1
|
Randall CA, Sun D, Randall PA. A novel alcohol+nicotine co-use self-administration procedure reveals sex differences and differential alteration of mesocorticolimbic TLR- and cholinergic-related neuroimmune gene expression in rats. Alcohol 2024; 121:115-131. [PMID: 39197504 DOI: 10.1016/j.alcohol.2024.08.003] [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: 05/03/2024] [Revised: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Although alcohol and nicotine are two of the most commonly co-used drugs with upwards of 90% of adults with an alcohol use disorder (AUD) in the US also smoking, we don't tend to study alcohol and nicotine use this way. The current studies sought to develop and assess a novel alcohol + nicotine co-access self-administration (SA) model in adult male and female Long-Evans rats. Further, both drugs are implicated in neuroimmune function, albeit in largely opposing ways. Chronic alcohol use increases neuroinflammation via toll-like receptors (TLRs) which in turn increases alcohol intake. By contrast, nicotine produces anti-inflammatory effects, in part, through the monomeric alpha7 receptor (ChRNa7). Following long-term co-access (6 months), rats reliably administered both drugs during daily sessions, however males generally responded for more alcohol and females for nicotine. This was reflected in plasma analysis with translationally relevant intake levels of both alcohol and nicotine, making it invaluable in studying the effects of co-use on behavior and CNS function. Moreover, male rats show sensitivity to alterations in alcohol concentration whereas females show sensitivity to alterations in nicotine concentration. Rats trained on this procedure also developed an anxiogenic phenotype. Finally, we assessed alterations in neuroimmune-related gene expression in the medial prefrontal cortex - prelimbic, (mPFC-PL), nucleus accumbens core (AcbC), and ventral tegmental area (VTA). In the AcbC, where α7 expression was increased and β2 was decreased, markers of pro-inflammatory activity were decreased, despite increases in TLR gene expression suggesting that co-use with nicotine modulates inflammatory state downstream from the receptor level. By contrast, in mPFC-PL where α7 was not increased, both TLRs and downstream proinflammatory markers were increased. Taken together, these findings support that there are brain regional and sex differences with co-use of alcohol + nicotine SA and suggest that targeting nicotinic α7 may represent a novel strategy for treating alcohol + nicotine co-dependence.
Collapse
Affiliation(s)
- Christie A Randall
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA
| | - Dongxiao Sun
- Department of Pharmacology, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA
| | - Patrick A Randall
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA; Department of Pharmacology, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA.
| |
Collapse
|
2
|
Crews FT, Coleman LG, Macht VA, Vetreno RP. Alcohol, HMGB1, and Innate Immune Signaling in the Brain. Alcohol Res 2024; 44:04. [PMID: 39135668 PMCID: PMC11318841 DOI: 10.35946/arcr.v44.1.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Binge drinking (i.e., consuming enough alcohol to achieve a blood ethanol concentration of 80 mg/dL, approximately 4-5 drinks within 2 hours), particularly in early adolescence, can promote progressive increases in alcohol drinking and alcohol-related problems that develop into compulsive use in the chronic relapsing disease, alcohol use disorder (AUD). Over the past decade, neuroimmune signaling has been discovered to contribute to alcohol-induced changes in drinking, mood, and neurodegeneration. This review presents a mechanistic hypothesis supporting high mobility group box protein 1 (HMGB1) and Toll-like receptor (TLR) signaling as key elements of alcohol-induced neuroimmune signaling across glia and neurons, which shifts gene transcription and synapses, altering neuronal networks that contribute to the development of AUD. This hypothesis may help guide further research on prevention and treatment. SEARCH METHODS The authors used the search terms "HMGB1 protein," "alcohol," and "brain" across PubMed, Scopus, and Embase to find articles published between 1991 and 2023. SEARCH RESULTS The database search found 54 references in PubMed, 47 in Scopus, and 105 in Embase. A total of about 100 articles were included. DISCUSSION AND CONCLUSIONS In the brain, immune signaling molecules play a role in normal development that differs from their functions in inflammation and the immune response, although cellular receptors and signaling are shared. In adults, pro-inflammatory signals have emerged as contributing to brain adaptation in stress, depression, AUD, and neurodegenerative diseases. HMGB1, a cytokine-like signaling protein released from activated cells, including neurons, is hypothesized to activate pro-inflammatory signals through TLRs that contribute to adaptations to binge and chronic heavy drinking. HMGB1 alone and in heteromers with other molecules activates TLRs and other immune receptors that spread signaling across neurons and glia. Both blood and brain levels of HMGB1 increase with ethanol exposure. In rats, an adolescent intermittent ethanol (AIE) binge drinking model persistently increases brain HMGB1 and its receptors; alters microglia, forebrain cholinergic neurons, and neuronal networks; and increases alcohol drinking and anxiety while disrupting cognition. Studies of human postmortem AUD brain have found elevated levels of HMGB1 and TLRs. These signals reduce cholinergic neurons, whereas microglia, the brain's immune cells, are activated by binge drinking. Microglia regulate synapses through complement proteins that can change networks affected by AIE that increase drinking, contributing to risks for AUD. Anti-inflammatory drugs, exercise, cholinesterase inhibitors, and histone deacetylase epigenetic inhibitors prevent and reverse the AIE-induced pathology. Further, HMGB1 antagonists and other anti-inflammatory treatments may provide new therapies for alcohol misuse and AUD. Collectively, these findings suggest that restoring the innate immune signaling balance is central to recovering from alcohol-related pathology.
Collapse
Affiliation(s)
- Fulton T. Crews
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Leon G. Coleman
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Victoria A. Macht
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
3
|
Heilig M, Witkiewitz K, Ray LA, Leggio L. Novel medications for problematic alcohol use. J Clin Invest 2024; 134:e172889. [PMID: 38828724 PMCID: PMC11142745 DOI: 10.1172/jci172889] [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: 06/05/2024] Open
Abstract
Alcohol-related harm, a major cause of disease burden globally, affects people along a spectrum of use. When a harmful pattern of drinking is present in the absence of significant behavioral pathology, low-intensity brief interventions that provide information about health consequences of continued use provide large health benefits. At the other end of the spectrum, profound behavioral pathology, including continued use despite knowledge of potentially fatal consequences, warrants a medical diagnosis, and treatment is strongly indicated. Available behavioral and pharmacological treatments are supported by scientific evidence but are vastly underutilized. Discovery of additional medications, with a favorable balance of efficacy versus safety and tolerability can improve clinical uptake of treatment, allow personalized treatment, and improve outcomes. Here, we delineate the clinical conditions when pharmacotherapy should be considered in relation to the main diagnostic systems in use and discuss clinical endpoints that represent meaningful clinical benefits. We then review specific developments in three categories of targets that show promise for expanding the treatment toolkit. GPCRs remain the largest category of successful drug targets across contemporary medicine, and several GPCR targets are currently pursued for alcohol-related indications. Endocrine systems are another established category, and several promising targets have emerged for alcohol indications. Finally, immune modulators have revolutionized treatment of multiple medical conditions, and they may also hold potential to produce benefits in patients with alcohol problems.
Collapse
Affiliation(s)
- Markus Heilig
- Center for Social and Affective Neuroscience, Linköping University, and Department of Psychiatry, Linköping University Hospital, Linköping, Sweden
| | - Katie Witkiewitz
- Department of Psychology and Center on Alcohol, Substance Use and Addictions, University of New Mexico, Albuquerque, New Mexico, USA
| | - Lara A. Ray
- Department of Psychology, UCLA, Los Angeles, California, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| |
Collapse
|
4
|
Dilly GA, Blednov YA, Warden AS, Ezerskiy L, Fleischer C, Plotkin JD, Patil S, Osterndorff-Kahanek EA, Mayfield J, Mayfield RD, Homanics GE, Messing RO. Knockdown of Tlr3 in dorsal striatum reduces ethanol consumption and acute functional tolerance in male mice. Brain Behav Immun 2024; 118:437-448. [PMID: 38499210 PMCID: PMC11007683 DOI: 10.1016/j.bbi.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Systemic activation of toll-like receptor 3 (TLR3) signaling using poly(I:C), a TLR3 agonist, drives ethanol consumption in several rodent models, while global knockout of Tlr3 reduces drinking in C57BL/6J male mice. To determine if brain TLR3 pathways are involved in drinking behavior, we used CRISPR/Cas9 genome editing to generate a Tlr3 floxed (Tlr3F/F) mouse line. After sequence confirmation and functional validation of Tlr3 brain transcripts, we injected Tlr3F/F male mice with an adeno-associated virus expressing Cre recombinase (AAV5-CMV-Cre-GFP) to knockdown Tlr3 in the medial prefrontal cortex, nucleus accumbens, or dorsal striatum (DS). Only Tlr3 knockdown in the DS decreased two-bottle choice, every-other-day (2BC-EOD) ethanol consumption. DS-specific deletion of Tlr3 also increased intoxication and prevented acute functional tolerance to ethanol. In contrast, poly(I:C)-induced activation of TLR3 signaling decreased intoxication in male C57BL/6J mice, consistent with its ability to increase 2BC-EOD ethanol consumption in these mice. We also found that TLR3 was highly colocalized with DS neurons. AAV5-Cre transfection occurred predominantly in neurons, but there was minimal transfection in astrocytes and microglia. Collectively, our previous and current studies show that activating or inhibiting TLR3 signaling produces opposite effects on acute responses to ethanol and on ethanol consumption. While previous studies, however, used global knockout or systemic TLR3 activation (which alter peripheral and brain innate immune responses), the current results provide new evidence that brain TLR3 signaling regulates ethanol drinking. We propose that activation of TLR3 signaling in DS neurons increases ethanol consumption and that a striatal TLR3 pathway is a potential target to reduce excessive drinking.
Collapse
Affiliation(s)
- Geoffrey A Dilly
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Lubov Ezerskiy
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Caleb Fleischer
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Jesse D Plotkin
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Shruti Patil
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | | | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Gregg E Homanics
- Departments of Anesthesiology & Perioperative Medicine, Neurobiology, and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Robert O Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
5
|
Hitzemann R, Gao L, Fei SS, Ray K, Vigh-Conrad KA, Phillips TJ, Searles R, Cervera-Juanes RP, Khadka R, Carlson TL, Gonzales SW, Newman N, Grant KA. Effects of repeated alcohol abstinence on within-subject prefrontal cortical gene expression in rhesus macaques. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2024; 4:12528. [PMID: 38737578 PMCID: PMC11082748 DOI: 10.3389/adar.2024.12528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/12/2024] [Indexed: 05/14/2024]
Abstract
Male rhesus monkeys (n = 24) had a biopsy of prefrontal cortical area 46 prior to chronic ethanol self-administration (n = 17) or caloric control (n = 7). Fourteen months of daily self-administration (water vs. 4% alcohol, 22 h access/day termed "open-access") was followed by two cycles of prolonged abstinence (5 weeks) each followed by 3 months of open-access alcohol and a final abstinence followed by necropsy. At necropsy, a biopsy of Area 46, contralateral to the original biopsy, was obtained. Gene expression data (RNA-Seq) were collected comparing biopsy/necropsy samples. Monkeys were categorized by drinking status during the final post-abstinent drinking phase as light (LD), binge (BD), heavy (HD) and very heavy (VHD drinkers). Comparing pre-ethanol to post-abstinent biopsies, four animals that converted from HD to VHD status had significant ontology enrichments in downregulated genes (necropsy minus biopsy n = 286) that included immune response (FDR < 9 × 10-7) and plasma membrane changes (FDR < 1 × 10-7). Genes in the immune response category included IL16 and 18, CCR1, B2M, TLR3, 6 and 7, SP2 and CX3CR1. Upregulated genes (N = 388) were particularly enriched in genes associated with the negative regulation of MAP kinase activity (FDR < 3 × 10-5), including DUSP 1, 4, 5, 6 and 18, SPRY 2, 3, and 4, SPRED2, BMP4 and RGS2. Overall, these data illustrate the power of the NHP model and the within-subject design of genomic changes due to alcohol and suggest new targets for treating severe escalated drinking following repeated alcohol abstinence attempts.
Collapse
Affiliation(s)
- Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
- Portland Alcohol Research Center, Oregon Health and Science University, Portland, OR, United States
- Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Lina Gao
- Portland Alcohol Research Center, Oregon Health and Science University, Portland, OR, United States
- Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Suzanne S. Fei
- Portland Alcohol Research Center, Oregon Health and Science University, Portland, OR, United States
- Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Karina Ray
- Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Katinka A. Vigh-Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Tamara J. Phillips
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
- Portland Alcohol Research Center, Oregon Health and Science University, Portland, OR, United States
- Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Robert Searles
- Portland Alcohol Research Center, Oregon Health and Science University, Portland, OR, United States
- Integrated Genomics Laboratory, Oregon Health and Science University, Portland, OR, United States
| | - Rita P. Cervera-Juanes
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Rupak Khadka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Timothy L. Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Steven W. Gonzales
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Natali Newman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Kathleen A. Grant
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
- Portland Alcohol Research Center, Oregon Health and Science University, Portland, OR, United States
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| |
Collapse
|
6
|
Airapetov M, Eresko S, Ignatova P, Lebedev A, Bychkov E, Shabanov P. Effect of rifampicin on TLR4-signaling pathways in the nucleus accumbens of the rat brain during abstinence of long-term alcohol treatment. Alcohol Alcohol 2024; 59:agae016. [PMID: 38520481 DOI: 10.1093/alcalc/agae016] [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: 01/12/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 03/25/2024] Open
Abstract
AIMS The treatment with the antibiotic rifampicin (Rif) led to a decrease in the frequency of neurodegenerative pathologies. There are suggestions that the mechanism of action of Rif may be mediated by its effect on toll-like receptor (TLR)4-dependent pathways. We evaluated the expression status of TLR4-dependent genes during abstinence from long-term alcohol treatments in the nucleus accumbens (NAc) of the rat brain, and also studied the effects of Rif to correct these changes. METHODS The long-term alcohol treatment was performed by intragastric delivery of ethanol solution. At the end of alcohol treatment intraperitoneal injections of Rif (100 mg/kg) or saline were made. Extraction of the brain structures was performed on the 10th day of abstinence from alcohol. We used the SYBR Green qPCR method to quantitatively analyze the relative expression levels of the studied genes. RESULTS The long-term alcohol treatment promotes an increase in the level of TLR4 mRNA and mRNA of its endogenous ligand high-mobility group protein B1 during abstinence drop alcohol in NAc of rats. The use of Rif in our study led to a decrease in the increased expression of high-mobility group protein B1, Tlr4, and proinflammatory cytokine genes (Il1β, Il6) in the NAc of the rat brain during abstinence of long-term alcohol treatment. In addition, Rif administration increased the decreased mRNA levels of anti-inflammatory cytokines (Il10, Il11). CONCLUSION The data obtained indicate the ability of Rif to correct the mechanisms of the TLR4 system genes in the NAc of the rat brain during alcohol abstinence.
Collapse
Affiliation(s)
- Marat Airapetov
- Department of Neuropharmacology, Institute of Experimental Medicine, P.O. Box 197376, 12 Academician Pavlova str., St. Petersburg, Russian Federation
- Department of Pathological Physiology, Military Medical Academy of S.M. Kirov, P.O. Box 194044, 6G Akademika Lebedeva str., St. Petersburg, Russian Federation
| | - Sergei Eresko
- Department of Neuropharmacology, Institute of Experimental Medicine, P.O. Box 197376, 12 Academician Pavlova str., St. Petersburg, Russian Federation
- Center for Chemical Engineering, Saint-Petersburg National Research University of Information Technologies, Mechanics and Optics, P.O. Box 197101, 49 Kronverksky pr., St. Petersburg, Russian Federation
| | - Polina Ignatova
- Department of Neuropharmacology, Institute of Experimental Medicine, P.O. Box 197376, 12 Academician Pavlova str., St. Petersburg, Russian Federation
| | - Andrei Lebedev
- Department of Neuropharmacology, Institute of Experimental Medicine, P.O. Box 197376, 12 Academician Pavlova str., St. Petersburg, Russian Federation
| | - Evgenii Bychkov
- Department of Neuropharmacology, Institute of Experimental Medicine, P.O. Box 197376, 12 Academician Pavlova str., St. Petersburg, Russian Federation
| | - Petr Shabanov
- Department of Neuropharmacology, Institute of Experimental Medicine, P.O. Box 197376, 12 Academician Pavlova str., St. Petersburg, Russian Federation
- Department of Pathological Physiology, Military Medical Academy of S.M. Kirov, P.O. Box 194044, 6G Akademika Lebedeva str., St. Petersburg, Russian Federation
| |
Collapse
|
7
|
Grodin EN. Neuroimmune modulators as novel pharmacotherapies for substance use disorders. Brain Behav Immun Health 2024; 36:100744. [PMID: 38435721 PMCID: PMC10906159 DOI: 10.1016/j.bbih.2024.100744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/20/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
One promising avenue of research is the use of neuroimmune modulators to treat substance use disorders (SUDs). Neuroimmune modulators target the interactions between the nervous system and immune system, which have been found to play a crucial role in the development and maintenance of SUDs. Multiple classes of substances produce alterations to neuroimmune signaling and peripheral immune function, including alcohol, opioids, and psychostimulants Preclinical studies have shown that neuroimmune modulators can reduce drug-seeking behavior and prevent relapse in animal models of SUDs. Additionally, early-phase clinical trials have demonstrated the safety and feasibility of using neuroimmune modulators as a treatment for SUDs in humans. These therapeutics can be used as stand-alone treatments or as adjunctive. This review summarizes the current state of the field and provides future directions with a specific focus on personalized medicine.
Collapse
Affiliation(s)
- Erica N. Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
- Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
8
|
Lin X, Li X, Li C, Wang H, Zou L, Pan J, Zhang X, He L, Rong X, Peng Y. Activation of STING signaling aggravates chronic alcohol exposure-induced cognitive impairment by increasing neuroinflammation and mitochondrial apoptosis. CNS Neurosci Ther 2024; 30:e14689. [PMID: 38516831 PMCID: PMC10958405 DOI: 10.1111/cns.14689] [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: 11/21/2023] [Revised: 02/18/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
AIMS Chronic alcohol exposure leads to persistent neurological disorders, which are mainly attributed to neuroinflammation and apoptosis. Stimulator of IFN genes (STING) is essential in the cytosolic DNA sensing pathway and is involved in inflammation and cellular death processes. This study was to examine the expression pattern and biological functions of STING signaling in alcohol use disorder (AUD). METHODS Cell-free DNA was extracted from human and mouse plasma. C57BL/6J mice were given alcohol by gavage for 28 days, and behavior tests were used to determine their mood and cognition. Cultured cells were treated with ethanol for 24 hours. The STING agonist DMXAA, STING inhibitor C-176, and STING-siRNA were used to intervene the STING. qPCR, western blot, and immunofluorescence staining were used to assess STING signaling, inflammation, and apoptosis. RESULTS Circulating cell-free mitochondrial DNA (mtDNA) was increased in individuals with AUD and mice chronically exposed to alcohol. Upregulation of STING signaling under alcohol exposure led to inflammatory responses in BV2 cells and mitochondrial apoptosis in PC12 cells. DMXAA exacerbated alcohol-induced cognitive impairment and increased the activation of microglia, neuroinflammation, and apoptosis in the medial prefrontal cortex (mPFC), while C-176 exerted neuroprotection. CONCLUSION Activation of STING signaling played an essential role in alcohol-induced inflammation and mitochondrial apoptosis in the mPFC. This study identifies STING as a promising therapeutic target for AUD.
Collapse
Affiliation(s)
- Xinrou Lin
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
- Nanhai Translational Innovation Center of Precision ImmunologySun Yat‐Sen Memorial HospitalFoshanChina
| | - Xiangpen Li
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
- Shenshan Medical Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityShanweiChina
| | - Chenguang Li
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Hongxuan Wang
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Lubin Zou
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
- Nanhai Translational Innovation Center of Precision ImmunologySun Yat‐Sen Memorial HospitalFoshanChina
| | - Jingrui Pan
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
- Shenshan Medical Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityShanweiChina
| | - Xiaoni Zhang
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Lei He
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Xiaoming Rong
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Ying Peng
- Department of Neurology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
- Nanhai Translational Innovation Center of Precision ImmunologySun Yat‐Sen Memorial HospitalFoshanChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
9
|
Holloway KN, Douglas JC, Rafferty TM, Kane CJM, Drew PD. Ethanol Induces Neuroinflammation in a Chronic Plus Binge Mouse Model of Alcohol Use Disorder via TLR4 and MyD88-Dependent Signaling. Cells 2023; 12:2109. [PMID: 37626919 PMCID: PMC10453365 DOI: 10.3390/cells12162109] [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: 05/17/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Ethanol induces neuroinflammation, which is believed to contribute to the pathogenesis of alcohol use disorder (AUD). Toll-like receptors (TLRs) are a group of pattern recognition receptors (PRRs) expressed on both immune cells, including microglia and astrocytes, and non-immune cells in the central nervous system (CNS). Studies have shown that alcohol activates TLR4 signaling, resulting in the induction of pro-inflammatory cytokines and chemokines in the CNS. However, the effect of alcohol on signaling pathways downstream of TLR4, such as MyD88 and TRIF (TICAM) signaling, has not been evaluated extensively. In the current study, we treated male wild-type, TLR4-, MyD88-, and TRIF-deficient mice using a chronic plus binge mouse model of AUD. Evaluation of mRNA expression by qRT-PCR revealed that ethanol increased IL-1β, TNF-α, CCL2, COX2, FosB, and JunB in the cerebellum in wild-type and TRIF-deficient mice, while ethanol generally did not increase the expression of these molecules in TLR4- and MyD88-deficient mice. Furthermore, IRF3, IRF7, and IFN-β1, which are associated with the TRIF-dependent signaling cascade, were largely unaffected by alcohol. Collectively, these results suggest that the TLR4 and downstream MyD88-dependent signaling pathways are essential in ethanol-induced neuroinflammation in this mouse model of AUD.
Collapse
Affiliation(s)
- Kalee N. Holloway
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - James C. Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tonya M. Rafferty
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Cynthia J. M. Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Paul D. Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
10
|
Atkinson NS. The Role of Toll and Nonnuclear NF-κB Signaling in the Response to Alcohol. Cells 2023; 12:1508. [PMID: 37296629 PMCID: PMC10252657 DOI: 10.3390/cells12111508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
An understanding of neuroimmune signaling has become central to a description of how alcohol causes addiction and how it damages people with an AUD. It is well known that the neuroimmune system influences neural activity via changes in gene expression. This review discusses the roles played by CNS Toll-like receptor (TLR) signaling in the response to alcohol. Also discussed are observations in Drosophila that show how TLR signaling pathways can be co-opted by the nervous system and potentially shape behavior to a far greater extent and in ways different than generally recognized. For example, in Drosophila, TLRs substitute for neurotrophin receptors and an NF-κB at the end of a TLR pathway influences alcohol responsivity by acting non-genomically.
Collapse
Affiliation(s)
- Nigel S Atkinson
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
11
|
Xiao Q, Chen YH, Chen YL, Chien YS, Hsieh LH, Shirakawa H, Yang SC. Potential Benefits of Epidermal Growth Factor for Inhibiting Muscle Degrative Markers in Rats with Alcoholic Liver Damage. Int J Mol Sci 2023; 24:ijms24108845. [PMID: 37240190 DOI: 10.3390/ijms24108845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/29/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
This study investigated the beneficial effects of epidermal growth factor (EGF) on muscle loss in rats with chronic ethanol feeding. Six-week-old male Wistar rats were fed either a control liquid diet without EGF (C group, n = 12) or EGF (EGF-C group, n = 18) for two weeks. From the 3rd to 8th week, the C group was divided into two groups. One was continually fed with a control liquid diet (C group), and the other one was fed with an ethanol-containing liquid diet (E group); moreover, the EGF-C group was divided into three groups, such as the AEGF-C (continually fed with the same diet), PEGF-E (fed with the ethanol-containing liquid diet without EGF), and AEGF-E (fed with the ethanol-containing liquid diet with EGF). As a result, the E group had significantly higher plasma ALT and AST, endotoxin, ammonia, and interleukin 1b (IL-1b) levels, along with liver injuries, such as hepatic fatty changes and inflammatory cell infiltration. However, plasma endotoxin and IL-1b levels were significantly decreased in the PEGF-E and AEGF-E groups. In addition, the protein level of muscular myostatin and the mRNA levels of forkhead box transcription factors (FOXO), muscle RING-finger protein-1 (MURF-1) and atorgin-1 was increased considerably in the E group but inhibited in the PEGF-E and AEGF-E groups. According to the principal coordinate analysis findings, the gut microbiota composition differed between the control and ethanol liquid diet groups. In conclusion, although there was no noticeable improvement in muscle loss, EGF supplementation inhibited muscular protein degradation in rats fed with an ethanol-containing liquid diet for six weeks. The mechanisms might be related to endotoxin translocation inhibition, microbiota composition alteration as well as the amelioration of liver injury. However, the reproducibility of the results must be confirmed in future studies.
Collapse
Affiliation(s)
- Qian Xiao
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Hsiu Chen
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Ya-Ling Chen
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Shan Chien
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Li-Hsuan Hsieh
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Hitoshi Shirakawa
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8857, Japan
| | - Suh-Ching Yang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- School of Gerontology and Long-Term Care, College of Nursing, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
12
|
Varodayan FP, Pahng AR, Davis TD, Gandhi P, Bajo M, Steinman MQ, Kiosses WB, Blednov YA, Burkart MD, Edwards S, Roberts AJ, Roberto M. Chronic ethanol induces a pro-inflammatory switch in interleukin-1β regulation of GABAergic signaling in the medial prefrontal cortex of male mice. Brain Behav Immun 2023; 110:125-139. [PMID: 36863493 PMCID: PMC10106421 DOI: 10.1016/j.bbi.2023.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Neuroimmune pathways regulate brain function to influence complex behavior and play a role in several neuropsychiatric diseases, including alcohol use disorder (AUD). In particular, the interleukin-1 (IL-1) system has emerged as a key regulator of the brain's response to ethanol (alcohol). Here we investigated the mechanisms underlying ethanol-induced neuroadaptation of IL-1β signaling at GABAergic synapses in the prelimbic region of the medial prefrontal cortex (mPFC), an area responsible for integrating contextual information to mediate conflicting motivational drives. We exposed C57BL/6J male mice to the chronic intermittent ethanol vapor-2 bottle choice paradigm (CIE-2BC) to induce ethanol dependence, and conducted ex vivo electrophysiology and molecular analyses. We found that the IL-1 system regulates basal mPFC function through its actions at inhibitory synapses on prelimbic layer 2/3 pyramidal neurons. IL-1β can selectively recruit either neuroprotective (PI3K/Akt) or pro-inflammatory (MyD88/p38 MAPK) mechanisms to produce opposing synaptic effects. In ethanol naïve conditions, there was a strong PI3K/Akt bias leading to a disinhibition of pyramidal neurons. Ethanol dependence produced opposite IL-1 effects - enhanced local inhibition via a switch in IL-1β signaling to the canonical pro-inflammatory MyD88 pathway. Ethanol dependence also increased cellular IL-1β in the mPFC, while decreasing expression of downstream effectors (Akt, p38 MAPK). Thus, IL-1β may represent a key neural substrate in ethanol-induced cortical dysfunction. As the IL-1 receptor antagonist (kineret) is already FDA-approved for other diseases, this work underscores the high therapeutic potential of IL-1 signaling/neuroimmune-based treatments for AUD.
Collapse
Affiliation(s)
- F P Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA; Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - A R Pahng
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - T D Davis
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY, USA
| | - P Gandhi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - M Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - M Q Steinman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - W B Kiosses
- Microscopy Core Imaging Facility, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Y A Blednov
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - M D Burkart
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - S Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - A J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, USA
| | - M Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
13
|
Randall CA, Sun D, Randall PA. Differential Effects of Nicotine, Alcohol, and Coexposure on Neuroimmune-Related Protein and Gene Expression in Corticolimbic Brain Regions of Rats. ACS Chem Neurosci 2023; 14:628-644. [PMID: 36705334 DOI: 10.1021/acschemneuro.2c00413] [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: 01/28/2023] Open
Abstract
Nicotine and alcohol co-use is extremely common and their use constitutes two of the most common causes of preventable death, yet the underlying biological mechanisms are largely understudied. Activation of neuroimmune toll-like receptors (TLRs) promotes the induction of proinflammatory cascades and increases alcohol intake in rodents, which further promotes TLRs in the brain; nicotine may decrease central proinflammatory signaling. The current studies sought to determine the effects of nicotine ± alcohol (alone or in combination) on circulating blood plasma and TLR protein/gene expression in addiction-associated corticolimbic brain regions, including the prefrontal cortex-prelimbic (mPFC-PL) and nucleus accumbens core (AcbC). Adult rats were treated with alcohol (0 or 2 g/kg, IG) and exposed to nicotine vapor (0 or 30 mg/mL solution) daily for 2, 14, or 28 days. Plasma studies indicated no effects of independent exposure or coexposure in males. Coexposure decreased plasma nicotine levels versus nicotine-only treated females, yet alcohol and cotinine concentrations were unchanged. By 28 days, the anti-inflammatory cytokine IL-13 was decreased in alcohol-only females. Divergent changes in TLR3 (but not TLR4) protein occurred for independent-drug exposed males (but not coexposure), with reductions in the mPFC-PL after 14 days and increases in the AcbC by 28 days. Gene expression following chronic coexposure suggests nicotine may regionally counteract alcohol-induced inflammation, including increased AcbC-TLR3/4/7 and several downstream markers in females and increased mPFC-PL-TLR3 and -STAT3 (but not IRF3) evident in males with exposure to either drug alone. These findings give further insight into the role of sex and the neuroimmune system in independent exposure and coexposure to nicotine ± alcohol.
Collapse
Affiliation(s)
- Christie A Randall
- Department of Anesthesiology and Perioperative Medicine, College of Medicine, Pennsylvania State University, 500 University Drive, Hershey, Pennsylvania 17033, United States
| | - Dongxiao Sun
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania 17033 United States
| | - Patrick A Randall
- Department of Anesthesiology and Perioperative Medicine, College of Medicine, Pennsylvania State University, 500 University Drive, Hershey, Pennsylvania 17033, United States.,Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania 17033 United States
| |
Collapse
|
14
|
Grantham EK, Barchiesi R, Salem NA, Mayfield RD. Neuroimmune pathways as targets to reduce alcohol consumption. Pharmacol Biochem Behav 2023; 222:173491. [PMID: 36400266 PMCID: PMC9906983 DOI: 10.1016/j.pbb.2022.173491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/04/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Affiliation(s)
| | - Riccardo Barchiesi
- Waggoner Center for Alcohol and Addiction Research and the Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, United States.
| | | | | |
Collapse
|
15
|
Airapetov MI, Eresko SO, Kochkin DV, Nosov AM, Bychkov ER, Lebedev AA, Shabanov PD. [Ginsenosides affect the system of Toll-like receptors in the brain of rats under conditions of long-term alcohol withdrawal]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:459-469. [PMID: 36573411 DOI: 10.18097/pbmc20226806459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Long-term alcohol consumption causes the development of neuroinflammation in various brain structures. One of the mechanisms involved in this process is the increased activity of TLR-signaling intracellular pathways. Studies confirm the ability of ginseng extract or its individual ginsenosides to reduce the increased activity of TLR-signaling pathways. The aim of our study was to study the effect of the amount of ginsenosides obtained from the extract of the Panax japonicus cell line on the state of the TLR-signaling system in the nucleus accumbens and hippocampus of the rat brain in a model of long-term alcohol consumption during alcohol withdrawal. The results of the study showed that ginsenosides were able to make changes in the TLR signaling system, which has been altered by long-term alcohol consumption. A significant effect of ginsenosides on the level of TLR3 and TLR4 mRNA in the nucleus accumbens was found, while in the hippocampus, ginsenosides significantly affected the level of TLR7 mRNA. The effect of ginsenosides on the level of mRNA of transcription factors and cytokines involved in TLR-signaling was evaluated. Thus, results of our study confirm that ginsenosides are able to influence the state of TLR-signaling pathways, but this effect is multidirectional in relation to different brain structures. In the future, it seems interesting to evaluate the role of individual ginsenosides in relation to genes of TLR-signaling, as well as the effect of ginsenosides on other brain structures.
Collapse
Affiliation(s)
- M I Airapetov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia; St. Petersburg State Pediatric Medical University, St. Petersburg, Russia
| | - S O Eresko
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia; Research and Training Center of Molecular and Cellular Technologies, St. Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | - D V Kochkin
- Department of Plant Physiology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - A M Nosov
- Department of Plant Physiology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - E R Bychkov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - A A Lebedev
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - P D Shabanov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia; Department of Pharmacology, Kirov Military Medical Academy, St. Petersburg, Russia
| |
Collapse
|
16
|
Squillace S, Salvemini D. Toll-like receptor-mediated neuroinflammation: relevance for cognitive dysfunctions. Trends Pharmacol Sci 2022; 43:726-739. [PMID: 35753845 PMCID: PMC9378500 DOI: 10.1016/j.tips.2022.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/13/2022] [Accepted: 05/27/2022] [Indexed: 10/17/2022]
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) with a well-documented role in the innate and adaptive immune responses. Interestingly, TLR activation has also been linked to several brain functions including neurogenesis and synaptogenesis. Increasing evidence supports TLR involvement in peripheral and central inflammation underlying normal aging and the pathogenesis of clinical conditions characterized by cognitive decline. These include not only major neurodegenerative diseases but also traumatic brain injuries, surgeries, and alcohol consumption- and chemotherapy-induced cognitive impairment. We first summarize the physiological roles of TLRs in the nervous system, and then illustrate the emerging involvement of TLRs in cognitive functions, pointing to these receptors as novel enticing pharmacological targets to develop more efficient drugs for the treatment of cognitive impairment.
Collapse
Affiliation(s)
- Silvia Squillace
- Department of Pharmacology and Physiology, and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St. Louis, MO 63104, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St. Louis, MO 63104, USA.
| |
Collapse
|
17
|
Airapetov MI, Eresko SO, Skabelkin DA, Iskalieva AR, Lebedev AA, Bychkov ER, Shabanov PD. [The effect of rifampicin on the system of Toll-like receptors in the nucleus accumbens of the brain of long-term alcoholized rats during alcohol withdrawal]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:279-287. [PMID: 36005846 DOI: 10.18097/pbmc20226804279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nucleus accumbens (NAc) is the ventral part of the striatum of the brain; it is an important part of the mesolimbic pathway involved in the reward system that mediates the formation of various forms of addiction, in particular alcohol addiction. Neuroimaging data and in vitro studies indicate the development of a pronounced neurodegenerative process in the NAc, with long-term alcohol use, but the key mechanisms mediating this process remain unknown. In recent years, the attention of researchers has been focused on studying the system of Toll-like receptors (TLRs), the increased activity of which is clearly shown in the cerebral cortex and hippocampus during prolonged alcohol exposure, but there is a need to study the role of this system in other brain structures. In this study, we have shown that prolonged alcohol exposure (2 months) with moderate doses of ethanol (2 g/kg) promotes a pronounced increase in the expression of the Tlr4 gene and its endogenous ligand Hmgb1 in NAc during the period of alcohol withdrawal in rats. Injections of rifampicin (100 mg/kg) reduced the elevated expression level of Hmgb1, Tlr4, as well as pro-inflammatory cytokine genes (IL1β, IL6), while the administration of the drug increased the reduced level of mRNA of anti-inflammatory cytokines (IL10, IL11).
Collapse
Affiliation(s)
- M I Airapetov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia; Department of Pharmacology, St. Petersburg State Pediatric Medical University
| | - S O Eresko
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia; Research and Training Center of Molecular and Cellular Technologies, St. Petersburg, Russia
| | - D A Skabelkin
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - A R Iskalieva
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - A A Lebedev
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - E R Bychkov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - P D Shabanov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia; Department of Pharmacology, Kirov Military Medical Academy, St. Petersburg, Russia
| |
Collapse
|
18
|
Lovelock DF, Randall PA, Van Voorhies K, Vetreno RP, Crews FT, Besheer J. Increased alcohol self-administration following repeated Toll-like receptor 3 agonist treatment in male and female rats. Pharmacol Biochem Behav 2022; 216:173379. [PMID: 35395252 PMCID: PMC9263963 DOI: 10.1016/j.pbb.2022.173379] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023]
Abstract
Toll-like receptor (TLR) signaling may play an important role in the neuroimmune system's involvement in the development and maintenance of alcohol use disorder (AUD). In the present study we administered the TLR3 agonist poly(I:C) in male and female Long-Evans rats to determine whether TLR3 agonism can increase alcohol consumption on a daily 15% alcohol operant self-administration paradigm. We found few effects when poly(I:C) was given every-other-day at 0.3 or 1.0 mg/kg. However, when 1.0 mg/kg was given on consecutive days, alcohol intake increased in the days following injections specifically in females. In a second experiment, we found that this effect only emerged when rats had a history of multiple poly(I:C) injections. In the final experiment the poly(I:C) dose was increased to 3.0 mg/kg on consecutive days which resulted in significant reductions in alcohol intake on injection days in females that were not accompanied by subsequent increases. The poly(I:C) dose was increased to 9.0 mg/kg for one final pair of injections which led to reductions in intake in both males and females followed by a male specific delayed increase in alcohol intake. Overall, repeated poly(I:C) administration was able to increase subsequent alcohol consumption in both sexes, with females showing an increase at a lower dose than males. These findings support TLR3 agonism in contributing to increased alcohol consumption and add to the body of work identifying the neuroimmune system as a potential therapeutic target for AUD.
Collapse
Affiliation(s)
- Dennis F Lovelock
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick A Randall
- Department of Anesthesiology & Perioperative Medicine, Penn State College of Medicine, Hershey, PA 17033, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Kalynn Van Voorhies
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
19
|
Prenatal and adolescent alcohol exposure programs immunity across the lifespan: CNS-mediated regulation. Pharmacol Biochem Behav 2022; 216:173390. [PMID: 35447157 DOI: 10.1016/j.pbb.2022.173390] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/28/2022] [Accepted: 04/11/2022] [Indexed: 12/31/2022]
Abstract
For many individuals, first exposure to alcohol occurs either prenatally due to maternal drinking, or during adolescence, when alcohol consumption is most likely to be initiated. Prenatal Alcohol Exposure (PAE) and its associated Fetal Alcohol Spectrum Disorders (FASD) in humans is associated with earlier initiation of alcohol use and increased rates of Alcohol Use Disorders (AUD). Initiation of alcohol use and misuse in early adolescence correlates highly with later AUD diagnosis as well. Thus, PAE and adolescent binge drinking set the stage for long-term health consequences due to adverse effects of alcohol on subsequent immune function, effects that may persist across the lifespan. The overarching goal of this review, therefore, is to determine the extent to which early developmental exposure to alcohol produces long-lasting, and potentially life-long, changes in immunological function. Alcohol affects the whole body, yet most studies are narrowly focused on individual features of immune function, largely ignoring the systems-level interactions required for effective host defense. We therefore emphasize the crucial role of the Central Nervous System (CNS) in orchestrating host defense processes. We argue that alcohol-mediated disruption of host immunity can occur through both (a) direct action of ethanol on neuroimmune processes, that subsequently disrupt peripheral immune function (top down); and (b) indirect action of ethanol on peripheral immune organs/cells, which in turn elicit consequent changes in CNS neuroimmune function (bottom up). Recognizing that alcohol consumption across the entire body, we argue in favor of integrative, whole-organism approaches toward understanding alcohol effects on immune function, and highlight the need for more work specifically examining long-lasting effects of early developmental exposure to alcohol (prenatal and adolescent periods) on host immunity.
Collapse
|
20
|
Astrocytes play a critical role in mediating the effect of acute ethanol on central amygdala glutamatergic transmission. Neuropharmacology 2022; 205:108918. [PMID: 34896402 PMCID: PMC8792276 DOI: 10.1016/j.neuropharm.2021.108918] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022]
Abstract
The Central Amygdala (CeA) has been heavily implicated in many aspects of alcohol use disorder. Ethanol (EtOH) has been shown to modulate glutamatergic transmission in the lateral subdivision of the CeA, however, the exact mechanism of this modulation is still unclear. EtOH exposure is associated with increased pro-inflammatory cytokines in the CeA, and inhibition of neuroimmune cells (microglia and astrocytes) has previously been shown to reduce EtOH drinking in animal models. Since neuroimmune activation seems to be involved in many of the effects of EtOH, we hypothesized that acute EtOH exposure will increase excitatory glutamatergic transmission in the CeA via modulation of neuroimmune cells. Using ex vivo brain slice whole-cell patch clamp electrophysiology, it was found that a physiologically relevant concentration of EtOH (20 mM) significantly increased presynaptic glutamatergic transmission in the CeA. Pharmacologic and chemogenetic inhibition of astrocyte function significantly reduced the ability of EtOH to modulate CeA glutamatergic transmission with minimal impact of microglia inhibition. This finding prompted additional studies examining whether direct neuroimmune activation through lipopolysaccharide (LPS) might lead to an increase in the glutamatergic transmission in the CeA. It was found that LPS modulation of glutamatergic transmission was limited by microglia activation and required astrocyte signaling. Taken together these results support the hypothesis that acute EtOH enhances lateral CeA glutamatergic transmission through an astrocyte mediated mechanism.
Collapse
|
21
|
Czerwińska-Błaszczyk A, Pawlak E, Pawłowski T. The Significance of Toll-Like Receptors in the Neuroimmunologic Background of Alcohol Dependence. Front Psychiatry 2022; 12:797123. [PMID: 35095609 PMCID: PMC8791063 DOI: 10.3389/fpsyt.2021.797123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/13/2021] [Indexed: 01/02/2023] Open
Abstract
Toll-like receptors (TLR) are a group of protein belonging to the family of Pattern Recognition Receptors (PRR) which have the ability to distinguish between an organism's own antigens and foreign ones and to induce immunological response. TLR play a significant part in non-specific immunity but at the same time they are also a vital element linking non-specific response to the specific one. A growing number of data seems to indicate that the non-specific immunity mechanisms affect the development and sustenance of alcohol addiction. Alcohol damages the organism's cells not only directly but also through an increase inintestinal permeability which induces innate immune response of peripheral blood cells. The signaling pathway of Toll-like receptors located on the surface of brain immune cells intensifies the inflammatory reaction and, through modifying gene expression of proinflammatory factors, unnaturally supports it. This overly protracted "sterile inflammatory reaction" positively correlates with alcohol craving affecting also the functioning of the reward system structures and increasing the risk of relapse of alcoholism. Recurrent alcoholic binges sensitize the microglia and cause an escalation in inflammatory reaction which also leads to neurodegeneration. The induction of innate immunity signaling pathways exposes clinical symptoms of alcohol addiction such as increased impulsivity, loss of behavioral control, depressive-anxiety symptoms and cognitive dysfunctions. Traditional methods of treating alcohol addiction have tended to focus predominantly on reducing symptoms which-given the frequency of relapses-seems insufficient. The aim of the present paper is to discuss the role of toll-like receptors as elements of the immunity system which, together with the nervous system, plays a crucial part in the pathogenesis of alcohol addiction. We also wish to present pharmacotherapeutic perspectives targeted at the neuroimmunological mechanisms of alcohol addiction.
Collapse
Affiliation(s)
| | - Edyta Pawlak
- Laboratory of Immunopatology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Tomasz Pawłowski
- Division of Psychotherapy and Psychosomatic Medicine, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
22
|
Neuroadaptations and TGF-β signaling: emerging role in models of neuropsychiatric disorders. Mol Psychiatry 2022; 27:296-306. [PMID: 34131268 PMCID: PMC8671568 DOI: 10.1038/s41380-021-01186-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023]
Abstract
Neuropsychiatric diseases are manifested by maladaptive behavioral plasticity. Despite the greater understanding of the neuroplasticity underlying behavioral adaptations, pinpointing precise cellular mediators has remained elusive. This has stymied the development of pharmacological interventions to combat these disorders both at the level of progression and relapse. With increased knowledge on the putative role of the transforming growth factor (TGF- β) family of proteins in mediating diverse neuroadaptations, the influence of TGF-β signaling in regulating maladaptive cellular and behavioral plasticity underlying neuropsychiatric disorders is being increasingly elucidated. The current review is focused on what is currently known about the TGF-β signaling in the central nervous system in mediating cellular and behavioral plasticity related to neuropsychiatric manifestations.
Collapse
|
23
|
Morcuende A, Navarrete F, Nieto E, Manzanares J, Femenía T. Inflammatory Biomarkers in Addictive Disorders. Biomolecules 2021; 11:biom11121824. [PMID: 34944470 PMCID: PMC8699452 DOI: 10.3390/biom11121824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Substance use disorders are a group of diseases that are associated with social, professional, and family impairment and that represent a high socio-economic impact on the health systems of countries around the world. These disorders present a very complex diagnosis and treatment regimen due to the lack of suitable biomarkers supporting the correct diagnosis and classification and the difficulty of selecting effective therapies. Over the last few years, several studies have pointed out that these addictive disorders are associated with systemic and central nervous system inflammation, which could play a relevant role in the onset and progression of these diseases. Therefore, identifying different immune system components as biomarkers of such addictive disorders could be a crucial step to promote appropriate diagnosis and treatment. Thus, this work aims to provide an overview of the immune system alterations that may be biomarkers of various addictive disorders.
Collapse
Affiliation(s)
- Alvaro Morcuende
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Elena Nieto
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Teresa Femenía
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-965-919-553
| |
Collapse
|
24
|
Meredith LR, Burnette EM, Grodin EN, Irwin MR, Ray LA. Immune treatments for alcohol use disorder: A translational framework. Brain Behav Immun 2021; 97:349-364. [PMID: 34343618 PMCID: PMC9044974 DOI: 10.1016/j.bbi.2021.07.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/10/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
While the immune system is essential for survival, an excessive or prolonged inflammatory response, such as that resulting from sustained heavy alcohol use, can damage the host and contribute to psychiatric disorders. A growing body of literature indicates that the immune system plays a critical role in the development and maintenance of alcohol use disorder (AUD). As such, there is enthusiasm for treatments that can restore healthy levels of inflammation as a mechanism to reduce drinking and promote recovery. In this qualitative literature review, we provide a conceptual rationale for immune therapies and discuss progress in medications development for AUD focused on the immune system as a treatment target. This review is organized into sections based on primary signaling pathways targeted by the candidate therapies, namely: (a) toll-like receptors, (b) phosphodiesterase inhibitors, (c) peroxisome proliferator-activated receptors, (d) microglia and astrocytes, (e) other immune pharmacotherapies, and (f) behavioral therapies. As relevant within each section, we examine the basic biological mechanisms of each class of therapy and evaluate preclinical research testing the role of the therapy on mitigating alcohol-related behaviors in animal models. To the extent available, translational findings are reviewed with discussion of completed and ongoing randomized clinical trials and their findings to date. An applied and clinically focused approach is taken to identify the potential clinical applications of the various treatments reviewed. We conclude by delineating the most promising candidate treatments and discussing future directions by considering opportunities for immune treatment development and personalized medicine for AUD.
Collapse
Affiliation(s)
- Lindsay R Meredith
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elizabeth M Burnette
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erica N Grodin
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael R Irwin
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA; Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA, USA; Cousins Center for Psychoneuroimmunology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Vetreno RP, Qin L, Coleman LG, Crews FT. Increased Toll-like Receptor-MyD88-NFκB-Proinflammatory neuroimmune signaling in the orbitofrontal cortex of humans with alcohol use disorder. Alcohol Clin Exp Res 2021; 45:1747-1761. [PMID: 34415075 PMCID: PMC8526379 DOI: 10.1111/acer.14669] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/02/2021] [Accepted: 07/07/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND Many brain disorders, including alcohol use disorder (AUD), are associated with induction of multiple proinflammatory genes. One aspect of proinflammatory signaling is progressive increases in expression across cells and induction of other innate immune genes. High-mobility group box 1 (HMGB1) heteromers contribute to amplification by potentiating multiple proinflammatory responses, including Toll-like receptors (TLRs). TLR signaling recruits coupling proteins linked to nuclear transcription factors that induce proinflammatory cytokines and chemokines and their respective receptors. We tested the hypothesis that AUD induction of TLR expression increases levels of proinflammatory genes and cellular signaling cascades in association with neurodegeneration in the orbitofrontal cortex (OFC). METHODS Postmortem human OFC tissue samples (n = 10) from males diagnosed with AUD were compared to age-matched moderate drinking controls (CON). Neuroimmune signaling molecules were assessed using immunohistochemistry for protein and reverse transcription polymerase chain reaction for messenger RNA (mRNA). RESULTS In the AUD OFC, we report induction of the endogenous TLR agonist HMGB1 as well as all TLRs assessed (i.e., TLR2-TLR9) except TLR1. This was accompanied by increased expression of the TLR adaptor protein myeloid differentiation primary response 88 (MyD88), activation of the proinflammatory nuclear transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), and downstream induction of proinflammatory cytokines, chemokines, and their corresponding receptors. Several of these proinflammatory signaling markers are expressed in glia and neurons. The induction of HMGB1-TLR-MyD88-NFκB proinflammatory signaling pathways correlates with neurodegeneration (i.e., Fluoro-Jade B), lifetime alcohol consumption, and age of drinking onset. CONCLUSION These data implicate the induction of HMGB1-TLR-MyD88-NFκB cascades through coordinated glial and neuronal signaling as contributors to the neurodegeneration seen in the postmortem human OFC of individuals with AUD.
Collapse
Affiliation(s)
- Ryan P. Vetreno
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PsychiatrySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Liya Qin
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Leon G. Coleman
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PharmacologySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Fulton T. Crews
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PsychiatrySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PharmacologySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
26
|
Kisby BR, Farris SP, McManus MM, Varodayan FP, Roberto M, Harris RA, Ponomarev I. Alcohol Dependence in Rats Is Associated with Global Changes in Gene Expression in the Central Amygdala. Brain Sci 2021; 11:1149. [PMID: 34573170 PMCID: PMC8468792 DOI: 10.3390/brainsci11091149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/06/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol dependence is associated with adverse consequences of alcohol (ethanol) use and is evident in most severe cases of alcohol use disorder (AUD). The central nucleus of the amygdala (CeA) plays a critical role in the development of alcohol dependence and escalation of alcohol consumption in dependent subjects. Molecular mechanisms underlying the CeA-driven behavioral changes are not well understood. Here, we examined the effects of alcohol on global gene expression in the CeA using a chronic intermittent ethanol (CIE) vapor model in rats and RNA sequencing (RNA-Seq). The CIE procedure resulted in robust changes in CeA gene expression during intoxication, as the number of differentially expressed genes (DEGs) was significantly greater than those expected by chance. Over-representation analysis of cell types, functional groups and molecular pathways revealed biological categories potentially important for the development of alcohol dependence in our model. Genes specific for astrocytes, myelinating oligodendrocytes, and endothelial cells were over-represented in the DEG category, suggesting that these cell types were particularly affected by the CIE procedure. The majority of the over-represented functional groups and molecular pathways were directly related to the functions of glial and endothelial cells, including extracellular matrix (ECM) organization, myelination, and the regulation of innate immune response. A coordinated regulation of several ECM metalloproteinases (e.g., Mmp2; Mmp14), their substrates (e.g., multiple collagen genes and myelin basic protein; Mbp), and a metalloproteinase inhibitor, Reck, suggests a specific mechanism for ECM re-organization in response to chronic alcohol, which may modulate neuronal activity and result in behavioral changes, such as an escalation of alcohol drinking. Our results highlight the importance of glial and endothelial cells in the effects of chronic alcohol exposure on the CeA, and demonstrate further insight into the molecular mechanisms of alcohol dependence in rats. These molecular targets may be used in future studies to develop therapeutics to treat AUD.
Collapse
Affiliation(s)
- Brent R. Kisby
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.R.K.); (M.M.M.)
| | - Sean P. Farris
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78715, USA; (S.P.F.); (R.A.H.)
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Michelle M. McManus
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.R.K.); (M.M.M.)
| | - Florence P. Varodayan
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY 13902, USA;
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - R. Adron Harris
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78715, USA; (S.P.F.); (R.A.H.)
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78741, USA
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.R.K.); (M.M.M.)
| |
Collapse
|
27
|
Airapetov MI, Eresko SO, Vasilev AK, Vasileva VY, Bychkov ER, Lebedev AA, Shabanov PD. [The TLR3 induction increases content of interferons in rat's brain by TRAIL signaling during long-term alcoholization]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:331-337. [PMID: 34414891 DOI: 10.18097/pbmc20216704331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The pathogenetic mechanisms associated with alcohol use include dysregulation of the innate immune system mechanisms in the brain. TLR3 expression is increased in the postmortem material of the prefrontal cortex of humans. An increase in the TLR3 signaling activity leads to the induction of interferons (IFN). IFN are associated with depressive symptoms and, therefore, may play a role in the pathogenesis of alcoholism; however, the exact mechanisms of intracellular signaling mediated by the influence of ethanol are not fully elucidated and their study was the purpose of this work. The experimental results showed that ethanol and the TLR3 agonist Poly (I:C) increased the content of TLR3, IFNβ, and IFNγ mRNA in the prefrontal cortex. In addition, expression of the TRAIL encoding gene also increased, and this increase positively correlaed with the mRNA content of TLR3, IFNβ and IFNγ both under alcoholization conditions and after injections of the TLR3 agonist. The data obtained may indicate that alcoholization is able to activate TLR3-TRAIL-IFN-signaling in the prefrontal cortex of the brain.
Collapse
Affiliation(s)
- M I Airapetov
- Institute of Experimental Medicine, St. Petersburg, Russia; Saint Petersburg State Pediatric Medical University, St. Petersburg, Russia
| | - S O Eresko
- Institute of Experimental Medicine, St. Petersburg, Russia; Saint-Petersburg State Chemical and Pharmaceutical University, St. Petersburg, Russia
| | - A K Vasilev
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - V Y Vasileva
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - E R Bychkov
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - A A Lebedev
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - P D Shabanov
- Institute of Experimental Medicine, St. Petersburg, Russia; Kirov Military Medical Academy, St. Petersburg, Russia
| |
Collapse
|
28
|
Airapetov M, Eresko S, Lebedev A, Bychkov E, Shabanov P. The role of Toll-like receptors in neurobiology of alcoholism. Biosci Trends 2021; 15:74-82. [PMID: 33716257 DOI: 10.5582/bst.2021.01041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Alcoholism is a global socially significant problem and still remains one of the leading causes of disability and premature death. One of the main signs of the disease is the loss of cognitive control over the amount of alcohol consumed. Among the mechanisms of the development of this pathology, changes in neuroimmune mechanisms occurring in the brain during prolonged alcohol consumption and its withdrawal have recently become the focus of numerous studies. Ethanol consumption leads to the activation of neuroimmune signaling in the central nervous system through many subtypes of Toll-like receptors (TLRs), as well as release of their endogenous agonists (high-mobility group protein B1 (HMGB1), S100 protein, heat shock proteins (HSPs), and extracellular matrix degradation proteins). TLR activation triggers intracellular molecular cascades of reactions leading to increased expression of genes of the innate immune system, particularly, proinflammatory cytokines, causing further development of a persistent neuroinflammatory process in the central nervous system. This leads to death of neurons and neuroglial cells in various brain structures, primarily in those associated with the development of a pathological craving for alcohol. In addition, there is evidence that some subtypes of TLRs (TLR3, TLR4) are able to form heterodimers with neuropeptide receptors, thereby possibly playing other roles in the central nervous system, in addition to participating in the activation of the innate immune system.
Collapse
Affiliation(s)
- Marat Airapetov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia.,Department of Pharmacology, St. Petersburg State Pediatric Medical University, St. Petersburg, Russia
| | - Sergei Eresko
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia.,Research and Education Center for Molecular and Cellular Technologies, St. Petersburg State Chemical Pharmaceutical University, St Petersburg, Russia
| | - Andrei Lebedev
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Evgenii Bychkov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Petr Shabanov
- Department of Neuropharmacology, Institute of Experimental Medicine, St. Petersburg, Russia.,Department of Pharmacology, Kirov Military Medical Academy, St. Petersburg, Russia
| |
Collapse
|
29
|
Blednov YA, Da Costa A, Mayfield J, Harris RA, Messing RO. Deletion of Tlr3 reduces acute tolerance to alcohol and alcohol consumption in the intermittent access procedure in male mice. Addict Biol 2021; 26:e12932. [PMID: 32604471 DOI: 10.1111/adb.12932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 04/28/2020] [Accepted: 06/09/2020] [Indexed: 02/02/2023]
Abstract
Pharmacological studies implicate toll-like receptor 3 (TLR3) signaling in alcohol drinking. We examined the role of TLR3 in behavioral responses to alcohol and GABAergic drugs by studying Tlr3 -/- mice. Because of opposing signaling between TLR3 and MyD88 pathways, we also evaluated Myd88 -/- mice. Ethanol consumption and preference decreased in male but not in female Tlr3 -/- mice during two-bottle choice every-other-day (2BC-EOD) drinking. There were no genotype differences in either sex during continuous or limited-access drinking. Null mutations in Tlr3 or Myd88 did not alter conditioned taste aversion to alcohol and had small or no effects on conditioned place preference. The Tlr3 null mutation did not alter acute alcohol withdrawal. Male, but not female, Tlr3 -/- mice took longer than wild-type littermates to recover from ataxia by ethanol or diazepam and longer to recover from sedative-hypnotic effects of ethanol or gaboxadol, indicating regulation of GABAergic signaling by TLR3. Acute functional tolerance (AFT) to alcohol-induced ataxia was decreased in Tlr3 -/- mice but was increased in Myd88 -/- mice. Thus, MyD88 and TLR3 pathways coordinately regulate alcohol consumption and tolerance to intoxicating doses of alcohol and GABAergic drugs. Despite similar alcohol metabolism and similar amounts of total alcohol consumed during 2BC and 2BC-EOD procedures in C57BL/6J mice, only 2BC-EOD drinking induced tolerance to alcohol-induced ataxia. Ataxia recovery was inversely correlated with level of drinking in wild-type and Tlr3 -/- littermates. Thus, deleting Tlr3 reduces alcohol consumption by reducing AFT to alcohol and not by altering tolerance induced by 2BC-EOD drinking.
Collapse
Affiliation(s)
- Yuri A. Blednov
- Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - Adriana Da Costa
- Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
| | - R. Adron Harris
- Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
- Department of Neuroscience The University of Texas at Austin Austin Texas USA
| | - Robert O. Messing
- Waggoner Center for Alcohol and Addiction Research The University of Texas at Austin Austin Texas USA
- Department of Neuroscience The University of Texas at Austin Austin Texas USA
- Department of Neurology, Dell Medical School The University of Texas at Austin Austin Texas USA
| |
Collapse
|
30
|
Crews FT, Zou J, Coleman LG. Extracellular microvesicles promote microglia-mediated pro-inflammatory responses to ethanol. J Neurosci Res 2021; 99:1940-1956. [PMID: 33611821 PMCID: PMC8451840 DOI: 10.1002/jnr.24813] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
Alcohol use disorder (AUD) pathology features pro-inflammatory gene induction and microglial activation. The underlying cellular processes that promote this activation remain unclear. Previously considered cellular debris, extracellular vesicles (EVs) have emerged as mediators of inflammatory signaling in several disease states. We investigated the role of microvesicles (MVs, 50 nm-100 µm diameter EVs) in pro-inflammatory and microglial functional gene expression using primary organotypic brain slice culture (OBSC). Ethanol caused a unique immune gene signature that featured: temporal induction of pro-inflammatory TNF-α and IL-1β, reduction of homeostatic microglia state gene Tmem119, progressive increases in purinergic receptor P2RY12 and the microglial inhibitory fractalkine receptor CX3CR1, an increase in the microglial presynaptic gene C1q, and a reduction in the phagocytic gene TREM2. MV signaling was implicated in this response as reduction of MV secretion by imipramine blocked pro-inflammatory TNF-α and IL-1β induction by ethanol, and ethanol-conditioned MVs (EtOH-MVs) reproduced the ethanol-associated immune gene signature in naïve OBSC slices. Depletion of microglia prior to ethanol treatment prevented pro-inflammatory activity of EtOH-MVs, as did incubation of EtOH-MVs with the HMGB1 inhibitor glycyrrhizin. Ethanol caused HMGB1 secretion from cultured BV2 microglia in MVs through activation of PI3 kinase. In summary, these studies find MVs modulate pro-inflammatory gene induction and microglial activation changes associated with ethanol. Thus, MVs may represent a novel therapeutic target to reduce neuroinflammation in the setting of alcohol abuse or other diseases that feature a neuroimmune component. [Correction added on 5 April 2021, after first online publication: The copyright line was changed.].
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, 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.,Department of Psychiatry, The University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Jian Zou
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, 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
| |
Collapse
|
31
|
Cao Y, Ao T, Wang X, Wei W, Fan J, Tian X. CD300a and CD300f molecules regulate the function of leukocytes. Int Immunopharmacol 2021; 93:107373. [PMID: 33548578 DOI: 10.1016/j.intimp.2021.107373] [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: 11/19/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
The CD300 molecule family is a type I transmembrane glycoprotein expressed on cell membrane of human and other mammals, and of its eight members, only CD300a and CD300f are classified as inhibitory receptors. CD300a and CD300f play an important role in regulating the function of leukocytes, such as activation, proliferation, differentiation, migration and immunity function. They are considered as potential targets for studying the development and progression of inflammation, infection and other diseases. Here, we review the expression and regulatory mechanisms of CD300a and CD300f on leukocytes, as well as their effects on relevant diseases.
Collapse
Affiliation(s)
- Yue Cao
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Tianrang Ao
- Department of Cardiology, Peking Union Medical College Hospital, Tsinghua University, Beijing 100730, China
| | - Xiaohong Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Wumei Wei
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Jun Fan
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaohong Tian
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
32
|
Wen B, Zhang C, Zhou J, Zhang Z, Che Q, Cao H, Bai Y, Guo J, Su Z. Targeted treatment of alcoholic liver disease based on inflammatory signalling pathways. Pharmacol Ther 2020; 222:107752. [PMID: 33253739 DOI: 10.1016/j.pharmthera.2020.107752] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Targeted therapy is an emerging treatment strategy for alcoholic liver disease (ALD). Inflammation plays an important role in the occurrence and development of ALD, and is a key choice for its targeted treatment, and anti-inflammatory treatment has been considered beneficial for liver disease. Surprisingly, immune checkpoint inhibitors have become important therapeutic agents for hepatocellular carcinoma (HCC). Moreover, studies have shown that the combination of inflammatory molecule inhibitors and immune checkpoint inhibitors can exert better effects than either alone in mouse models of HCC. This review discusses the mechanism of hepatic ethanol metabolism and the conditions under which inflammation occurs. In addition, we focus on the potential molecular targets in inflammatory signalling pathways and summarize the potential targeted inhibitors and immune checkpoint inhibitors, providing a theoretical basis for the targeted treatment of ALD and the development of new combination therapy strategies for HCC.
Collapse
Affiliation(s)
- Bingjian Wen
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chengcheng Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingwen Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengyan Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd., Guangzhou 510663, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
33
|
Wang X, Yu H, Wang C, Liu Y, You J, Wang P, Xu G, Shen H, Yao H, Lan X, Zhao R, Wu X, Zhang G. Chronic ethanol exposure induces neuroinflammation in H4 cells through TLR3 / NF-κB pathway and anxiety-like behavior in male C57BL/6 mice. Toxicology 2020; 446:152625. [PMID: 33161052 DOI: 10.1016/j.tox.2020.152625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/22/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
Chronic alcoholism has become a major public health problem. Long-term and excessive drinking can lead to a variety of diseases. Chronic ethanol exposure can induce neuroinflammation and anxiety-like behavior, and this may be induced through the Toll-like receptor 3/nuclear factor-κB (TLR3/NF-κB) pathway. Animal experiments were performed using healthy adult male C57BL/6 N mice given 10 % (m/V) or 20 % ethanol solution as the only choice of drinkable fluid for 60, 90 or 180 d. In cell culture experiments, H4 human glioma cells were treated with 100 mM ethanol for 2 d, with the TLR3 gene silenced by RNAi and NF-κB inhibited by ammonium pyrrolidine dithiocarbamate (PDTC, 10 μM). After treatment with ethanol solution for a specific time, the anxiety-like behavior of the mice was tested using the open field test and the elevated plus maze test. Western blotting was used to detect the expression of TLR3, TLR4, NF-κB, IL-1β, IL-6, and TNF-α in the mouse hippocampus and H4 cells. The expression of IL-1β, IL-6 and TNF-α in the supernatant of cell culture medium was detected by ELISA. The open field test showed a decrease in time spent in the central area, and the elevated plus maze test showed a decrease in activity time in the open arm region. These behavioral tests indicated that ethanol caused anxiety-like behavior in mice. The expression levels of TLR3, TLR4, NF-κB, IL-1β, IL-6, and TNF-α increased after ethanol exposure in both the hippocampus of mice and H4 cells. Silencing of the TLR3 gene by RNAi or inhibition of NF-κB by PDTC attenuated the ethanol-induced increase in the expression of inflammatory factors in H4 cells. These findings indicated that chronic ethanol exposure increases the expression of TLR3 and NF-κB and produces neuroinflammation and anxiety-like behavior in male C57BL/6 mice and that ethanol-induced neuroinflammation can be caused through the TLR3/NF-κB pathway.
Collapse
Affiliation(s)
- Xiaolong Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hao Yu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Changliang Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China; The People's Procuratorate of Liaoning Province Judicial Authentication Center, Shenyang, Liaoning, 110032, PR China; Collaborative Laboratory of Intelligentized Forensic Science (CLIFS), Shenyang, Liaoning, 110032, PR China
| | - Yang Liu
- The People's Procuratorate of Liaoning Province Judicial Authentication Center, Shenyang, Liaoning, 110032, PR China; Collaborative Laboratory of Intelligentized Forensic Science (CLIFS), Shenyang, Liaoning, 110032, PR China
| | - Jiabin You
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Pengfei Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Guohui Xu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hui Shen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hui Yao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xinze Lan
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Rui Zhao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xu Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Guohua Zhang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, 110122, PR China.
| |
Collapse
|
34
|
Airapetov MI, Eresko SO, Lebedev AA, Bychkov ER, Shabanov PD. [Involvement of TOLL-like receptors in the neuroimmunology of alcoholism]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:208-215. [PMID: 32588826 DOI: 10.18097/pbmc20206603208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alcohol use is a global socially significant problem that remains one of the leading risk factors for disability and premature death. One of the main pathological characteristics of alcoholism is the loss of cognitive control over the amount of consumed alcohol. Growing body of evidence suggests that alterations of neuroimmune communication occurring in the brain during prolonged alcoholization are one of the main mechanisms responsible for the development of this pathology. Ethanol consumption leads to activation of neuroimmune signaling in the central nervous system through many types of Toll-like receptors (TLRs), as well as the release of their endogenous agonists (HMGB1 protein, S100 protein, heat shock proteins, extracellular matrix breakdown proteins). Activation of TLRs triggers intracellular molecular cascades leading to increased expression of the innate immune system genes, particularly proinflammatory cytokines, subsequently causing the development of a persistent neuroinflammatory process in the central nervous system, which results in massive death of neurons and glial cells in the brain structures, which are primarily associated with the development of a pathological craving for alcohol. In addition, some subtypes of TLRs are capable of forming heterodimers with neuropeptide receptors (corticoliberin, orexin, ghrelin receptors), and may also have other functional relationships.
Collapse
Affiliation(s)
- M I Airapetov
- Institute of Experimental Medicine, St. Petersburg, Russia; St. Petersburg State Medical Pediatric University, St. Petersburg, Russia
| | - S O Eresko
- University ITMO (National Research University), St. Petersburg, Russia
| | - A A Lebedev
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - E R Bychkov
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - P D Shabanov
- Institute of Experimental Medicine, St. Petersburg, Russia; Kirov Military Medical Academy, St. Petersburg, Russia
| |
Collapse
|
35
|
Grantham EK, Warden AS, McCarthy GS, DaCosta A, Mason S, Blednov Y, Mayfield RD, Harris RA. Role of toll-like receptor 7 (TLR7) in voluntary alcohol consumption. Brain Behav Immun 2020; 89:423-432. [PMID: 32726684 PMCID: PMC7572874 DOI: 10.1016/j.bbi.2020.07.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/09/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
Overactivation of neuroimmune signaling has been linked to excessive ethanol consumption. Toll-like receptors (TLRs) are a major component of innate immune signaling and initiate anti- and pro-inflammatory responses via intracellular signal transduction cascades. TLR7 is upregulated in post-mortem brain tissue from humans with alcohol use disorder (AUD) and animals with prior exposure to ethanol. Despite this evidence, the role of TLR7 in the regulation of voluntary ethanol consumption has not been studied. We test the hypothesis that TLR7 activation regulates voluntary ethanol drinking behavior by administering a TLR7 agonist (R848) during an intermittent access drinking procedure in mice. Acute activation of TLR7 reduced ethanol intake, preference, and total fluid intake due, at least in part, to an acute sickness response. However, chronic pre-treatment with R848 resulted in tolerance to the adverse effects of the drug and a subsequent increase in ethanol consumption. To determine the molecular machinery that mediates these behavioral changes, we evaluated gene expression after acute and chronic TLR7 activation. We found that acute TLR7 activation produces brain region specific changes in expression of immune pathway genes, whereas chronic TLR7 activation causes downregulation of TLRs and blunted cytokine induction, suggesting molecular tolerance. Our results demonstrate a novel role for TLR7 signaling in regulating voluntary ethanol consumption. Taken together, our findings suggest TLR7 may be a viable target for development of therapies to treat AUD.
Collapse
Affiliation(s)
- E K Grantham
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| | - A S Warden
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - G S McCarthy
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - A DaCosta
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA
| | - S Mason
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA
| | - Y Blednov
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA
| | - R D Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
36
|
Warden AS, DaCosta A, Mason S, Blednov YA, Mayfield RD, Harris RA. Inbred Substrain Differences Influence Neuroimmune Response and Drinking Behavior. Alcohol Clin Exp Res 2020; 44:1760-1768. [PMID: 32640038 DOI: 10.1111/acer.14410] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/03/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The inbred mouse strain C57BL/6 is widely used in both models of addiction and immunological disease. However, there are pronounced phenotypic differences in ethanol (EtOH) consumption and innate immune response between C57BL/6 substrains. The focus of this study was to examine the effects of substrain on innate immune response and neuroimmune-induced escalation of voluntary EtOH consumption. The main goal was to identify whether substrain differences in immune response can account for differences in EtOH behavior. METHODS We compared acute innate immune response with a viral dsRNA mimic, polyinosinic:polycytidylic acid (poly(I:C)), in brain using qRT-PCR in both C57BL/6N and C57BL/6J mice. Next, we used a neuroimmune model of escalation using poly(I:C) to compare drinking behavior between substrains. Finally, we compared brain neuroimmune response with both EtOH and repeated poly(I:C) in both substrains as a way to account for differences in EtOH behavior. RESULTS We found that C57BL/6 substrains have differing immune response and drinking behaviors. C57BL/6N mice have a shorter but more robust inflammatory response to acute poly(I:C). In contrast, C57BL/6J mice have a smaller but longer-lasting acute immune response to poly(I:C). In our neuroimmune-induced escalation model, C57BL/6J mice but not C57BL/6N mice escalate EtOH intake after poly(I:C). Finally, only C57BL/6J mice show enhanced proinflammatory transcript abundance after poly(I:C) and EtOH, suggesting that longer-lasting immune responses are critical to neuroimmune drinking phenotypes. CONCLUSIONS Altogether, this work has elucidated additional influences that substrain has on both innate immune response and drinking phenotypes. Our observations highlight the importance of considering and reporting the source and background used for production of transgenic and knockout mice. These data provide further evidence that genetic background must be carefully considered when investigating the role of neuroimmune signaling in EtOH abuse.
Collapse
Affiliation(s)
- Anna S Warden
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA.,Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA.,Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA
| | - Adriana DaCosta
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA
| | - Sonia Mason
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA
| | - Yuri A Blednov
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA
| | - Roy Dayne Mayfield
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA.,Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Robert Adron Harris
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA.,Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
37
|
Deep proteome profiling reveals novel pathways associated with pro-inflammatory and alcohol-induced microglial activation phenotypes. J Proteomics 2020; 220:103753. [PMID: 32200115 DOI: 10.1016/j.jprot.2020.103753] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/09/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
Microglia, the resident immune cells of the brain, can exhibit a broad range of activation phenotypes, many of which have been implicated in several diseases and disorders of the central nervous system including those related to alcohol abuse. Given the complexity of global-scale molecular changes that define microglial activation, accurate phenotypic classification in the context of alcohol exposure is still lacking. We employed an optimized method for deep, quantitative proteome profiling of primary microglia in order to characterize their response to acute exposure to alcohol (ethanol) as well as the pro-inflammatory driver and TLR4 agonist, LPS. From this analysis, 5,062 total proteins were identified where 4,857 and 4,928 of those proteins were quantifiable by label-free quantitation in ethanol and LPS treatment groups, respectively. This study highlights the subtle, yet significant proteomic changes that occur in ethanol-treated microglia, which do not align with the robust pro-inflammatory phenotype induced by TLR4 activation. Specifically, our results indicate inhibition of several upstream regulators associated with inflammation, opposing effects on pathways such as phagocytosis upon comparison to TLR4-mediated pro-inflammatory phenotype, and a potential metabolic shift associated with increased expression of proteins related to OXPHOS and lipid homeostasis. Data are available via ProteomeXchange with identifier PXD14466. SIGNIFICANCE: Alcohol abuse has a significant impact on the central nervous system, which includes the pathophysiological mechanisms resulting from glial cell activation. Microglia, in particular, are the resident immune cells of the brain and exhibit a broad range of activation phenotypes. The molecular changes that drive microglial activation phenotype are complex and have yet to be fully characterized in the context of alcohol exposure. Our study highlights the first and most comprehensive characterization of alcohol-induced proteomic changes in primary microglia to date and has shed light on novel immune-related and metabolic pathways that are altered due to alcohol exposure. The results from this study provide an important foundation for future work aimed to understand the complexity of alcohol-induced microglial activation in vivo and other translational models of acute and chronic alcohol exposure.
Collapse
|
38
|
Petralia MC, Mazzon E, Mangano K, Fagone P, Di Marco R, Falzone L, Basile MS, Nicoletti F, Cavalli E. Transcriptomic analysis reveals moderate modulation of macrophage migration inhibitory factor superfamily genes in alcohol use disorders. Exp Ther Med 2020; 19:1755-1762. [PMID: 32104230 PMCID: PMC7026954 DOI: 10.3892/etm.2020.8410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022] Open
Abstract
Alcohol use disorder (AUD) is a primary, chronic and relapsing disease of brain reward, motivation and memory, which is associated with several comorbidities, including major depression and post-traumatic stress disorder. It has been revealed that Ibudilast (IBUD), a dual inhibitor of phosphodiesterase-4 and −10 and of macrophage migration inhibitory factor (MIF), exerts beneficial effects on AUD in rodent models and human patients. Therefore, IBUD has attracted increasing interest, with research focusing on the elucidation of the pathogenic role of MIF and its homologue, D-dopachrome tautomerase (DDT), in the pathogenesis and maintenance of AUD. By using DNA microarray analysis, the current study performed a transcriptomic expression analysis of MIF, DDT and their co-receptors, including CD74, C-X-C chemokine receptor (CXCR)2, CXCR4 and CXCR7 in patients with AUD. The results revealed that the transcriptomic levels of MIF, DDT and their receptors were superimposable in the prefrontal cortex of rodents and patients with AUD and human patients. Furthermore, peripheral blood cells from heavy drinkers exhibited a moderate increase in MIF and DDT levels, both at the baseline and following exposure to alcohol-associated cues, based on individual situations that included alcohol-related stimuli resulting in subsequent alcohol use (buying alcohol and being at a bar, watching others drink alcohol). Considering the overlapping effects of MIF and DDT, the inverse Fisher's χ2 test was performed on unadjusted P-values to evaluate the combined effect of MIF and DDT. The results revealed a significant increase in these cytokines in heavy drinkers compared with controls (moderate drinkers). To the best of our knowledge, the present study demonstrated for the first time that MIF and DDT expression was upregulated in the blood of patients with AUD. These results therefore warrant further study to evaluate the role of MIF and DDT in the development and maintenance of AUD, to evaluate their use as biomarkers to predict the psychotherapeutic and pharmacological response of patients with AUD and for use as therapeutic targets.
Collapse
Affiliation(s)
- Maria Cristina Petralia
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Emanuela Mazzon
- IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Centro Neurolesi 'Bonino-Pulejo', I-98124 Messina, Italy
| | - Katia Mangano
- IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Centro Neurolesi 'Bonino-Pulejo', I-98124 Messina, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Roberto Di Marco
- Department of Medicine and Health Sciences 'Vincenzo Tiberio', University of Molise, I-86100 Campobasso, Italy
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Eugenio Cavalli
- IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Centro Neurolesi 'Bonino-Pulejo', I-98124 Messina, Italy
| |
Collapse
|
39
|
Ferguson LB, Patil S, Moskowitz BA, Ponomarev I, Harris RA, Mayfield RD, Messing RO. A Pathway-Based Genomic Approach to Identify Medications: Application to Alcohol Use Disorder. Brain Sci 2019; 9:E381. [PMID: 31888299 PMCID: PMC6956180 DOI: 10.3390/brainsci9120381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic, excessive alcohol use alters brain gene expression patterns, which could be important for initiating, maintaining, or progressing the addicted state. It has been proposed that pharmaceuticals with opposing effects on gene expression could treat alcohol use disorder (AUD). Computational strategies comparing gene expression signatures of disease to those of pharmaceuticals show promise for nominating novel treatments. We reasoned that it may be sufficient for a treatment to target the biological pathway rather than lists of individual genes perturbed by AUD. We analyzed published and unpublished transcriptomic data using gene set enrichment of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways to identify biological pathways disrupted in AUD brain and by compounds in the Library of Network-based Cellular Signatures (LINCS L1000) and Connectivity Map (CMap) databases. Several pathways were consistently disrupted in AUD brain, including an up-regulation of genes within the Complement and Coagulation Cascade, Focal Adhesion, Systemic Lupus Erythematosus, and MAPK signaling, and a down-regulation of genes within the Oxidative Phosphorylation pathway, strengthening evidence for their importance in AUD. Over 200 compounds targeted genes within those pathways in an opposing manner, more than twenty of which have already been shown to affect alcohol consumption, providing confidence in our approach. We created a user-friendly web-interface that researchers can use to identify drugs that target pathways of interest or nominate mechanism of action for drugs. This study demonstrates a unique systems pharmacology approach that can nominate pharmaceuticals that target pathways disrupted in disease states such as AUD and identify compounds that could be repurposed for AUD if sufficient evidence is attained in preclinical studies.
Collapse
Affiliation(s)
- Laura B. Ferguson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Shruti Patil
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Bailey A. Moskowitz
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Robert A. Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Roy D. Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O. Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
40
|
Bajo M, Patel RR, Hedges DM, Varodayan FP, Vlkolinsky R, Davis TD, Burkart MD, Blednov YA, Roberto M. Role of MyD88 in IL-1β and Ethanol Modulation of GABAergic Transmission in the Central Amygdala. Brain Sci 2019; 9:brainsci9120361. [PMID: 31817854 PMCID: PMC6956324 DOI: 10.3390/brainsci9120361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
Myeloid differentiation primary response protein (MyD88) is a critical neuroimmune adaptor protein in TLR (Toll-like receptor) and IL-1R (Interleukin-1 receptor) signaling complexes. These two pro-inflammatory families play an important role in the neurobiology of alcohol use disorder, specifically MyD88 regulates ethanol drinking, ethanol-induced sedation, and ethanol-induced deficits in motor coordination. In this study, we examined the role of MyD88 in mediating the effects of IL-1β and ethanol on GABAergic transmission in the central amygdala (CeA) of male mice using whole-cell patch-clamp recordings in combination with pharmacological (AS-1, a mimetic that prevents MyD88 recruitment by IL-1R) and genetic (Myd88 knockout mice) approaches. We demonstrate through both approaches that IL-1β and ethanol’s modulatory effects at CeA GABA synapses are not dependent on MyD88. Myd88 knockout potentiated IL-1β’s actions in reducing postsynaptic GABAA receptor function. Pharmacological inhibition of MyD88 modulates IL-1β’s action at CeA GABA synapses similar to Myd88 knockout mice. Additionally, ethanol-induced CeA GABA release was greater in Myd88 knockout mice compared to wildtype controls. Thus, MyD88 is not essential to IL-1β or ethanol regulation of CeA GABA synapses but plays a role in modulating the magnitude of their effects, which may be a potential mechanism by which it regulates ethanol-related behaviors.
Collapse
Affiliation(s)
- Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
- Correspondence: ; Tel.: +1-858-784-7259
| | - Reesha R. Patel
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - David M. Hedges
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - Florence P. Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - Tony D. Davis
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA; (T.D.D.); (M.D.B.)
| | - Michael D. Burkart
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA; (T.D.D.); (M.D.B.)
| | - Yuri A. Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| |
Collapse
|
41
|
Nunes PT, Kipp BT, Reitz NL, Savage LM. Aging with alcohol-related brain damage: Critical brain circuits associated with cognitive dysfunction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:101-168. [PMID: 31733663 PMCID: PMC7372724 DOI: 10.1016/bs.irn.2019.09.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcoholism is associated with brain damage and impaired cognitive functioning. The relative contributions of different etiological factors, such as alcohol, thiamine deficiency and age vulnerability, to the development of alcohol-related neuropathology and cognitive impairment are still poorly understood. One reason for this quandary is that both alcohol toxicity and thiamine deficiency produce brain damage and cognitive problems that can be modulated by age at exposure, aging following alcohol toxicity or thiamine deficiency, and aging during chronic alcohol exposure. Pre-clinical models of alcohol-related brain damage (ARBD) have elucidated some of the contributions of ethanol toxicity and thiamine deficiency to neuroinflammation, neuronal loss and functional deficits. However, the critical variable of age at the time of exposure or long-term aging with ARBD has been relatively ignored. Acute thiamine deficiency created a massive increase in neuroimmune genes and proteins within the thalamus and significant increases within the hippocampus and frontal cortex. Chronic ethanol treatment throughout adulthood produced very minor fluctuations in neuroimmune genes, regardless of brain region. Intermittent "binge-type" ethanol during the adolescent period established an intermediate neuroinflammatory response in the hippocampus and frontal cortex, that can persist into adulthood. Chronic excessive drinking throughout adulthood, adolescent intermittent ethanol exposure, and thiamine deficiency all led to a loss of the cholinergic neuronal phenotype within the basal forebrain, reduced hippocampal neurogenesis, and alterations in the frontal cortex. Only thiamine deficiency results in gross pathological lesions of the thalamus. The behavioral impairment following these types of treatments is hierarchical: Thiamine deficiency produces the greatest impairment of hippocampal- and prefrontal-dependent behaviors, chronic ethanol drinking ensues mild impairments on both types of tasks and adolescent intermittent ethanol exposure leads to impairments on frontocortical tasks, with sparing on most hippocampal-dependent tasks. However, our preliminary data suggest that as rodents age following adolescent intermittent ethanol exposure, hippocampal functional deficits began to emerge. A necessary requirement for the advancement of understanding the neural consequences of alcoholism is a more comprehensive assessment and understanding of how excessive alcohol drinking at different development periods (adolescence, early adulthood, middle-aged and aged) influences the trajectory of the aging process, including pathological aging and disease.
Collapse
Affiliation(s)
- Polliana Toledo Nunes
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Brian T Kipp
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Nicole L Reitz
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Lisa M Savage
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States.
| |
Collapse
|
42
|
Loftis JM, Taylor J, Hudson R, Firsick EJ. Neuroinvasion and cognitive impairment in comorbid alcohol dependence and chronic viral infection: An initial investigation. J Neuroimmunol 2019; 335:577006. [PMID: 31325774 DOI: 10.1016/j.jneuroim.2019.577006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022]
Abstract
Viruses that invade the central nervous system (CNS) can cause neuropsychiatric impairments. Similarly, chronic alcohol exposure can induce inflammatory responses that alter brain function. However, the effects of a chronic viral infection and comorbid alcohol use on neuroinflammation and behavior are not well-defined. We investigated the role of heavy alcohol intake in regulating inflammatory responses and behavioral signs of cognitive impairments in mice infected with lymphocytic choriomeningitis virus (LCMV) clone 13. LCMV-infected mice exposed to alcohol had increased peripheral inflammation and impaired cognitive function (as indicated by performance on the novel object recognition test). Initial findings suggest that brain region-specific dysregulation of microglial response to viral infection may contribute to cognitive impairments in the context of heavy alcohol use.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA; Methamphetamine Abuse Research Center, Veterans Affairs Portland Health Care System, Oregon Health & Science University, Portland, OR, USA.
| | - Jonathan Taylor
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - Rebekah Hudson
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Evan J Firsick
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA
| |
Collapse
|
43
|
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: 2.6] [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
|
44
|
Ethanol Induction of Innate Immune Signals Across BV2 Microglia and SH-SY5Y Neuroblastoma Involves Induction of IL-4 and IL-13. Brain Sci 2019; 9:brainsci9090228. [PMID: 31510019 PMCID: PMC6770440 DOI: 10.3390/brainsci9090228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/03/2019] [Accepted: 09/08/2019] [Indexed: 02/07/2023] Open
Abstract
Innate immune signaling molecules, such as Toll-like receptors (TLRs), cytokines and transcription factor NFκB, are increased in post-mortem human alcoholic brain and may play roles in alcohol dependence and neurodegeneration. Innate immune signaling involves microglia -neuronal signaling which while poorly understood, may impact learning and memory. To investigate mechanisms of ethanol induction of innate immune signaling within and between brain cells, we studied immortalized BV2 microglia and SH-SY5Y human neuroblastoma to model microglial and neuronal signaling. Cells were treated alone or in co-culture using a Transwell system, which allows transfer of soluble mediators. We determined immune signaling mRNA using real-time polymerase chain reaction. Ethanol induced innate immune genes in both BV2 and SH-SY5Y cultured alone, with co-culture altering gene expression at baseline and following ethanol exposure. Co-culture blunted ethanol-induced high mobility group box protein 1 (HMGB1)-TLR responses, corresponding with reduced ethanol induction of several proinflammatory NFκB target genes. In contrast, co-culture resulted in ethanol upregulation of cytokines IL-4 and IL-13 in BV2 and corresponding receptors, that is, IL-4 and IL-13 receptors, in SH-SY5Y, suggesting induction of a novel signaling pathway. Co-culture reduction in HMGB1-TLR levels occurs in parallel with reduced proinflammatory gene induction and increased IL-4 and IL-13 ligands and receptors. Findings from these immortalized and tumor-derived cell lines could provide insight into microglial-neuronal interactions via release of soluble mediators in vivo.
Collapse
|
45
|
Melbourne JK, Thompson KR, Peng H, Nixon K. Its complicated: The relationship between alcohol and microglia in the search for novel pharmacotherapeutic targets for alcohol use disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 167:179-221. [PMID: 31601404 DOI: 10.1016/bs.pmbts.2019.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder (AUD) is a chronic relapsing disorder with wide-ranging health consequences. Alcohol targets the central nervous system producing neurodegeneration and subsequent cognitive and behavioral deficits, but the mechanisms behind these effects remain unclear. Recently, evidence has been mounting for the role of neuroimmune activation in the pathogenesis of AUDs, but our nascent state of knowledge about the interaction of alcohol with the neuroimmune system supports that the relationship is complicated. As the resident macrophage of the central nervous system, microglia are a central focus. Human and animal research on the interplay between microglia and alcohol in AUDs has proven to be complex, and though early research focused on a pro-inflammatory phenotype of microglia, the anti-inflammatory and homeostatic roles of microglia must be considered. How these new roles for microglia should be incorporated into our thinking about the neuroimmune system in AUDs is discussed in the context of developing novel pharmacotherapies for AUDs.
Collapse
Affiliation(s)
- Jennifer K Melbourne
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States
| | - K Ryan Thompson
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States
| | - Hui Peng
- University of Kentucky, College of Pharmacy, Department of Pharmaceutical Sciences, Lexington, KY, United States
| | - Kimberly Nixon
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States.
| |
Collapse
|
46
|
Erickson EK, Blednov YA, Harris RA, Mayfield RD. Glial gene networks associated with alcohol dependence. Sci Rep 2019; 9:10949. [PMID: 31358844 PMCID: PMC6662804 DOI: 10.1038/s41598-019-47454-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic alcohol abuse alters the molecular structure and function of brain cells. Recent work suggests adaptations made by glial cells, such as astrocytes and microglia, regulate physiological and behavioral changes associated with addiction. Defining how alcohol dependence alters the transcriptome of different cell types is critical for developing the mechanistic hypotheses necessary for a nuanced understanding of cellular signaling in the alcohol-dependent brain. We performed RNA-sequencing on total homogenate and glial cell populations isolated from mouse prefrontal cortex (PFC) following chronic intermittent ethanol vapor exposure (CIE). Compared with total homogenate, we observed unique and robust gene expression changes in astrocytes and microglia in response to CIE. Gene co-expression network analysis revealed biological pathways and hub genes associated with CIE in astrocytes and microglia that may regulate alcohol-dependent phenotypes. Astrocyte identity and synaptic calcium signaling genes were enriched in alcohol-associated astrocyte networks, while TGF-β signaling and inflammatory response genes were disrupted by CIE treatment in microglia gene networks. Genes related to innate immune signaling, specifically interferon pathways, were consistently up-regulated across CIE-exposed astrocytes, microglia, and total homogenate PFC tissue. This study illuminates the cell-specific effects of chronic alcohol exposure and provides novel molecular targets for studying alcohol dependence.
Collapse
Affiliation(s)
- Emma K Erickson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA.
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712-01095, USA
| |
Collapse
|
47
|
Abstract
Innate immune signaling is an important feature in the pathology of alcohol use disorders. Alcohol abuse causes persistent innate immune activation in the brain. This is seen in postmortem human alcoholic brain specimens, as well as in primate and rodent models of alcohol consumption. Further, in vitro models of alcohol exposure in neurons and glia also demonstrate innate immune activation. The activation of the innate immune system seems to be important in the development of alcohol use pathology, as anti-immune therapies reduce pathology and ethanol self-administration in rodent models. Further, innate immune activation has been identified in each of the stages of addiction: binge/intoxication, withdrawal/negative affect, and preoccupation/craving. This suggests that innate immune activation may play a role both in the development and maintenance of alcoholic pathology. In this chapter, we discuss the known contributions of innate immune signaling in the pathology of alcohol use disorders, and present potential therapeutic interventions that may be beneficial for alcohol use disorders.
Collapse
Affiliation(s)
- Leon G Coleman
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
48
|
Warden AS, Azzam M, DaCosta A, Mason S, Blednov YA, Messing RO, Mayfield RD, Harris RA. Toll-like receptor 3 dynamics in female C57BL/6J mice: Regulation of alcohol intake. Brain Behav Immun 2019; 77:66-76. [PMID: 30550930 PMCID: PMC6399033 DOI: 10.1016/j.bbi.2018.12.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 11/19/2018] [Accepted: 12/10/2018] [Indexed: 11/30/2022] Open
Abstract
Although there are sex differences in the effects of alcohol on immune responses, it is unclear if sex differences in immune response can influence drinking behavior. Activation of toll-like receptor 3 (TLR3) by polyinosinic:polycytidylic acid (poly(I:C)) produced a rapid proinflammatory response in males that increased alcohol intake over time (Warden et al., 2019). Poly(I:C) produced a delayed and prolonged innate immune response in females. We hypothesized that the timecourse of innate immune activation could regulate drinking behavior in females. Therefore, we chose to test the effect of two time points in the innate immune activation timecourse on every-other-day two-bottle-choice drinking: (1) peak activation; (2) descending limb of activation. Poly(I:C) reduced ethanol consumption when alcohol access occurred during peak activation. Poly(I:C) did not change ethanol consumption when alcohol access occurred on the descending limb of activation. Decreased levels of MyD88-dependent pathway correlated with decreased alcohol intake and increased levels of TRIF-dependent pathway correlated with increased alcohol intake in females. To validate the effects of poly(I:C) were mediated through MyD88, we tested female mice lacking Myd88. Poly(I:C) did not change alcohol intake in Myd88 knockouts, indicating that poly(I:C)-induced changes in alcohol intake are dependent on MyD88 in females. We next determined if the innate immune timecourse also regulated drinking behavior in males. Poly(I:C) reduced ethanol consumption in males when alcohol was presented at peak activation. Therefore, the timecourse of innate immune activation regulates drinking behavior and sex-specific dynamics of innate immune response must be considered when designing therapeutics to treat excessive drinking.
Collapse
Affiliation(s)
- Anna S Warden
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| | - Moatasem Azzam
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Adriana DaCosta
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Sonia Mason
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Yuri A Blednov
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O Messing
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - R Adron Harris
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
49
|
Warden AS, Azzam M, DaCosta A, Mason S, Blednov YA, Messing RO, Mayfield RD, Harris RA. Toll-like receptor 3 activation increases voluntary alcohol intake in C57BL/6J male mice. Brain Behav Immun 2019; 77:55-65. [PMID: 30550931 PMCID: PMC6399060 DOI: 10.1016/j.bbi.2018.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 11/21/2018] [Accepted: 12/10/2018] [Indexed: 12/30/2022] Open
Abstract
Many genes differentially expressed in brain tissue from human alcoholics and animals that have consumed large amounts of alcohol are components of the innate immune toll-like receptor (TLR) pathway. TLRs initiate inflammatory responses via two branches: (1) MyD88-dependent or (2) TRIF-dependent. All TLRs signal through MyD88 except TLR3. Prior work demonstrated a direct role for MyD88-dependent signaling in regulation of alcohol consumption. However, the role of TLR3 as a potential regulator of excessive alcohol drinking has not previously been investigated. To test the possibility TLR3 activation regulates alcohol consumption, we injected mice with the TLR3 agonist polyinosinic:polycytidylic acid (poly(I:C)) and tested alcohol consumption in an every-other-day two-bottle choice test. Poly(I:C) produced a persistent increase in alcohol intake that developed over several days. Repeated poly(I:C) and ethanol exposure altered innate immune transcript abundance; increased levels of TRIF-dependent pathway components correlated with increased alcohol consumption. Administration of poly(I:C) before exposure to alcohol did not alter alcohol intake, suggesting that poly(I:C) and ethanol must be present together to change drinking behavior. To determine which branch of TLR signaling mediates poly(I:C)-induced changes in drinking behavior, we tested either mice lacking MyD88 or mice administered a TLR3/dsRNA complex inhibitor. MyD88 null mutants showed poly(I:C)-induced increases in alcohol intake. In contrast, mice pretreated with a TLR3/dsRNA complex inhibitor reduced their alcohol intake, suggesting poly(I:C)-induced escalations in alcohol intake are, at least partially, dependent on TLR3. Together, these results strongly suggest that TLR3-dependent signaling drives excessive alcohol drinking behavior.
Collapse
Affiliation(s)
- Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| | - Moatasem Azzam
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Adriana DaCosta
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Sonia Mason
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O Messing
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
50
|
Gene and cell therapy on the acquisition and relapse-like binge drinking in a model of alcoholism: translational options. Gene Ther 2019; 26:407-417. [DOI: 10.1038/s41434-019-0064-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 12/11/2022]
|