC. Ethanol Alters Variability, But Not Rate, of Firing in Medial Prefrontal Cortex Neurons of Awake-Behaving Rats. Alcohol Clin Exp Res 2020; 44:2225-2238. [PMID: 32966634 PMCID: PMC7680402 DOI: 10.1111/acer.14463] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/12/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) is a brain region involved in the evaluation and selection of motivationally relevant outcomes. Neural activity in mPFC is altered following acute ethanol (EtOH) use and, in rodent models, doses as low as 0.75 g/kg yield cognitive deficits. Deficits in decision making following acute EtOH are thought to be mediated, at least in part, by decreases in mPFC firing rates (FRs). However, the data leading to this conclusion have been generated exclusively in anesthetized rodents. The present study characterizes the effects of acute EtOH injections on mPFC neural activity in awake-behaving rodents. METHODS Awake-behaving and anesthetized in vivo electrophysiological recordings were performed. We utilized 3 groups: the first received 2 saline injections, the second received a saline injection followed by 1.0 g/kg EtOH, and the last received saline followed by 2 g/kg EtOH. One week later, an anesthetized recording occurred where a saline injection was followed by an injection of 1.0 g/kg EtOH. RESULTS The anesthetized condition showed robust decreases in neural activity and differences in up-down states (UDS) dynamics. In the awake-behaving condition, FRs were grouped according to behavioral state: moving, not-moving, and sleep. The differences in median FRs were found for each treatment and behavioral state combination. A FR decrease was only found in the 2.0 g/kg EtOH treatment during not-moving states. However, robust decreases in FR variability were found across behavioral state in both the 1.0 and 2.0 g/kg EtOH treatment. Sleep was separately analyzed. EtOH modulated the UDS during sleep producing decreases in FRs. CONCLUSIONS In conclusion, the changes in neural activity following EtOH administration in anesthetized animals are not conserved in awake-behaving animals. The most prominent difference following EtOH was a decrease in FR variability suggesting that acute EtOH may be affecting decision making via this mechanism.
Collapse
|
21
|
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: 2.2] [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
|
22
|
Shang P, Lindberg D, Starski P, Peyton L, Hong SI, Choi S, Choi DS. Chronic Alcohol Exposure Induces Aberrant Mitochondrial Morphology and Inhibits Respiratory Capacity in the Medial Prefrontal Cortex of Mice. Front Neurosci 2020; 14:561173. [PMID: 33192248 PMCID: PMC7646256 DOI: 10.3389/fnins.2020.561173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Abstract
Alcohol use disorder (AUD) is characterized as a chronic, relapsing disease with a pattern of excessive drinking despite negative consequences to an individual's life. Severe chronic alcohol use impairs the function of the medial prefrontal cortex (mPFC), which contributes to alcohol-induced cognitive and executive dysfunction. The mPFC contains more mitochondria compared to other cortical areas, which suggests mitochondrial damage may occur in AUD and trigger subsequent behavior change. Here, we identified morphological and functional changes in mitochondria in the mPFC in C57BL6/J mice after 8 h of withdrawal from chronic intermittent alcohol (CIA) exposure. Three-dimensional serial block-face scanning electron microscopy (SBFSEM) reconstruction revealed that CIA exposure elongated mPFC mitochondria and formed mitochondria-on-a-string (MOAS). Furthermore, alcohol significantly affected mitochondrial bioenergetics, including oxidative phosphorylation and electron transport, with inhibited aerobic respiration in mPFC mitochondria after CIA exposure. We also found decreased expression of fusion (mitofusin 2, Mfn2) and increased fission (mitochondrial fission 1 protein, Fis1) proteins in the mPFC of alcohol-treated mice. In sum, our study suggests that CIA exposure impairs mitochondrial dynamics and function in the mPFC.
Collapse
Affiliation(s)
- Pei Shang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Neurology, First Hospital of Jilin University, Changchun, China
| | - Daniel Lindberg
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Phillip Starski
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sun Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
23
|
Hughes BA, Crofton EJ, O'Buckley TK, Herman MA, Morrow AL. Chronic ethanol exposure alters prelimbic prefrontal cortical Fast-Spiking and Martinotti interneuron function with differential sex specificity in rat brain. Neuropharmacology 2020; 162:107805. [PMID: 31589884 PMCID: PMC7027948 DOI: 10.1016/j.neuropharm.2019.107805] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/17/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
Chronic ethanol exposure results in numerous neurobiological adaptations that promote deficits in medial prefrontal cortical (mPFC) function associated with blunted inhibitory control and elevated anxiety during withdrawal. Studies exploring alcohol dependence-related changes in this region have largely investigated adaptations in glutamatergic signaling, with inhibitory neurotransmission remaining relatively understudied. To address this, we used biochemical and electrophysiological methods to evaluate the effects of ethanol on the activity of deep-layer prelimbic mPFC Fast-Spiking (FS) and Martinotti interneurons after chronic ethanol exposure in male and female rats. We report that chronic alcohol exposure significantly impairs FS neuron excitability in both males and females. Interestingly, we observed a marked sex difference in the baseline activity of Martinotti cells that furthermore displayed differential sex-specific responses to alcohol exposure. In addition, alcohol effects on Martinotti neuron excitability negatively correlated with hyperpolarization-activated currents mediated by hyperpolarization-activated cyclic nucleotide gated (HCN) channels, indicative of a causal relationship. Analysis of HCN1 protein expression also revealed a substantial sex difference, although no effect of ethanol on HCN1 protein expression was observed. Taken together, these findings further elucidate the complex adaptations that occur in the mPFC after chronic ethanol exposure and reveal fundamental differences in interneuron activity between sexes. Furthermore, this disparity may reflect innate differences in intracortical microcircuit function between male and female rats, and offers a tenable circuit-level explanation for sex-dependent behavioral responses to alcohol.
Collapse
Affiliation(s)
- Benjamin A Hughes
- Department of Psychiatry, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA
| | - Elizabeth J Crofton
- Department of Psychiatry, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA
| | - Todd K O'Buckley
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA
| | - Melissa A Herman
- Department of Pharmacology, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA
| | - A Leslie Morrow
- Department of Psychiatry, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, 27599, NC, USA.
| |
Collapse
|
24
|
Petruccelli E, Feyder M, Ledru N, Jaques Y, Anderson E, Kaun KR. Alcohol Activates Scabrous-Notch to Influence Associated Memories. Neuron 2018; 100:1209-1223.e4. [PMID: 30482693 DOI: 10.1016/j.neuron.2018.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 08/17/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Drugs of abuse, like alcohol, modulate gene expression in reward circuits and consequently alter behavior. However, the in vivo cellular mechanisms through which alcohol induces lasting transcriptional changes are unclear. We show that Drosophila Notch/Su(H) signaling and the secreted fibrinogen-related protein Scabrous in mushroom body (MB) memory circuitry are important for the enduring preference of cues associated with alcohol's rewarding properties. Alcohol exposure affects Notch responsivity in the adult MB and alters Su(H) targeting at the dopamine-2-like receptor (Dop2R). Alcohol cue training also caused lasting changes to the MB nuclear transcriptome, including changes in the alternative splicing of Dop2R and newly implicated transcripts like Stat92E. Together, our data suggest that alcohol-induced activation of the highly conserved Notch pathway and accompanying transcriptional responses in memory circuitry contribute to addiction. Ultimately, this provides mechanistic insight into the etiology and pathophysiology of alcohol use disorder.
Collapse
Affiliation(s)
- Emily Petruccelli
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Michael Feyder
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Nicolas Ledru
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Yanabah Jaques
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Edward Anderson
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Karla R Kaun
- Department of Neuroscience, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
25
|
Role of glutamatergic system and mesocorticolimbic circuits in alcohol dependence. Prog Neurobiol 2018; 171:32-49. [PMID: 30316901 DOI: 10.1016/j.pneurobio.2018.10.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/08/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
Abstract
Emerging evidence demonstrates that alcohol dependence is associated with dysregulation of several neurotransmitters. Alterations in dopamine, glutamate and gamma-aminobutyric acid release are linked to chronic alcohol exposure. The effects of alcohol on the glutamatergic system in the mesocorticolimbic areas have been investigated extensively. Several studies have demonstrated dysregulation in the glutamatergic systems in animal models exposed to alcohol. Alcohol exposure can lead to an increase in extracellular glutamate concentrations in mesocorticolimbic brain regions. In addition, alcohol exposure affects the expression and functions of several glutamate receptors and glutamate transporters in these brain regions. In this review, we discussed the effects of alcohol exposure on glutamate receptors, glutamate transporters and glutamate homeostasis in each area of the mesocorticolimbic system. In addition, we discussed the genetic aspect of alcohol associated with glutamate and reward circuitry. We also discussed the potential therapeutic role of glutamate receptors and glutamate transporters in each brain region for the treatment of alcohol dependence. Finally, we provided some limitations on targeting the glutamatergic system for potential therapeutic options for the treatment alcohol use disorders.
Collapse
|
26
|
Alcohol Consumption during Adolescence in a Mouse Model of Binge Drinking Alters the Intrinsic Excitability and Function of the Prefrontal Cortex through a Reduction in the Hyperpolarization-Activated Cation Current. J Neurosci 2018; 38:6207-6222. [PMID: 29915134 DOI: 10.1523/jneurosci.0550-18.2018] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/24/2018] [Accepted: 05/26/2018] [Indexed: 12/20/2022] Open
Abstract
Periodic episodes of excessive alcohol consumption ("binge drinking") occur frequently among adolescents, and early binge drinking is associated with an increased risk of alcohol use disorders later in life. The PFC undergoes significant development during adolescence and hence may be especially susceptible to the effects of binge drinking. In humans and in animal models, adolescent alcohol exposure is known to alter PFC neuronal activity and produce deficits in PFC-dependent behaviors, such as decision making, response inhibition, and working memory. Using a voluntary intermittent access to alcohol (IA EtOH) procedure in male mice, we demonstrate that binge-level alcohol consumption during adolescence leads to altered drinking patterns and working memory deficits in young adulthood, two outcomes that suggest medial PFC dysfunction. We recorded from pyramidal neurons (PNs) in the prelimbic subregion of the medial PFC in slices obtained from mice that had IA EtOH and found that they display altered excitability, including a hyperpolarization of the resting membrane potential and reductions in the hyperpolarization-activated cation current (Ih) and in intrinsic persistent activity (a mode of neuronal firing that is dependent on Ih). Many of these effects on intrinsic excitability were sustained following abstinence and observed in mice that showed working memory deficits. In addition, we found that resting membrane potential and the Ih-dependent voltage "sag" in prelimbic PFC PNs are developmentally regulated during adolescence, suggesting that adolescent alcohol exposure may compromise PFC function by arresting the normal developmental trajectory of PN intrinsic excitability.SIGNIFICANCE STATEMENT Binge alcohol drinking during adolescence has negative consequences for the function of the developing PFC. Using a mouse model of voluntary binge drinking during adolescence, we found that this behavior leads to working memory deficits and altered drinking behavior in adulthood. In addition, we found that adolescent drinking is associated with specific changes to the intrinsic excitability of pyramidal neurons in the PFC, reducing the ability of these neurons to generate intrinsic persistent activity, a phenomenon thought to be important for working memory. These findings may help explain why human adolescent binge drinkers show performance deficits on tasks mediated by the PFC.
Collapse
|
27
|
Histone deacetylases mediate GABA A receptor expression, physiology, and behavioral maladaptations in rat models of alcohol dependence. Neuropsychopharmacology 2018; 43. [PMID: 29520058 PMCID: PMC5983537 DOI: 10.1038/s41386-018-0034-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Alcohol use disorders are chronic debilitating diseases characterized by severe withdrawal symptoms that contribute to morbidity and relapse. GABAA receptor (GABAAR) adaptations have long been implicated in the chronic effects of alcohol and contribute to many withdrawal symptoms associated with alcohol dependence. In rodents, GABAAR hypofunction results from decreases in Gabra1 expression, although the underlying mechanism controlling Gabra1 expression after chronic ethanol exposure is still unknown. We found that chronic ethanol exposure using either ethanol gavage or two-bottle choice voluntary access paradigms decreased Gabra1 expression and increased Hdac2 and Hdac3 expression. Administration of the HDAC inhibitor trichostatin A (TSA) after chronic ethanol exposure prevents the decrease in Gabra1 expression and function as well as the increase in Hdac2 and Hdac3 expression in both the cortex and the medial prefrontal cortex (mPFC). Chronic ethanol exposure and withdrawal, but not acute ethanol exposure or acute withdrawal, cause a selective upregulation of HDAC2 and HDAC3 associated with the Gabra1 promoter that accompanies a decrease in H3 acetylation of the Gabra1 promoter and the reduction in GABAAR α1 subunit expression. TSA administration prevented each of these molecular events as well as behavioral manifestations of ethanol dependence, including tolerance to zolpidem-induced loss of righting reflex, reduced open-arm time in the elevated plus maze, reduced center-time and locomotor activity in the open-field assay, and TSA reduced voluntary ethanol consumption. The results show how chronic ethanol exposure regulates the highly prominent GABAAR α1 subunit by an epigenetic mechanism that represents a potential treatment modality for alcohol dependence.
Collapse
|
28
|
Zamudio-Bulcock PA, Homanics GE, Woodward JJ. Loss of Ethanol Inhibition of N-Methyl-D-Aspartate Receptor-Mediated Currents and Plasticity of Cerebellar Synapses in Mice Expressing the GluN1(F639A) Subunit. Alcohol Clin Exp Res 2018; 42:698-705. [PMID: 29323417 DOI: 10.1111/acer.13597] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Glutamatergic N-methyl-d-aspartate receptors (NMDARs) are well known for their sensitivity to ethanol (EtOH) inhibition. However, the specific manner in which EtOH inhibits channel activity and how such inhibition affects neurotransmission, and ultimately behavior, remains unclear. Replacement of phenylalanine 639 with alanine (F639A) in the GluN1 subunit reduces EtOH inhibition of recombinant NMDARs. Mice expressing this subunit show reduced EtOH-induced anxiolysis, blunted locomotor stimulation following low-dose EtOH administration, and faster recovery of motor function after moderate doses of EtOH, suggesting that cerebellar dysfunction may contribute to some of these behaviors. In the mature mouse cerebellum, NMDARs at the cerebellar climbing fiber (CF) to Purkinje cell (PC) synapse are inhibited by low concentrations of EtOH and the long-term depression (LTD) of parallel fiber (PF)-mediated currents induced by concurrent activation of PFs and CFs (PF-LTD) requires activation of EtOH-sensitive NMDARs. In this study, we examined cerebellar NMDA responses and NMDA-mediated synaptic plasticity in wild-type (WT) and GluN1(F639A) mice. METHODS Patch-clamp electrophysiological recordings were performed in acute cerebellar slices from adult WT and GluN1(F639A) mice. NMDAR-mediated currents at the CF-PC synapse and NMDAR-dependent PF-LTD induction were compared for genotype-dependent differences. RESULTS Stimulation of CFs evoked robust NMDA-mediated excitatory postsynaptic currents (EPSCs) in PCs that were similar in amplitude and kinetics between WT and GluN1(F639A) mice. NMDA-mediated CF-PC EPSCs in WT mice were significantly inhibited by EtOH (50 mM) while those in mutant mice were unaffected. Concurrent stimulation of CF and PF inputs induced synaptic depression of PF-PC EPSCs in both WT and mutant mice, and this depression was blocked by the NMDA antagonist DL-APV. The synaptic depression of PF-PC EPSCs in WT mice was also blocked by a low concentration of EtOH (10 mM) that had no effect on plasticity in GluN1(F639A) mice. CONCLUSIONS These results demonstrate that inhibition of cerebellar NMDARs may be a key mechanism by which EtOH affects cerebellar-dependent behaviors.
Collapse
Affiliation(s)
- Paula A Zamudio-Bulcock
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Gregg E Homanics
- Department of Anesthesiology, Univeristy of Pittsburgh, Pittsburgh, PA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
29
|
Klenowski PM. Emerging role for the medial prefrontal cortex in alcohol-seeking behaviors. Addict Behav 2018; 77:102-106. [PMID: 28992574 DOI: 10.1016/j.addbeh.2017.09.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/01/2017] [Accepted: 09/27/2017] [Indexed: 01/01/2023]
Abstract
The medial prefrontal cortex (mPFC) plays an important role in high-order executive processes and sends highly organized projections to sub-cortical regions controlling mood, motivation and impulsivity. Recent preclinical and clinical studies have demonstrated alcohol-induced effects on the activity and composition of the PFC which are implicated in associative learning processes and may disrupt executive control over impulsivity, leading to an inability to self-limit alcohol intake. Animal studies have begun to dissect the role of the mPFC circuitry in alcohol-seeking behavior and withdrawal, and have identified a key role for projections to sub-cortical sites including the extended amygdala and the nucleus accumbens (NAc). Importantly, these studies have highlighted that alcohol can have contrasting effects on the mPFC compared to other addictive substances and also produce differential effects on the structure and activity of the mPFC following short-term versus long-term consumption. Because of these differences, how the mPFC influences the initial aspects of alcohol-seeking behavior and how we can better understand the long-term effects of alcohol use on the activity and connectivity of the mPFC need to be considered. Given the lack of preclinical data from long-term drinking models, an increased focus should be directed towards identifying how long-term alcohol use changes the mPFC, in order to provide new insights into the mechanisms underlying the transition to dependence.
Collapse
|
30
|
Abstract
Major depressive disorder (MDD) is a chronic and potentially life threatening illness that carries a staggering global burden. Characterized by depressed mood, MDD is often difficult to diagnose and treat owing to heterogeneity of syndrome and complex etiology. Contemporary antidepressant treatments are based on improved monoamine-based formulations from serendipitous discoveries made > 60 years ago. Novel antidepressant treatments are necessary, as roughly half of patients using available antidepressants do not see long-term remission of depressive symptoms. Current development of treatment options focuses on generating efficacious antidepressants, identifying depression-related neural substrates, and better understanding the pathophysiological mechanisms of depression. Recent insight into the brain's mesocorticolimbic circuitry from animal models of depression underscores the importance of ionic mechanisms in neuronal homeostasis and dysregulation, and substantial evidence highlights a potential role for ion channels in mediating depression-related excitability changes. In particular, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are essential regulators of neuronal excitability. In this review, we describe seminal research on HCN channels in the prefrontal cortex and hippocampus in stress and depression-related behaviors, and highlight substantial evidence within the ventral tegmental area supporting the development of novel therapeutics targeting HCN channels in MDD. We argue that methods targeting the activity of reward-related brain areas have significant potential as superior treatments for depression.
Collapse
Affiliation(s)
- Stacy M Ku
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
31
|
Shah A, Zuo W, Kang S, Li J, Fu R, Zhang H, Bekker A, Ye JH. The lateral habenula and alcohol: Role of glutamate and M-type potassium channels. Pharmacol Biochem Behav 2017. [PMID: 28624587 DOI: 10.1016/j.pbb.2017.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder (AUD) or alcoholism is a chronic relapsing disorder. Our knowledge of alcoholism hinges on our understanding of its effects on the brain. This review will center on the effects of alcohol in the lateral habenula (LHb), an epithalamic structure that connects the forebrain with the midbrain and encodes aversive signaling. Like many addictive drugs, alcohol has both rewarding and aversive properties. While alcohol's euphoric property is believed to be important for the initiation of drinking, increasing evidence suggests that alcohol's negative affect plays a critical role in excessive drinking and alcohol dependence. During withdrawal and abstinence, alcoholics often experience anxiety and depressions, both of which have been implicated in relapse drinking. This review focuses on the recent accumulation of knowledge about the effects of acute and chronic alcohol exposure on the activity of and synaptic transmissions on LHb neurons, as well as the effects of manipulation of LHb function on alcohol consumption and related behaviors. Recent evidence highlights a critical role for the LHb in AUD and related psychiatric ailments. Multidisciplinary work in animals collectively suggests that LHb function and activity, including M-type potassium channels and glutamatergic transmission are altered by acute and repeated chronic alcohol exposure. We will also discuss how functional, pharmacological, and chemogenetic manipulation of the LHb affects ethanol drinking and psychiatric disorders occurring in animals withdrawn from chronic alcohol exposure. Conceivable mechanisms behind these effects and their potential as targets for therapies will also be discussed.
Collapse
Affiliation(s)
- Avi Shah
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Wanhong Zuo
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Seungwoo Kang
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Jing Li
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Rao Fu
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Haifeng Zhang
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA.
| |
Collapse
|
32
|
Harrison NL, Skelly MJ, Grosserode EK, Lowes DC, Zeric T, Phister S, Salling MC. Effects of acute alcohol on excitability in the CNS. Neuropharmacology 2017; 122:36-45. [PMID: 28479395 DOI: 10.1016/j.neuropharm.2017.04.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 01/23/2023]
Abstract
Alcohol has many effects on brain function and hence on human behavior, ranging from anxiolytic and mild disinhibitory effects, sedation and motor incoordination, amnesia, emesis, hypnosis and eventually unconsciousness. In recent years a variety of studies have shown that acute and chronic exposure to alcohol can modulate ion channels that regulate excitability. Modulation of intrinsic excitability provides another way in which alcohol can influence neuronal network activity, in addition to its actions on synaptic inputs. In this review, we review "low dose" effects [between 2 and 20 mM EtOH], and "medium dose"; effects [between 20 and 50 mM], by considering in turn each of the many networks and brain regions affected by alcohol, and thereby attempt to integrate in vitro physiological studies in specific brain regions (e.g. amygdala, ventral tegmental area, prefrontal cortex, thalamus, cerebellum etc.) within the context of alcohol's behavioral actions in vivo (e.g. anxiolysis, euphoria, sedation, motor incoordination). This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Neil L Harrison
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States.
| | - Mary Jane Skelly
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Emma K Grosserode
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Daniel C Lowes
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Tamara Zeric
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Sara Phister
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Michael C Salling
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| |
Collapse
|
33
|
Delli Pizzi S, Chiacchiaretta P, Mantini D, Bubbico G, Ferretti A, Edden RA, Di Giulio C, Onofrj M, Bonanni L. Functional and neurochemical interactions within the amygdala-medial prefrontal cortex circuit and their relevance to emotional processing. Brain Struct Funct 2017; 222:1267-1279. [PMID: 27566606 PMCID: PMC5549263 DOI: 10.1007/s00429-016-1276-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/15/2016] [Indexed: 02/02/2023]
Abstract
The amygdala-medial prefrontal cortex (mPFC) circuit plays a key role in emotional processing. GABA-ergic inhibition within the mPFC has been suggested to play a role in the shaping of amygdala activity. However, the functional and neurochemical interactions within the amygdala-mPFC circuits and their relevance to emotional processing remain unclear. To investigate this circuit, we obtained resting-state functional magnetic resonance imaging (rs-fMRI) and proton MR spectroscopy in 21 healthy subjects to assess the potential relationship between GABA levels within mPFC and the amygdala-mPFC functional connectivity. Trait anxiety was assessed using the State-Trait Anxiety Inventory (STAI-Y2). Partial correlations were used to measure the relationships among the functional connectivity outcomes, mPFC GABA levels and STAI-Y2 scores. Age, educational level and amount of the gray and white matters within 1H-MRS volume of interest were included as nuisance variables. The rs-fMRI signals of the amygdala and the vmPFC were significantly anti-correlated. This negative functional coupling between the two regions was inversely correlated with the GABA+/tCr level within the mPFC and the STAI-Y2 scores. We suggest a close relationship between mPFC GABA levels and functional interactions within the amygdala-vmPFC circuit, providing new insights in the physiology of emotion.
Collapse
Affiliation(s)
- Stefano Delli Pizzi
- Department of Neuroscience, Imaging and Clinical Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
- Aging Research Centre, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Piero Chiacchiaretta
- Department of Neuroscience, Imaging and Clinical Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Dante Mantini
- Research Centre for Motor Control and Neuroplasticity, KU Leuven, Louvain, Belgium
- Department of Health Sciences and Technology, Neural Control of Movement Lab, ETH Zurich, Switzerland
- Department of Experimental Psychology, Oxford University, Oxford, UK
| | - Giovanna Bubbico
- Department of Neuroscience, Imaging and Clinical Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Antonio Ferretti
- Department of Neuroscience, Imaging and Clinical Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Richard A Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Center for Functional MRI, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Camillo Di Giulio
- Department of Neuroscience, Imaging and Clinical Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Marco Onofrj
- Department of Neuroscience, Imaging and Clinical Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
- Aging Research Centre, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy
| | - Laura Bonanni
- Department of Neuroscience, Imaging and Clinical Sciences, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy.
- Aging Research Centre, ''G. d'Annunzio'' University of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
34
|
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: 3.3] [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
|
35
|
Klenowski PM, Fogarty MJ, Shariff M, Belmer A, Bellingham MC, Bartlett SE. Increased Synaptic Excitation and Abnormal Dendritic Structure of Prefrontal Cortex Layer V Pyramidal Neurons following Prolonged Binge-Like Consumption of Ethanol. eNeuro 2016; 3:ENEURO.0248-16.2016. [PMID: 28032119 PMCID: PMC5179982 DOI: 10.1523/eneuro.0248-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/26/2016] [Accepted: 11/17/2016] [Indexed: 01/23/2023] Open
Abstract
Long-term alcohol use causes a multitude of neurochemical changes in cortical regions that facilitate the transition to dependence. Therefore, we used a model of long-term, binge-like ethanol consumption in rats to determine the effects on morphology and synaptic physiology of medial prefrontal cortex (mPFC) layer V pyramidal neurons. Following 10 weeks of ethanol consumption, we recorded synaptic currents from mPFC neurons and used neurobiotin filling to analyze their morphology. We then compared these data to measurements obtained from age-matched, water-drinking control rats. We found that long-term ethanol consumption caused a significant increase in total dendrite arbor length of mPFC layer V pyramidal neurons. Dendritic restructuring was primarily observed in basal dendrite arbors, with mPFC neurons from animals engaged in long-term ethanol drinking having significantly larger and more complex basal arbors compared with controls. These changes were accompanied by significantly increased total spine densities and spontaneous postsynaptic excitatory current frequency, suggesting that long-term binge-like ethanol consumption enhances basal excitatory synaptic transmission in mPFC layer V pyramidal neurons. Our results provide insights into the morphological and functional changes in mPFC layer V pyramidal neuronal physiology following prolonged exposure to ethanol and support changes in mPFC activity during the development of alcohol dependence.
Collapse
Affiliation(s)
- Paul M. Klenowski
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| | - Matthew J. Fogarty
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Queensland, Australia
| | - Masroor Shariff
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| | - Arnauld Belmer
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| | - Mark C. Bellingham
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Queensland, Australia
| | - Selena E. Bartlett
- Translational Research Institute and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4000, Queensland, Australia
| |
Collapse
|
36
|
Binge-like intake of HFD attenuates alcohol intake in rats. Physiol Behav 2016; 178:187-195. [PMID: 27765644 DOI: 10.1016/j.physbeh.2016.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/07/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022]
Abstract
Binge eating and binge alcohol intake are behavioral manifestations of pathological feeding and alcohol use disorder (AUD), respectively. Binge-feeding and AUD have high comorbidity with other psychiatric disorders such as depression, which could have important implications for the management of these conditions. Importantly, these behaviors share many common features suggesting a singular etiology. However, the nature by which binge-feeding affects the development or maintenance of AUD is unclear. The present study examined the impact of a binge-feeding from a nutritionally complete high-fat diet (HFD) on initiation and maintenance of alcohol intake, anxiolytic behavior and central genetic changes in brain regions that control alcohol-reinforced behaviors. To do this, male Long-Evans rats received chow (controls) or HFD every three days (HFD-3D) or every day (HFD-ED) for 5weeks. Rodent chow and water were available ad-libitum to all groups throughout the experiment. Following 5weeks of HFD cycling, 20.0% ethanol or 2.0% sucrose intake was evaluated. In addition, anxiety-like behavior was measured using a light-dark box apparatus. Both HFD-3D and -ED groups of rats consumed significantly large amount of food during 2h HFD access sessions and reduced their chow intake in the next 22h. Surprisingly, binge-fed rats displayed attenuated acquisition of alcohol intake whereas sucrose consumption was unaffected. Rats exposed to HFD spent more time in the light side compared to chow controls, indicating that binge-feeding induced anxiolytic effects. In addition, alterations in the brain neurotensin system were observed following HFD exposure. These data indicate that binge-feeding behavior induces behavioral and genetic changes that help explain how alcohol intake is influenced by co-morbid eating disorders.
Collapse
|
37
|
Geng KW, He T, Wang RR, Li CL, Luo WJ, Wu FF, Wang Y, Li Z, Lu YF, Guan SM, Chen J. Ethanol Increases Mechanical Pain Sensitivity in Rats via Activation of GABAA Receptors in Medial Prefrontal Cortex. Neurosci Bull 2016; 32:433-44. [PMID: 27628528 DOI: 10.1007/s12264-016-0063-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022] Open
Abstract
Ethanol is widely known for its ability to cause dramatic changes in emotion, social cognition, and behavior following systemic administration in humans. Human neuroimaging studies suggest that alcohol dependence and chronic pain may share common mechanisms through amygdala-medial prefrontal cortex (mPFC) interactions. However, whether acute administration of ethanol in the mPFC can modulate pain perception is unknown. Here we showed that bilateral microinjections of ethanol into the prelimbic and infralimbic areas of the mPFC lowered the bilateral mechanical pain threshold for 48 h without influencing thermal pain sensitivity in adult rats. However, bilateral microinjections of artificial cerebrospinal fluid into the mPFC or bilateral microinjections of ethanol into the dorsolateral PFC (also termed as motor cortex area 1 in Paxinos and Watson's atlas of The Rat Brain. Elsevier Academic Press, Amsterdam, 2005) failed to do so, suggesting regional selectivity of the effects of ethanol. Moreover, bilateral microinjections of ethanol did not change the expression of either pro-apoptotic (caspase-3 and Bax) or anti-apoptotic (Bcl-2) proteins, suggesting that the dose was safe and validating the method used in the current study. To determine whether γ-aminobutyric acid A (GABAA) receptors are involved in mediating the ethanol effects, muscimol, a selective GABAA receptor agonist, or bicuculline, a selective GABAA receptor antagonist, was administered alone or co-administered with ethanol through the same route into the bilateral mPFC. The results showed that muscimol mimicked the effects of ethanol while bicuculline completely reversed the effects of ethanol and muscimol. In conclusion, ethanol increases mechanical pain sensitivity through activation of GABAA receptors in the mPFC of rats.
Collapse
Affiliation(s)
- Kai-Wen Geng
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Ting He
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Rui-Rui Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Chun-Li Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Wen-Jun Luo
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Fang-Fang Wu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Yan Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Zhen Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yun-Fei Lu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Su-Min Guan
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Jun Chen
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China.
- Beijing Institute for Brain Disorders, Beijing, 100069, China.
| |
Collapse
|
38
|
Doherty JM, Schier CJ, Vena AA, Dilly GA, Gonzales RA. Medial Prefrontal Cortical Dopamine Responses During Operant Self-Administration of Sweetened Ethanol. Alcohol Clin Exp Res 2016; 40:1662-70. [PMID: 27435872 DOI: 10.1111/acer.13141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/31/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Medial prefrontal cortex (mPFC) dysfunction is present in heavy alcohol consumers. Dopamine signaling in mPFC is associated with executive functioning and affects drinking behavior; however, direct measurement of extracellular mPFC dopamine during appetitive and consummatory ethanol (EtOH) self-administration behavior has not been reported. METHODS We used in vivo microdialysis in freely behaving, adult, male, Long Evans rats to determine extracellular dopamine concentration in the mPFC during operant self-administration of an EtOH-plus-sucrose or sucrose solution. The model separated appetitive/seeking from consummatory phases of the operant session. Dopamine was also monitored in an untrained handling control group, and dialysate EtOH was measured in the EtOH-drinking group. RESULTS Home cage baseline dopamine was lower in rats that experienced a week of drinking sweetened EtOH compared with sucrose-drinking and handling controls. Transfer into the operant chamber and the initiation of consumption stimulated a relatively higher change in dopamine over baseline in the sweetened EtOH group compared with sucrose and handling controls. However, all groups show a dopamine response during transfer into the operant chamber, and the sucrose group had a relatively higher change in dopamine over baseline during initiation of consumption compared with handling controls. The time courses of dopamine and EtOH in the mPFC differ in the EtOH-consuming rats. CONCLUSIONS Differences in extracellular mPFC dopamine between EtOH drinkers compared with control groups suggest that mPFC dopamine is involved in the mechanism of operant self-administration of sweetened EtOH and sucrose. Furthermore, the increase in dopamine during consumption is consistent with a role of mPFC dopamine in reward prediction.
Collapse
Affiliation(s)
- James M Doherty
- Division of Pharmacology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Christina J Schier
- Division of Pharmacology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Ashley A Vena
- Division of Pharmacology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Geoffrey A Dilly
- Division of Pharmacology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Rueben A Gonzales
- Division of Pharmacology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
39
|
Gutman AL, Taha SA. Acute ethanol effects on neural encoding of reward size and delay in the nucleus accumbens. J Neurophysiol 2016; 116:1175-88. [PMID: 27169507 DOI: 10.1152/jn.00204.2014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/06/2016] [Indexed: 11/22/2022] Open
Abstract
Acute ethanol administration can cause impulsivity, resulting in increased preference for immediately available rewards over delayed but more valuable alternatives. The manner in which reward size and delay are represented in neural firing is not fully understood, and very little is known about ethanol effects on this encoding. To address this issue, we used in vivo electrophysiology to characterize neural firing in the core of the nucleus accumbens (NAcc) in rats responding for rewards that varied in size or delay after vehicle or ethanol administration. The NAcc is a central element in the circuit that governs decision-making and importantly, promotes choice of delayed rewards. We found that NAcc firing in response to reward-predictive cues encoded anticipated reward value after vehicle administration, but ethanol administration disrupted this encoding, resulting in a loss of discrimination between immediate and delayed rewards in cue-evoked neural responses. In addition, NAcc firing occurring at the time of the operant response (lever pressing) was inversely correlated with behavioral response latency, such that increased firing rates were associated with decreased latencies to lever press. Ethanol administration selectively attenuated this lever press-evoked firing when delayed but not immediate rewards were expected. These effects on neural firing were accompanied by increased behavioral latencies to respond for delayed rewards. Our results suggest that ethanol effects on NAcc cue- and lever press-evoked encoding may contribute to ethanol-induced impulsivity.
Collapse
Affiliation(s)
- Andrea L Gutman
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| | - Sharif A Taha
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| |
Collapse
|
40
|
den Hartog C, Zamudio-Bulcock P, Nimitvilai S, Gilstrap M, Eaton B, Fedarovich H, Motts A, Woodward JJ. Inactivation of the lateral orbitofrontal cortex increases drinking in ethanol-dependent but not non-dependent mice. Neuropharmacology 2016; 107:451-459. [PMID: 27016020 DOI: 10.1016/j.neuropharm.2016.03.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/05/2016] [Accepted: 03/21/2016] [Indexed: 12/01/2022]
Abstract
Long-term consumption of ethanol affects cortical areas that are important for learning and memory, cognition, and decision-making. Deficits in cortical function may contribute to alcohol-abuse disorders by impeding an individual's ability to control drinking. Previous studies from this laboratory show that acute ethanol reduces activity of lateral orbitofrontal cortex (LOFC) neurons while chronic exposure impairs LOFC-dependent reversal learning and induces changes in LOFC excitability. Despite these findings, the role of LOFC neurons in ethanol consumption is unknown. To address this issue, we examined ethanol drinking in adult C57Bl/6J mice that received an excitotoxic lesion or viral injection of the inhibitory DREADD (designer receptor exclusively activated by designer drug) into the LOFC. No differences in ethanol consumption were observed between sham and lesioned mice during access to increasing concentrations of ethanol (3-40%) every other day for 7 weeks. Adulterating the ethanol solution with saccharin (0.2%) or quinine (0.06 mM) enhanced or inhibited, respectively, consumption of the 40% ethanol solution similarly in both groups. Using a chronic intermittent ethanol (CIE) vapor exposure model that produces dependence, we found no difference in baseline drinking between sham and lesioned mice prior to vapor treatments. CIE enhanced drinking in both groups as compared to air-treated animals and CIE treated lesioned mice showed an additional increase in ethanol drinking as compared to CIE sham controls. This effect persisted during the first week when quinine was added to the ethanol solution but consumption decreased to control levels in CIE lesioned mice in the following 2 weeks. In viral injected mice, baseline drinking was not altered by expression of the inhibitory DREADD receptor and repeated cycles of CIE exposure enhanced drinking in DREADD and virus control groups. Consistent with the lesion study, treatment with clozapine-N-oxide (CNO) further enhanced consumption only in CIE exposed DREADD mice with no change in air-treated mice. These results suggest that the LOFC is not critical for the initiation and maintenance of ethanol drinking in non-dependent mice, but may regulate the escalated drinking observed during dependence.
Collapse
Affiliation(s)
- Carolina den Hartog
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Paula Zamudio-Bulcock
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sudarat Nimitvilai
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Meghin Gilstrap
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bethany Eaton
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Hleb Fedarovich
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Andrew Motts
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
41
|
Barker JM, Corbit LH, Robinson DL, Gremel CM, Gonzales RA, Chandler LJ. Corticostriatal circuitry and habitual ethanol seeking. Alcohol 2015; 49:817-24. [PMID: 26059221 PMCID: PMC4644517 DOI: 10.1016/j.alcohol.2015.03.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 03/22/2015] [Accepted: 03/26/2015] [Indexed: 01/01/2023]
Abstract
The development of alcohol-use disorders is thought to involve a transition from casual alcohol use to uncontrolled alcohol-seeking behavior. This review will highlight evidence suggesting that the shift toward inflexible alcohol seeking that occurs across the development of addiction consists, in part, of a progression from goal-directed to habitual behaviors. This shift in "response strategy" is thought to be largely regulated by corticostriatal network activity. Indeed, specific neuroanatomical substrates within the prefrontal cortex and the striatum have been identified as playing opposing roles in the expression of actions and habits. A majority of the research on the neurobiology of habitual behavior has focused on non-drug reward seeking. Here, we will highlight recent research identifying corticostriatal structures that regulate the expression of habitual alcohol seeking and a comparison will be made when possible to findings for non-drug rewards.
Collapse
Affiliation(s)
- Jacqueline M Barker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Laura H Corbit
- School of Psychology, University of Sydney, Sydney, Australia
| | - Donita L Robinson
- Bowles Center for Alcohol Studies, Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Christina M Gremel
- Department of Psychology, Neuroscience Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Rueben A Gonzales
- Department of Pharmacology, The University of Texas at Austin, Austin, TX, USA
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
42
|
Linsenbardt DN, Lapish CC. Neural Firing in the Prefrontal Cortex During Alcohol Intake in Alcohol-Preferring "P" Versus Wistar Rats. Alcohol Clin Exp Res 2015; 39:1642-53. [PMID: 26250465 DOI: 10.1111/acer.12804] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/05/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Neural activity within the prefrontal cortex (PFC) is altered by alcohol and alcohol-associated stimuli and is mediated by genetic susceptibility to alcoholism. However, very little is known about how genetic risk of excessive drinking might mediate neural firing in the PFC during alcohol consumption. METHODS To determine how genetic risk influences alcohol seeking, intake, and neural activity, a Pavlovian alcohol consumption task was used-the 2-Way Cued Access Protocol (2CAP). Alcohol-preferring "P" rats and relatives of their (heterogeneous) founding Wistar population were used for these studies. After acquisition of 2CAP, extinction of responding for alcohol was evaluated by substituting water for alcohol. Following these experiments, in vivo electrophysiological recordings were obtained during 2CAP from the PFC in a separate cohort of Wistar and P rats implanted with moveable tetrode microdrives. RESULTS P and Wistar rats increased daily alcohol seeking and intake with P rats consuming roughly twice as much alcohol as Wistar. Both rat populations decreased seeking behavior during extinction. However, P rats displayed persistent increases in seeking after controlling for intake versus Wistar. Higher firing rates (FRs) were observed in P rats prior to 2CAP and throughout alcohol and water consumption compared with Wistars that were matched for alcohol-drinking history. Differences in FR were driven, in part, by a larger percentage of neurons in P rats versus Wistars that increased FR compared with those that decreased, or did not change. CONCLUSIONS These data provide additional evidence of increased alcohol consumption and persistent alcohol seeking in P versus Wistar rats. Differences in PFC neural firing observed in P rats prior to drinking could be heritable and/or related to an enhanced response to alcohol-associated contextual cues. FR differences observed during alcohol drinking might be related to an augmented sensitivity of PFC neurons to orally consumed alcohol.
Collapse
Affiliation(s)
- David N Linsenbardt
- Addiction Neuroscience, Department of Psychology and Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, Indiana
| | - Christopher C Lapish
- Addiction Neuroscience, Department of Psychology and Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
43
|
Mishra D, Harrison NR, Gonzales CB, Schilström B, Konradsson-Geuken Å. Effects of age and acute ethanol on glutamatergic neurotransmission in the medial prefrontal cortex of freely moving rats using enzyme-based microelectrode amperometry. PLoS One 2015; 10:e0125567. [PMID: 25927237 PMCID: PMC4416039 DOI: 10.1371/journal.pone.0125567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/24/2015] [Indexed: 01/04/2023] Open
Abstract
Ethanol abuse during adolescence may significantly alter development of the prefrontal cortex which continues to undergo structural remodeling into adulthood. Glutamatergic neurotransmission plays an important role during these brain maturation processes and is modulated by ethanol. In this study, we investigated glutamate dynamics in the medial prefrontal cortex of freely moving rats, using enzyme-based microelectrode amperometry. We analyzed the effects of an intraperitoneal ethanol injection (1 g/kg) on cortical glutamate levels in adolescent and adult rats. Notably, basal glutamate levels decreased with age and these levels were found to be significantly different between postnatal day (PND) 28-38 vs PND 44-55 (p<0.05) and PND 28-38 vs adult animals (p<0.001). We also observed spontaneous glutamate release (transients) throughout the recordings. The frequency of transients (per hour) was significantly higher in adolescent rats (PND 28-38 and PND 44-55) compared to those of adults. In adolescent rats, post-ethanol injection, the frequency of glutamate transients decreased within the first hour (p<0.05), it recovered slowly and in the third hour there was a significant rebound increase of the frequency (p<0.05). Our data demonstrate age-dependent differences in extracellular glutamate levels in the medial prefrontal cortex and suggest that acute ethanol injections have both inhibitory and excitatory effects in adolescent rats. These effects of ethanol on the prefrontal cortex may disturb its maturation and possibly limiting individuals´ control over addictive behaviors.
Collapse
Affiliation(s)
- Devesh Mishra
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas R. Harrison
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Carolina B. Gonzales
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Schilström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience and Physiology, Section of Neuropsychiatry, Gothenburg University, Gothenburg, Sweden
| | - Åsa Konradsson-Geuken
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Navarro AI, Mandyam CD. Protracted abstinence from chronic ethanol exposure alters the structure of neurons and expression of oligodendrocytes and myelin in the medial prefrontal cortex. Neuroscience 2015; 293:35-44. [PMID: 25732140 DOI: 10.1016/j.neuroscience.2015.02.043] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/05/2015] [Accepted: 02/21/2015] [Indexed: 10/23/2022]
Abstract
In rodents, chronic intermittent ethanol vapor exposure (CIE) produces alcohol dependence, alters the structure and activity of pyramidal neurons and decreases the number of oligodendroglial progenitors in the medial prefrontal cortex (mPFC). In this study, adult Wistar rats were exposed to seven weeks of CIE and were withdrawn from CIE for 21 days (protracted abstinence; PA). Tissue enriched in the mPFC was processed for Western blot analysis and Golgi-Cox staining to investigate the long-lasting effects of CIE on the structure of mPFC neurons and the levels of myelin-associated proteins. PA increased dendritic arborization within apical dendrites of pyramidal neurons. These changes occurred concurrently with hypophosphorylation of the N-methyl-d-aspartate (NMDA) receptor 2B (NR2B) at Tyr-1472. PA increased myelin basic protein (MBP) levels which occurred concurrently with hypophosphorylation of the premyelinating oligodendrocyte bHLH transcription factor Olig2 in the mPFC. Given that PA is associated with increased sensitivity to stress and hypothalamic-pituitary-adrenal (HPA) axis dysregulation, and stress alters oligodendrocyte expression as a function of glucocorticoid receptor (GR) activation, the levels of total GR and phosphorylated GR were also evaluated. PA produced hypophosphorylation of the GR at Ser-232 without affecting expression of total protein. These findings demonstrate persistent and compensatory effects of ethanol in the mPFC long after cessation of CIE, including enhanced myelin production and impaired GR function. Collectively, these results suggest a novel relationship between oligodendrocytes and GR in the mPFC, in which stress may alter frontal cortex function in alcohol dependent subjects by promoting hypermyelination, thereby altering the cellular composition and white matter structure in the mPFC.
Collapse
Affiliation(s)
- A I Navarro
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - C D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
45
|
Korkotian E, Botalova A, Odegova T, Segal M. Chronic exposure to alcohol alters network activity and morphology of cultured hippocampal neurons. Neurotoxicology 2015; 47:62-71. [PMID: 25655208 DOI: 10.1016/j.neuro.2015.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/15/2015] [Accepted: 01/26/2015] [Indexed: 11/16/2022]
Abstract
The effects of chronic exposure to moderate concentrations of ethanol were studied in cultured hippocampal neurons. Network activity, assessed by imaging of [Ca(2+)]i variations, was markedly suppressed following 5 days of exposure to 0.25-1% ethanol. The reduced activity was sustained following extensive washout of ethanol, but the activity recovered by blockade of inhibition with bicuculline. This reduction of network activity was associated with a reduction in rates of mEPSCs, but not in a change in inhibitory synaptic activity. Chronic exposure to ethanol caused a significant reduction in the density of mature dendritic spines, without an effect on dendritic length or arborization. These results indicate that chronic exposure to ethanol causes a reduction in excitatory network drive in hippocampal neurons adding another dimension to the chronic effects of alcohol abuse.
Collapse
Affiliation(s)
- Eduard Korkotian
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel.
| | - Alena Botalova
- Neurobiological Research Center, Perm State Pharmaceutical Academy, Perm, Russia
| | - Tatiana Odegova
- Department of Microbiology, Perm State Pharmaceutical Academy, Perm, Russia
| | - Menahem Segal
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel
| |
Collapse
|
46
|
Geng X, Elmadhoun O, Peng C, Ji X, Hafeez A, Liu Z, Du H, Rafols JA, Ding Y. Ethanol and Normobaric Oxygen. Stroke 2015; 46:492-9. [PMID: 25563647 DOI: 10.1161/strokeaha.114.006994] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Xiaokun Geng
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Omar Elmadhoun
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Changya Peng
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Xunming Ji
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Adam Hafeez
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Zongjian Liu
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Huishan Du
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Jose A. Rafols
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| | - Yuchuan Ding
- From the China-America Institute of Neuroscience, Luhe Hospital (X.G., X.J., Z.L., H.D., Y.D.) and Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China; Departments of Neurological Surgery (X.G., O.E., C.P., A.H., Y.D.) and Anatomy and Cell Biology (J.A.R.), Wayne State University School of Medicine, Detroit, MI; and Beijing Institute for Brain Disorders, Beijing, China (X.J.)
| |
Collapse
|
47
|
Geng X, Sy CA, Kwiecien TD, Ji X, Peng C, Rastogi R, Cai L, Du H, Brogan D, Singh S, Rafols JA, Ding Y. Reduced cerebral monocarboxylate transporters and lactate levels by ethanol and normobaric oxygen therapy in severe transient and permanent ischemic stroke. Brain Res 2015; 1603:65-75. [PMID: 25641040 DOI: 10.1016/j.brainres.2015.01.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Neuroprotective benefits of ethanol (EtOH) and normobaric oxygenation (NBO) were previously demonstrated in transient and permanent ischemic stroke. Here we sought to identify whether the enhanced lactic acidosis and increased expression of monocarboxylate transporters (MCTs) observed after stroke might be attenuated by single and/or combined EtOH and NBO therapies. METHODS Sprague-Dawley rats (n=96) were subjected to right middle cerebral artery occlusion (MCAO) for 2 or 4h (transient ischemia), or 28 h (permanent ischemia) followed by 3, 24h, or no reperfusion. Rats received: (1) either an intraperitoneal injection of saline (sham treatment), one dose of EtOH (1.5 g/kg), two doses of EtOH (1.5 g/kg at 2h of MCAO, followed by 1.0 g/kg 2h after 1st dose), or (2) EtOH+95% NBO (at 2h of MCAO for 6h in permanent ischemia). Lactate levels were detected at 3 and 24h of reperfusion. Gene and protein expressions of MCT-1, -2, -4 were assessed by real-time PCR and western blotting. RESULTS A dose-dependent EtOH neuroprotection was found in transient ischemia. Following transient ischemia, a single dose of EtOH (in 2h-MCAO) or a double dose (in 4h-MCAO), significantly attenuated lactate levels, as well as the mRNAs and protein expressions of MCT-1, MCT-2, and MCT-4. However, while two doses of EtOH alone was ineffective in permanent stroke, the combined therapy (EtOH+95% NBO) resulted in a more significant attenuation in all the above levels and expressions. CONCLUSIONS Our study demonstrates that acute EtOH administration attenuated lactic acidosis in transient or permanent ischemic stroke. This EtOH-induced beneficial effect was potentiated by NBO therapy in permanent ischemia. Because both EtOH and NBO are readily available, inexpensive and easy to administer, their combination could be implemented in the clinics shortly after stroke.
Collapse
Affiliation(s)
- Xiaokun Geng
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Christopher A Sy
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Timothy D Kwiecien
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Xunming Ji
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Xuanwu Hospital, China-America Institute of Neuroscience, Luhe Hospital Capital Medical University, Beijing 100053, China.
| | - Changya Peng
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Radhika Rastogi
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Lipeng Cai
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Huishan Du
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| | - David Brogan
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Sunpreet Singh
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA
| | - Jose A Rafols
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuchuan Ding
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI 48201, USA.
| |
Collapse
|
48
|
Barker JM, Taylor JR. Habitual alcohol seeking: modeling the transition from casual drinking to addiction. Neurosci Biobehav Rev 2014; 47:281-94. [PMID: 25193245 PMCID: PMC4258136 DOI: 10.1016/j.neubiorev.2014.08.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/17/2014] [Accepted: 08/25/2014] [Indexed: 12/29/2022]
Abstract
The transition from goal-directed actions to habitual ethanol seeking models the development of addictive behavior that characterizes alcohol use disorders. The progression to habitual ethanol-seeking behavior occurs more rapidly than for natural rewards, suggesting that ethanol may act on habit circuit to drive the loss of behavioral flexibility. This review will highlight recent research that has focused on the formation and expression of habitual ethanol seeking, and the commonalities and distinctions between ethanol and natural reward-seeking habits, with the goal of highlighting important, understudied research areas that we believe will lead toward the development of novel treatment and prevention strategies for uncontrolled drinking.
Collapse
Affiliation(s)
- Jacqueline M Barker
- Department of Psychiatry, Yale University School of Medicine, Ribicoff Labs, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jane R Taylor
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
49
|
Pava MJ, Woodward JJ. Chronic ethanol alters network activity and endocannabinoid signaling in the prefrontal cortex. Front Integr Neurosci 2014; 8:58. [PMID: 25100953 PMCID: PMC4103173 DOI: 10.3389/fnint.2014.00058] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 06/27/2014] [Indexed: 11/22/2022] Open
Abstract
Chronic use of alcohol is associated with structural and functional alterations in brain areas that subserve cognitive processes. Of particular importance is the prefrontal cortex (PFC) that is involved in higher order behaviors such as decision making, risk assessment and judgment. Understanding the mechanisms that underlie alcohol's effects on PFC function is important for developing strategies to overcome the cognitive deficits that may predispose individuals to relapse. Our previous studies showed that acutely applied ethanol inhibits network activity in slices of prefrontal cortex and that exogenous and endogenous cannabinoids modulate up-state dynamics. In the present study, we examined the effects of repeated alcohol exposure on cannabinoid regulation of up-states in slice cultures of the prefrontal cortex. Compared to controls, up-state duration, but not amplitude was enhanced when measured 4 days after a 10 day ethanol exposure (44 mM ethanol; equivalent to 0.2% blood ethanol). Administration of the CB1 agonist WIN 55,212-2 enhanced the amplitude of up-states in control cultures but not in those treated previously with ethanol. This lack of effect occurred in the absence of any noticeable change in CB1 receptor protein expression. Chronic ethanol treatment and withdrawal also blunted WIN's inhibition of electrically evoked GABA IPSCs in layer II/III pyramidal neurons but not those in layer V/VI. WIN inhibited the amplitude of spontaneous GABA IPSCs in both layers and the magnitude of this effect was not altered by ethanol treatment. However, in layer V/VI neurons, WIN's effect on sIPSC frequency was greater in ethanol treated cultures. WIN also inhibited electrically evoked NMDA EPSCs in both layer II/III and V/VI neurons but this action was unaffected by ethanol treatment and withdrawal. Overall, these results suggest that ethanol's down-regulation of cannabinoid signaling results in altered network activity in the prefrontal cortex.
Collapse
Affiliation(s)
- Matthew J Pava
- Department of Neuroscience, Medical University of South Carolina Charleston, SC, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina Charleston, SC, USA
| |
Collapse
|
50
|
Abstract
Escalation of voluntary alcohol consumption is a hallmark of alcoholism, but its neural substrates remain unknown. In rats, escalation occurs following prolonged exposure to cycles of alcohol intoxication, and is associated with persistent, wide-ranging changes in gene expression within the medial prefrontal cortex (mPFC). Here, we examined whether induction of microRNA (miR) 206 in mPFC contributes to escalated alcohol consumption. Following up on a microarray screen, quantitative real-time reverse transcription PCR (qPCR) confirmed that a history of dependence results in persistent (>3weeks) up-regulation of miR-206 expression in the mPFC, but not in the ventral tegmental area, amygdala, or nucleus accumbens. Viral-mediated overexpression of miR-206 in the mPFC of nondependent rats reproduced the escalation of alcohol self-administration seen following a history of dependence and significantly inhibited BDNF expression. Bioinformatic analysis identified three conserved target sites for miR-206 in the 3'-UTR of the rat BDNF transcript. Accordingly, BDNF was downregulated in post-dependent rats on microarray analysis, and this was confirmed by qPCR. In vitro, BDNF expression was repressed by miR-206 but not miR-9 in a 3'-UTR reporter assay, confirming BDNF as a functional target of miR-206. Mutation analysis showed that repression was dependent on the presence of all three miR-206 target sites in the BDNF 3'-UTR. Inhibition of miR-206 expression in differentiated rat cortical primary neurons significantly increased secreted levels of BDNF. In conclusion, recruitment of miR-206 in the mPFC contributes to escalated alcohol consumption following a history of dependence, with BDNF as a possible mediator of its action.
Collapse
|