1
|
Mikhalitskaya EV, Vyalova NM, Bokhan NA, Ivanova SA. Alcohol-Induced Activation of Chemokine System and Neuroinflammation Development. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1889-1903. [PMID: 39647818 DOI: 10.1134/s0006297924110038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 12/10/2024]
Abstract
Chemokines are immunoregulatory proteins with pleiotropic functions involved in neuromodulation, neurogenesis, and neurotransmission. The way chemokines affect the CNS plays an important role in modulating various conditions that could have negative impact on CNS functions, including development of alcohol use disorders. In this review, we analyzed the literature data available on the problem of chemokine participation in pathogenesis, clinical presentation, and remission of alcohol use disorders both in animal models and in the study of patients with alcoholism. The presented information confirms the hypothesis that the alcohol-induced chemokine production could modulate chronic neuroinflammation. Thus, the data summarized and shown in this review are focused on the relevant direction of research in the field of psychiatry, which is in demand by both scientists and clinical specialists.
Collapse
Affiliation(s)
- Ekaterina V Mikhalitskaya
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia.
| | - Natalya M Vyalova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| |
Collapse
|
2
|
Cruz B, Borgonetti V, Bajo M, Roberto M. Sex-dependent factors of alcohol and neuroimmune mechanisms. Neurobiol Stress 2023; 26:100562. [PMID: 37601537 PMCID: PMC10432974 DOI: 10.1016/j.ynstr.2023.100562] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Excessive alcohol use disrupts neuroimmune signaling across various cell types, including neurons, microglia, and astrocytes. The present review focuses on recent, albeit limited, evidence of sex differences in biological factors that mediate neuroimmune responses to alcohol and underlying neuroimmune systems that may influence alcohol drinking behaviors. Females are more vulnerable than males to the neurotoxic and negative consequences of chronic alcohol drinking, reflected by elevations of pro-inflammatory cytokines and inflammatory mediators. Differences in cytokine, microglial, astrocytic, genomic, and transcriptomic evidence suggest females are more reactive than males to neuroinflammatory changes after chronic alcohol exposure. The growing body of evidence supports that innate immune factors modulate synaptic transmission, providing a mechanistic framework to examine sex differences across neurocircuitry. Targeting neuroimmune signaling may be a viable strategy for treating AUD, but more research is needed to understand sex-specific differences in alcohol drinking and neuroimmune mechanisms.
Collapse
Affiliation(s)
- Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Vittoria Borgonetti
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| |
Collapse
|
3
|
Li S, Zheng Y, Xiao L, Lan S, Xiang J, Liao L, Lin Y, Ye Y. Aldehyde dehydrogenase 2-associated changes in pharmacokinetics, locomotor function and peripheral glutamic acid and gamma-aminobutyric acid levels during acute alcohol intoxication in male mice. Behav Pharmacol 2022; 33:551-558. [PMID: 36256704 DOI: 10.1097/fbp.0000000000000702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The insufficiency of human aldehyde dehydrogenase 2 (ALDH2) has been consistently associated with high blood acetaldehyde levels and impaired locomotor function during acute alcohol intoxication. The ALDH2-associated change in peripheral glutamic acid (Glu) and gamma-aminobutyric acid (GABA) levels and its correlation with pharmacokinetics and psychomotor function remain unclear. In this study, ALDH2*2 mice were used to build an acute alcohol intoxication model after intraperitoneal administration. The blood ethanol and acetaldehyde concentrations were analyzed to generate concentration-time curves at two doses of alcohol (2.0 and 4.0 g/kg). The dose of 4.0 g/kg was selected in accordance with the preliminary behavioral evaluation result to perform the following behavioral tests (e.g. the rotarod test, the open field test, and the Y-maze test), so as to assess locomotor activity, anxiety and cognitive ability. Plasma Glu and GABA levels were determined through enzyme-linked immunosorbent assays. The results suggested that the ALDH2*2 mice had highly accumulated acetaldehyde levels, impaired locomotor activity and anxiety-like emotion but unimpaired cognitive function, compared to the wild type (WT) mice. The plasma Glu level and the ratio of Glu/GABA in the alcohol-treated WT and ALDH2*2 groups decreased from 2 to 5 h after intraperitoneal administration, whereas the GABA level did not change significantly. The blood alcohol concentration in the WT and ALDH2*2 mice was positively correlated with plasma Glu level, whereas the blood acetaldehyde level was found as the opposite. We speculate that the decline degree of Glu/GABA ratio could be associated with psychomotor retardation and needs to be further investigated.
Collapse
Affiliation(s)
- Songfan Li
- Shanghai Key Laboratory of Forensic Medicine, Shanghai
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yuzi Zheng
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Li Xiao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Shengnan Lan
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jin Xiang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yao Lin
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Ye
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Bloch S, Holleran KM, Kash TL, Vazey EM, Rinker JA, Lebonville CL, O'Hara K, Lopez MF, Jones SR, Grant KA, Becker HC, Mulholland PJ. Assessing negative affect in mice during abstinence from alcohol drinking: Limitations and future challenges. Alcohol 2022; 100:41-56. [PMID: 35181404 PMCID: PMC8983487 DOI: 10.1016/j.alcohol.2022.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/09/2023]
Abstract
Alcohol use disorder (AUD) is frequently comorbid with mood disorders, and these co-occurring neuropsychiatric disorders contribute to the development and maintenance of alcohol dependence and relapse. In preclinical models, mice chronically exposed to alcohol display anxiety-like and depressive-like behaviors during acute withdrawal and protracted abstinence. However, in total, results from studies using voluntary alcohol-drinking paradigms show variable behavioral outcomes in assays measuring negative affective behaviors. Thus, the main objective of this review is to summarize the literature on the variability of negative affective behaviors in mice after chronic alcohol exposure. We compare the behavioral phenotypes that emerge during abstinence across different exposure models, including models of alcohol and stress interactions. The complicated outcomes from these studies highlight the difficulties of assessing negative affective behaviors in mouse models designed for the study of AUD. We discuss new behavioral assays, comprehensive platforms, and unbiased machine-learning algorithms as promising approaches to better understand the interaction between alcohol and negative affect in mice. New data-driven approaches in the understanding of mouse behavior hold promise for improving the identification of mechanisms, cell subtypes, and neurocircuits that mediate negative affect. In turn, improving our understanding of the neurobehavioral basis of alcohol-associated negative affect will provide a platform to test hypotheses in mouse models that aim to improve the development of more effective strategies for treating individuals with AUD and co-occurring mood disorders.
Collapse
Affiliation(s)
- Solal Bloch
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Jennifer A Rinker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Christina L Lebonville
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Krysten O'Hara
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Marcelo F Lopez
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States
| | - Howard C Becker
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States.
| |
Collapse
|
5
|
Lee KM, Talikoti A, Shelton K, Grotewiel M. Tyramine synthesis, vesicular packaging, and the SNARE complex function coordinately in astrocytes to regulate Drosophila alcohol sedation. Addict Biol 2021; 26:e13019. [PMID: 33538092 PMCID: PMC8225576 DOI: 10.1111/adb.13019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/08/2021] [Accepted: 01/19/2021] [Indexed: 12/21/2022]
Abstract
Identifying mechanisms underlying alcohol-related behaviors could provide important insights regarding the etiology of alcohol use disorder. To date, most genetic studies on alcohol-related behavior in model organisms have focused on neurons, leaving the causal roles of glial mechanisms less comprehensively investigated. Here, we report our studies on the role of Tyrosine decarboxylase 2 (Tdc2), which converts tyrosine to the catecholamine tyramine, in glial cells in Drosophila alcohol sedation. Using genetic approaches that drove transgene expression constitutively in all glia, constitutively in astrocytes and conditionally in glia during adulthood, we found that knockdown and overexpression of Tdc2, respectively, increased and decreased the sensitivity to alcohol sedation in flies. Manipulation of the genes tyramine β-hydroxylase and tyrosine hydroxylase, which respectively synthesize octopamine and dopamine from tyramine and tyrosine, had no discernable effect on alcohol sedation, suggesting that Tdc2 affects alcohol sedation by regulating tyramine production. We also found that knockdown of the vesicular monoamine transporter (VMAT) and disruption of the SNARE complex in all glia or selectively in astrocytes increased sensitivity to alcohol sedation and that both VMAT and the SNARE complex functioned downstream of Tdc2. Our studies support a model in which the synthesis of tyramine and vesicle-mediated release of tyramine from adult astrocytes regulates alcohol sedation in Drosophila. Considering that tyramine is functionally orthologous to norepinephrine in mammals, our results raise the possibility that gliotransmitter synthesis release could be a conserved mechanism influencing behavioral responses to alcohol as well as alcohol use disorder.
Collapse
Affiliation(s)
- Kristen M. Lee
- Neuroscience Graduate Program, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ananya Talikoti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Keith Shelton
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Mike Grotewiel
- Neuroscience Graduate Program, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
- Virginia Commonwealth University Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
6
|
A novel mouse model for vulnerability to alcohol dependence induced by early-life adversity. Neurobiol Stress 2020; 13:100269. [PMID: 33344722 PMCID: PMC7739069 DOI: 10.1016/j.ynstr.2020.100269] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/16/2020] [Accepted: 11/14/2020] [Indexed: 12/13/2022] Open
Abstract
Childhood adversity increases vulnerability to alcohol use disorders and preclinical models are needed to investigate the underlying neurobiological mechanisms. The present study modeled early-life adversity by rearing male and female C57BL/6J mouse pups in a limited bedding and nesting (LBN) environment, which induces erratic maternal care. As adults, mice were given limited access to two-bottle choice (2BC) alcohol drinking, combined or not with chronic intermittent ethanol (CIE) vapor inhalation to induce alcohol dependence. We tested the hypothesis that LBN rearing might exacerbate or facilitate the emergence of the motivational and affective effects of CIE. Consistent with our hypothesis, although LBN-reared males consumed the same baseline levels of alcohol as controls, they escalated their ethanol intake at an earlier stage of CIE exposure, i.e., after 4 rounds vs. 5 rounds for controls. In contrast, females were insensitive to both LBN rearing and CIE exposure. Males were further subjected to a behavioral test battery. Withdrawal from CIE-2BC increased digging activity and lowered mechanical nociceptive thresholds regardless of early-life conditions. On the other hand, LBN-reared CIE-2BC males showed reduced open arm exploration in the elevated plus maze and increased immobility in the tail suspension test compared to alcohol-naïve counterparts, while no group differences were detected among control-reared males. Finally, LBN rearing and alcohol exposure did not affect grooming in response to a sucrose spray (splash test), novel object recognition, or corticosterone levels. In summary, the LBN experience accelerates the transition from moderate to excessive alcohol drinking and produces additional indices of affective dysfunction during alcohol withdrawal in C57BL/6J male mice. Early-life adversity was generated by rearing C57BL/6J mouse pups in a limited bedding and nesting (LBN) environment. Alcohol dependence was induced in adulthood via chronic intermittent ethanol (CIE) inhalation. The LBN experience accelerated alcohol intake escalation in males. LBN exacerbated affective disturbances upon CIE withdrawal in males. Alcohol intake in females was insensitive to both LBN and CIE.
Collapse
|
7
|
Aurelian L, Balan I. GABA AR α2-activated neuroimmune signal controls binge drinking and impulsivity through regulation of the CCL2/CX3CL1 balance. Psychopharmacology (Berl) 2019; 236:3023-3043. [PMID: 31030249 DOI: 10.1007/s00213-019-05220-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE Toll-like receptors (TLRs) are a family of innate immune system receptors that respond to pathogen-derived and tissue damage-related ligands and are increasingly recognized for their impact on homeostasis and its dysregulation in the nervous system. TLR signaling participates in brain injury and addiction, but its role in the alcohol-seeking behavior, which initiates alcohol drinking, is still poorly understood. In this review, we discuss our findings designed to elucidate the potential contribution of the activated TLR4 signal located in neurons, on impulsivity and the predisposition to initiate alcohol drinking (binge drinking). RESULTS Our findings indicate that the TLR4 signal is innately activated in neurons from alcohol-preferring subjects, identifying a genetic contribution to the regulation of impulsivity and the alcohol-seeking propensity. Signal activation is through the non-canonical, previously unknown, binding of TLR4 to the α2 subunit of the γ-aminobutyric 2 acid A receptor (GABAAR α2). Activation is sustained by the stress hormone corticotrophin-releasing factor (CRF) and additional still poorly recognized ligand/scaffold proteins. Focus is on the effect of TLR4 signal activation on the balance between pro- and anti-inflammatory chemokines [chemokine (C-C motif) ligand 2 (CCL2)/chemokine (C-X3-C motif) ligand 1 (CX3CL1)] and its effect on binge drinking. CONCLUSION The results are discussed within the context of current findings on the distinct activation and functions of TLR signals located in neurons, as opposed to immune cells. They indicate that the balance between pro- and anti-inflammatory TLR4 signaling plays a major role in binge drinking. These findings have major impact on future basic and translational research, including the development of potential therapeutic and preventative strategies.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Stanford University School of Medicine OFDD, Stanford, CA, 94305, USA.
| | - Irina Balan
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
8
|
Erickson EK, Blednov YA, Harris RA, Mayfield RD. Glial gene networks associated with alcohol dependence. Sci Rep 2019; 9:10949. [PMID: 31358844 PMCID: PMC6662804 DOI: 10.1038/s41598-019-47454-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic alcohol abuse alters the molecular structure and function of brain cells. Recent work suggests adaptations made by glial cells, such as astrocytes and microglia, regulate physiological and behavioral changes associated with addiction. Defining how alcohol dependence alters the transcriptome of different cell types is critical for developing the mechanistic hypotheses necessary for a nuanced understanding of cellular signaling in the alcohol-dependent brain. We performed RNA-sequencing on total homogenate and glial cell populations isolated from mouse prefrontal cortex (PFC) following chronic intermittent ethanol vapor exposure (CIE). Compared with total homogenate, we observed unique and robust gene expression changes in astrocytes and microglia in response to CIE. Gene co-expression network analysis revealed biological pathways and hub genes associated with CIE in astrocytes and microglia that may regulate alcohol-dependent phenotypes. Astrocyte identity and synaptic calcium signaling genes were enriched in alcohol-associated astrocyte networks, while TGF-β signaling and inflammatory response genes were disrupted by CIE treatment in microglia gene networks. Genes related to innate immune signaling, specifically interferon pathways, were consistently up-regulated across CIE-exposed astrocytes, microglia, and total homogenate PFC tissue. This study illuminates the cell-specific effects of chronic alcohol exposure and provides novel molecular targets for studying alcohol dependence.
Collapse
Affiliation(s)
- Emma K Erickson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA.
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| |
Collapse
|
9
|
Bai D, Jin G, Zhang D, Zhao L, Wang M, Zhu Q, Zhu L, Sun Y, Liu X, Chen X, Zhang L, Li W, Cui Y. Natural silibinin modulates amyloid precursor protein processing and amyloid-β protein clearance in APP/PS1 mice. J Physiol Sci 2019; 69:643-652. [PMID: 31087219 PMCID: PMC10717595 DOI: 10.1007/s12576-019-00682-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 04/29/2019] [Indexed: 11/28/2022]
Abstract
Silibinin has been shown to attenuate cognitive dysfunction and inhibit amyloid-beta (Aβ) aggregation in Alzheimer's disease (AD) models. However, the underlying mechanism by which silibinin improves cognition remains poorly understood. In this study, we investigated the effect of silibinin on β-secretase levels, Aβ enzymatic degradation, and oxidative stress in the brains of APP/PS1 mice with cognitive impairments. Oral administration of silibinin for 2 months significantly attenuated the cognitive deficits of APP/PS1 mice in the Y-maze test, novel object recognition test, and Morris water maze test. Biochemical analyses revealed that silibinin decreased Aβ deposition and the levels of soluble Aβ1-40/1-42 in the hippocampus by downregulating APP and BACE1 and upregulating NEP in APP/PS1 mice. In addition, silibinin decreased the MDA content and increased the activities of the antioxidant enzymes CAT, SOD, and NO. Based on our findings, silibinin is a potentially promising agent for preventing AD-associated Aβ pathology.
Collapse
Affiliation(s)
- Dafeng Bai
- Department of Pharmacology, The Eleventh People's Hospital of Shenyang, 103 Hai Tang Street, Sujiatun District, Shenyang, Liaoning, 110016, People's Republic of China
| | - Ge Jin
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China.
| | - Dajun Zhang
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Lini Zhao
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Mingyue Wang
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Qiwen Zhu
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Lin Zhu
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Yan Sun
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Xuan Liu
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Xueying Chen
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Liqian Zhang
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Wenbo Li
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| | - Yan Cui
- Department of Pharmacology, Key Laboratory of Behavioral and Cognitive Neuroscience of Liaoning Province, Shenyang Medical Colleges, 146 Huanghe North Street, Yuhong District, Shenyang, Liaoning, 110034, People's Republic of China
| |
Collapse
|
10
|
Erickson EK, Grantham EK, Warden AS, Harris RA. Neuroimmune signaling in alcohol use disorder. Pharmacol Biochem Behav 2018; 177:34-60. [PMID: 30590091 DOI: 10.1016/j.pbb.2018.12.007] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/25/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder (AUD) is a widespread disease with limited treatment options. Targeting the neuroimmune system is a new avenue for developing or repurposing effective pharmacotherapies. Alcohol modulates innate immune signaling in different cell types in the brain by altering gene expression and the molecular pathways that regulate neuroinflammation. Chronic alcohol abuse may cause an imbalance in neuroimmune function, resulting in prolonged perturbations in brain function. Likewise, manipulating the neuroimmune system may change alcohol-related behaviors. Psychiatric disorders that are comorbid with AUD, such as post-traumatic stress disorder, major depressive disorder, and other substance use disorders, may also have underlying neuroimmune mechanisms; current evidence suggests that convergent immune pathways may be involved in AUD and in these comorbid disorders. In this review, we provide an overview of major neuroimmune cell-types and pathways involved in mediating alcohol behaviors, discuss potential mechanisms of alcohol-induced neuroimmune activation, and present recent clinical evidence for candidate immune-related drugs to treat AUD.
Collapse
Affiliation(s)
- Emma K Erickson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA.
| | - Emily K Grantham
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| |
Collapse
|
11
|
Sidhu H, Kreifeldt M, Contet C. Affective Disturbances During Withdrawal from Chronic Intermittent Ethanol Inhalation in C57BL/6J and DBA/2J Male Mice. Alcohol Clin Exp Res 2018; 42:1281-1290. [PMID: 29687895 PMCID: PMC6028290 DOI: 10.1111/acer.13760] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/16/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND Alcohol use disorders are characterized by a complex behavioral symptomatology, which includes the loss of control over alcohol consumption and the emergence of a negative affective state when alcohol is not consumed. Some of these symptoms can be recapitulated in rodent models, for instance following chronic intermittent ethanol (EtOH; CIE) vapor inhalation. However, the detection of negative affect in mice withdrawn from CIE has proven challenging and variable between strains. This study aimed to detect reliable indices of negative emotionality in CIE-exposed C57BL/6J (C57) and DBA/2J (DBA) mice. Males were used because they are known to escalate their voluntary EtOH consumption upon CIE exposure, which is hypothesized to be driven by negative reinforcement (relief from negative affect). METHODS Adult male mice were exposed to 4 to 6 weeks of CIE and were evaluated 3 to 10 days into withdrawal in the social approach, novelty-suppressed feeding, digging, marble burying, and bottle brush tests. RESULTS Withdrawal from CIE decreased sociability in DBA mice but not in C57 mice. Conversely, hyponeophagia was exacerbated by CIE in C57 mice but not in DBA mice. Withdrawal from CIE robustly increased digging activity in both strains, even in the absence of marbles. Aggressive responses to bottle brush attacks were elevated in both C57 and DBA mice following CIE exposure, but CIE had an opposite effect on defensive responses in the 2 strains (increase in C57 vs. decrease in DBA). CONCLUSIONS Our results indicate that withdrawal from CIE elicits negative emotionality in both C57 and DBA mice, but different tests need to be used to measure the anxiogenic-like effects of withdrawal in each strain. Increased digging activity and irritability-like behavior represent novel indices of affective dysfunction associated with withdrawal from CIE in both mouse strains. Our findings enrich the characterization of the affective symptomatology of protracted withdrawal from CIE in mice.
Collapse
Affiliation(s)
- Harpreet Sidhu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Max Kreifeldt
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Candice Contet
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
12
|
Bray JG, Reyes KC, Roberts AJ, Gruol DL. Altered hippocampal synaptic function in transgenic mice with increased astrocyte expression of CCL2 after withdrawal from chronic alcohol. Neuropharmacology 2018; 135:113-125. [PMID: 29499275 DOI: 10.1016/j.neuropharm.2018.02.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 02/22/2018] [Accepted: 02/26/2018] [Indexed: 11/29/2022]
Abstract
CNS actions of the chemokine CCL2 are thought to play a role in a variety of conditions that can have detrimental consequences to CNS function, including alcohol use disorders. We used transgenic mice that express elevated levels of CCL2 in the CNS (CCL2-tg) and their non-transgenic (non-tg) littermate control mice to investigate long-term consequences of CCL2/alcohol/withdrawal interactions on hippocampal synaptic function, including excitatory synaptic transmission, somatic excitability, and synaptic plasticity. Two alcohol exposure paradigms were tested, a two-bottle choice alcohol (ethanol) drinking protocol (2BC drinking) and a chronic intermittent alcohol (ethanol) (CIE/2BC) protocol. Electrophysiological measurements of hippocampal function were made ex vivo, starting ∼0.6 months after termination of alcohol exposure. Both alcohol exposure/withdrawal paradigms resulted in CCL2-dependent interactions that altered the effects of alcohol on synaptic function. The synaptic alterations differed for the two alcohol exposure paradigms. The 2BC drinking/withdrawal treatment had no apparent long-term consequences on synaptic responses and long-term potentiation (LTP) in hippocampal slices from non-tg mice, whereas synaptic transmission was reduced but LTP was enhanced in hippocampal slices from CCL2-tg mice. In contrast, the CIE/2BC/withdrawal treatment enhanced synaptic transmission but reduced LTP in the non-tg hippocampus, whereas there were no apparent long-term consequences to synaptic transmission and LTP in hippocampus from CCL2-tg mice, although somatic excitability was enhanced. These results support the idea that alcohol-induced CCL2 production can modulate the effects of alcohol exposure/withdrawal on synaptic function and indicate that CCL2/alcohol interactions can vary depending on the alcohol exposure/withdrawal protocol used.
Collapse
Affiliation(s)
- Jennifer G Bray
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kenneth C Reyes
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Amanda J Roberts
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Donna L Gruol
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Huitron-Resendiz S, Nadav T, Krause S, Cates-Gatto C, Polis I, Roberts AJ. Effects of Withdrawal from Chronic Intermittent Ethanol Exposure on Sleep Characteristics of Female and Male Mice. Alcohol Clin Exp Res 2018; 42:540-550. [PMID: 29265376 DOI: 10.1111/acer.13584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/11/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Sleep disruptions are an important consequence of alcohol use disorders. There is a dearth of preclinical studies examining sex differences in sleep patterns associated with ethanol (EtOH) dependence despite documented sex differences in alcohol-related behaviors and withdrawal symptoms. The purpose of this study was to investigate the effects of chronic intermittent EtOH on sleep characteristics in female and male mice. METHODS Female and male C57BL6/J mice had access to EtOH/water 2-bottle choice (2BC) 2 h/d for 3 weeks followed by exposure to EtOH vapor (vapor-2BC) or air for 5 cycles of 4 days. An additional group never experienced EtOH (naïve). Mice were implanted with electroencephalographic (EEG) electrodes, and vigilance states were recorded across 24 hours on the fourth day of withdrawal. The amounts of wakefulness, slow-wave sleep (SWS), and rapid eye movement sleep were calculated, and spectral analysis was performed by fast Fourier transformation. RESULTS Overall, vapor-2BC mice showed a decrease in the amount of SWS 4 days into withdrawal as well as a decrease in the power density of slow waves, indicating disruptions in both the amount and quality of sleep in EtOH-dependent mice. This was associated with a decrease in duration and an increase in number of SWS episodes in males and an increase in latency to sleep in females. CONCLUSIONS Our results revealed overall deficits in sleep regulation in EtOH-dependent mice of both sexes. Female mice appeared to be more affected with regard to the triggering of sleep, while male mice appeared more sensitive to disruptions in the maintenance of sleep.
Collapse
Affiliation(s)
| | - Tali Nadav
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Stephanie Krause
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Chelsea Cates-Gatto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Ilham Polis
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Amanda J Roberts
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| |
Collapse
|