1
|
Mashayekhi-Sardoo H, Razazpour F, Hakemi Z, Hedayati-Moghadam M, Baghcheghi Y. Ethanol-Induced Depression: Exploring the Underlying Molecular Mechanisms. Cell Mol Neurobiol 2025; 45:49. [PMID: 40405002 PMCID: PMC12098258 DOI: 10.1007/s10571-025-01569-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/12/2025] [Indexed: 05/24/2025]
Abstract
Ethanol consumption is widely recognized for its detrimental effects on mental health, particularly its association with depressive disorders. This narrative review aims to explore the intricate molecular mechanisms underlying ethanol-induced depression, synthesizing findings from preclinical and clinical studies. We begin by providing an overview of the relationship between chronic ethanol consumption and depression, highlighting compelling evidence from diverse populations. Subsequently, we delve into insights from animal models that elucidate the pathophysiological changes triggered by prolonged ethanol exposure. Key mechanisms identified include oxidative stress, which contributes to cellular damage; neuroinflammation, characterized by the activation of glial cells and altered cytokine profiles; and disruptions in neurotrophic factors that impair neuronal growth and survival. Furthermore, we discuss the induction of apoptosis in neural cells and the significant impact of ethanol on neurotransmitter receptor remodeling and regulation, leading to altered synaptic transmission. While substantial progress has been made in understanding these mechanisms, we also acknowledge the limitations of current research methodologies and call for further investigations to translate these findings into effective therapeutic strategies for individuals affected by ethanol-induced depression. This review ultimately underscores the need for a comprehensive understanding of the molecular underpinnings of ethanol's impact on mood disorders, paving the way for improved interventions and preventative measures.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
- School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fateme Razazpour
- Oral and Dental Diseases Research Center, Kerman University of Medical Science, Kerman, Iran
| | - Zohreh Hakemi
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahdiyeh Hedayati-Moghadam
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran.
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
2
|
Li X, Ni Z, Shi W, Zhao K, Zhang Y, Liu L, Wang Z, Chen J, Yu Z, Gao X, Qin Y, Zhao J, Peng W, Shi J, Kosten TR, Lu L, Su L, Xue Y, Sun H. Nitrate ameliorates alcohol-induced cognitive impairment via oral microbiota. J Neuroinflammation 2025; 22:106. [PMID: 40234914 PMCID: PMC12001487 DOI: 10.1186/s12974-025-03439-x] [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: 01/29/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025] Open
Abstract
Alcohol use is associated with cognitive impairment and dysregulated inflammation. Oral nitrate may benefit cognitive impairment in aging through altering the oral microbiota. Similarly, the beneficial effects of nitrate on alcohol-induced cognitive decline and the roles of the oral microbiota merit investigation. Here we found that nitrate supplementation effectively mitigated cognitive impairment induced by chronic alcohol exposure in mice, reducing both systemic and neuroinflammation. Furthermore, nitrate restored the dysbiosis of the oral microbiota caused by alcohol consumption. Notably, removing the oral microbiota led to a subsequent loss of the beneficial effects of nitrate. Oral microbiota from donor alcohol use disordered humans who had been taking the nitrate intervention were transplanted into germ-free mice which then showed increased cognitive function and reduced neuroinflammation. Finally, we examined 63 alcohol drinkers with varying levels of cognitive impairment and found that lower concentrations of nitrate metabolism-related bacteria were associated with higher cognitive impairment and lower nitrate levels in plasma. These findings highlight the protective role of nitrate against alcohol-induced cognition impairment and neuroinflammation and suggest that the oral microbiota associated with nitrate metabolism and brain function may form part of a "microbiota-mouth-brain axis".
Collapse
Affiliation(s)
- Xiangxue Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Zhaojun Ni
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Weixiong Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China
| | - Kangqing Zhao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Yanjie Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Lina Liu
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhong Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Jie Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Zhoulong Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Xuejiao Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Ying Qin
- Addiction Medicine Department, The Second People's Hospital of Guizhou Province, Guizhou, China
| | - Jingwen Zhao
- Addiction Medicine Department, The Second People's Hospital of Guizhou Province, Guizhou, China
| | - Wenjuan Peng
- Addiction Medicine Department, The Second People's Hospital of Guizhou Province, Guizhou, China
| | - Jie Shi
- National Institute On Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Thomas R Kosten
- Department of Psychiatry, Pharmacology, Neuroscience, Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
- National Institute On Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Lei Su
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - Yanxue Xue
- National Institute On Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China.
- Chinese Institute for Brain Research, Beijing, China.
| | - Hongqiang Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
3
|
Hossaini D, Alipour AK, Sajjadi M, Ansari M, Haidary M. Biotin Mitigates Alcohol Withdrawal-Induced Anxiety and Depression by Regulating Serotonin Metabolism, BDNF, Inflammation, and Oxidative Stress in Rats. Neuropsychopharmacol Rep 2025; 45:e12523. [PMID: 39754400 DOI: 10.1002/npr2.12523] [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: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/21/2024] [Indexed: 01/06/2025] Open
Abstract
INTRODUCTION Substance use disorders, particularly alcohol use disorders, represent a significant public health problem, with adolescents particularly vulnerable to their adverse effects. This study examined the possible anxiolytic and antidepressant effects of biotin, a crucial vitamin for brain function, in attenuating the behavioral and neurobiological changes associated with alcohol withdrawal in adolescent rats. MATERIALS AND METHODS Sixty male Sprague-Dawley rats were exposed to a 20% ethanol solution for 21 days, followed by a 21-day drug-free period to assess long-term behavioral and physiological changes. Behavioral assessments included the Open Field Test, Elevated Plus Maze, and Forced Swimming Test, administered post-withdrawal to evaluate anxiety and depression behaviors. Additionally, biochemical analyses were performed to measure serotonin levels, monoamine oxidase-A (MAO-A) activity, and BDNF concentrations. RESULTS The results indicate that ethanol withdrawal significantly induced anxiety- and depression-like behavior in the rats. However, treatment with biotin, particularly at higher doses, effectively attenuated these withdrawal-related behavioral changes. Mechanistically, biotin administration was found to regulate serotonin levels, monoamine oxidase activity, brain-derived neurotrophic factor, and glial fibrillary acidic protein, and alleviate oxidative stress markers in cortical tissue. DISCUSSION The results of this study suggest that biotin may have therapeutic potential for alleviating the negative effects of alcohol withdrawal, particularly those related to anxiety and depression. Further research is needed to elucidate the underlying mechanisms and examine the clinical effects of biotin supplementation for individuals undergoing alcohol withdrawal.
Collapse
Affiliation(s)
- Dawood Hossaini
- Department of Biology and Microbiology, Faculty of Medical Laboratory Technology, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Adam Khan Alipour
- Department of Chemistry and Biochemistry, Faculty of Medical Laboratory Technology, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Meysam Sajjadi
- Department of Chemistry and Biochemistry, Faculty of Medical Laboratory Technology, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Mustafa Ansari
- Department of Biology and Microbiology, Faculty of Medical Laboratory Technology, Khatam Al-Nabieen University, Kabul, Afghanistan
- Medical Research and Technology Center, Khatam Al-Nabieen University, Kabul, Afghanistan
| | - Murtaza Haidary
- Department of Biology and Microbiology, Faculty of Medical Laboratory Technology, Khatam Al-Nabieen University, Kabul, Afghanistan
- Medical Research and Technology Center, Khatam Al-Nabieen University, Kabul, Afghanistan
| |
Collapse
|
4
|
Domínguez-Pino M, Mellado S, Cuesta CM, Grillo-Risco R, García-García F, Pascual M. Metagenomics Reveals Sex-Based Differences in Murine Fecal Microbiota Profiles Induced by Chronic Alcohol Consumption. Int J Mol Sci 2024; 25:12534. [PMID: 39684246 DOI: 10.3390/ijms252312534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic ethanol exposure induces an inflammatory response within the intestinal tract, compromising mucosal and epithelial integrity and leading to dysbiosis of the gut microbiome. However, the specific roles of the gut microbiota in mediating ethanol-induced effects, as well as their interactions with the immune system, remain poorly characterized. This study aimed to evaluate sex-based differences in fecal microbiota profiles induced by chronic alcohol consumption and to assess whether TLR4 is involved in these effects. We analyzed the 16S rRNA gene sequencing of fecal samples from male and female wild-type (WT) and TLR4-knockout (TLR4-KO) mice with and without chronic ethanol exposure over a three-month period. Our findings provide evidence, for the first time, that male mice are more susceptible to the effects of ethanol on the fecal microbiota, since ethanol exposure induced greater alterations in the Gram-negative and -positive bacteria with immunogenic capacity in the WT male mice than in the female mice. We also demonstrate that the absence of immune receptor TLR4 leads to different microbiota in both sexes, showing anti-inflammatory and protective properties for intestinal barrier function and resulting in a phenotype more resistant to ethanol's effects. These findings may open new avenues for understanding the relationship between gut microbiota profiles and inflammation in the digestive system induced by chronic alcohol consumption.
Collapse
Affiliation(s)
- Manuel Domínguez-Pino
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Carlos M Cuesta
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Rubén Grillo-Risco
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Francisco García-García
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| |
Collapse
|
5
|
Koss KM, Son T, Li C, Hao Y, Cao J, Churchward MA, Zhang ZJ, Wertheim JA, Derda R, Todd KG. Toward discovering a novel family of peptides targeting neuroinflammatory states of brain microglia and astrocytes. J Neurochem 2024; 168:3386-3414. [PMID: 37171455 PMCID: PMC10640667 DOI: 10.1111/jnc.15840] [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: 12/16/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
Microglia are immune-derived cells critical to the development and healthy function of the brain and spinal cord, yet are implicated in the active pathology of many neuropsychiatric disorders. A range of functional phenotypes associated with the healthy brain or disease states has been suggested from in vivo work and were modeled in vitro as surveying, reactive, and primed sub-types of primary rat microglia and mixed microglia/astrocytes. It was hypothesized that the biomolecular profile of these cells undergoes a phenotypical change as well, and these functional phenotypes were explored for potential novel peptide binders using a custom 7 amino acid-presenting M13 phage library (SX7) to identify unique peptides that bind differentially to these respective cell types. Surveying glia were untreated, reactive were induced with a lipopolysaccharide treatment, recovery was modeled with a potent anti-inflammatory treatment dexamethasone, and priming was determined by subsequently challenging the cells with interferon gamma. Microglial function was profiled by determining the secretion of cytokines and nitric oxide, and expression of inducible nitric oxide synthase. After incubation with the SX7 phage library, populations of SX7-positive microglia and/or astrocytes were collected using fluorescence-activated cell sorting, SX7 phage was amplified in Escherichia coli culture, and phage DNA was sequenced via next-generation sequencing. Binding validation was done with synthesized peptides via in-cell westerns. Fifty-eight unique peptides were discovered, and their potential functions were assessed using a basic local alignment search tool. Peptides potentially originated from proteins ranging in function from a variety of supportive glial roles, including synapse support and pruning, to inflammatory incitement including cytokine and interleukin activation, and potential regulation in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- K M Koss
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - T Son
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - C Li
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - Y Hao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - J Cao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - M A Churchward
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biology and Environmental Sciences, Concordia University of Edmonton, Alberta, Edmonton, Canada
| | - Z J Zhang
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - J A Wertheim
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - R Derda
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - K G Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biomedical Engineering, University of Alberta, Alberta, Edmonton, Canada
| |
Collapse
|
6
|
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
|
7
|
Turner BRH, Jenkinson PI, Huttman M, Mullish BH. Inflammation, oxidative stress and gut microbiome perturbation: A narrative review of mechanisms and treatment of the alcohol hangover. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1451-1465. [PMID: 38965644 DOI: 10.1111/acer.15396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
Alcohol is the most widely abused substance in the world, the leading source of mortality in 15-49-year-olds, and a major risk factor for heart disease, liver disease, diabetes, and cancer. Despite this, alcohol is regularly misused in wider society. Consumers of excess alcohol often note a constellation of negative symptoms, known as the alcohol hangover. However, the alcohol hangover is not considered to have long-term clinical significance by clinicians or consumers. We undertook a critical review of the literature to demonstrate the pathophysiological mechanisms of the alcohol hangover. Hereafter, the alcohol hangover is re-defined as a manifestation of sickness behavior secondary to alcohol-induced inflammation, using the Bradford-Hill criteria to demonstrate causation above correlation. Alcohol causes inflammation through oxidative stress and endotoxemia. Alcohol metabolism is oxidative and increased intake causes relative tissue hypoxia and increased free radical generation. Tissue damage ensues through lipid peroxidation and the formation of DNA/protein adducts. Byproducts of alcohol metabolism such as acetaldehyde and congeners, sleep deprivation, and the activation of nonspecific inducible CYP2E1 in alcohol-exposed tissues exacerbate free radical generation. Tissue damage and cell death lead to inflammation, but in the intestine loss of epithelial cells leads to intestinal permeability, allowing the translocation of pathogenic bacteria to the systemic circulation (endotoxemia). This leads to a well-characterized cascade of systemic inflammation, additionally activating toll-like receptor 4 to induce sickness behavior. Considering the evidence, it is suggested that hangover frequency and severity may be predictors of the development of later alcohol-related diseases, meriting formal confirmation in prospective studies. In light of the mechanisms of alcohol-mediated inflammation, research into gut permeability and the gut microbiome may be an exciting future therapeutic avenue to prevent alcohol hangover and other alcohol-related diseases.
Collapse
Affiliation(s)
| | - Poppy I Jenkinson
- Department of Anaesthetics, Royal Surrey County Hospital, Surrey, UK
| | - Marc Huttman
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
8
|
Hedayati-Moghadam M, Razazpour F, Pourfridoni M, Mirzaee F, Baghcheghi Y. Ethanol's impact on the brain: a neurobiological perspective on the mechanisms of memory impairment. Mol Biol Rep 2024; 51:782. [PMID: 38918289 DOI: 10.1007/s11033-024-09748-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
Alcohol consumption is known to have detrimental effects on memory function, with various studies implicating ethanol in the impairment of cognitive processes related to memory retention and retrieval. This review aims to elucidate the complex neurobiological mechanisms underlying ethanol-induced memory impairment. Through a thorough search of existing literature using electronic databases, relevant articles focusing on the neurobiological mechanisms of ethanol on memory were identified and critically evaluated. This review focuses on the molecular and neural pathways through which ethanol exerts its effects on memory formation, consolidation, and recall processes. Key findings from the included studies shed light on the impact of ethanol on neurotransmitter systems, synaptic plasticity, and neuroinflammation in relation to memory impairment. This review contributes to a better understanding of the intricate mechanisms by which alcohol impairs memory function, offering insights for future research directions and the development of targeted interventions to alleviate these cognitive impairments.
Collapse
Affiliation(s)
- Mahdiyeh Hedayati-Moghadam
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Fateme Razazpour
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Mohammad Pourfridoni
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Faezeh Mirzaee
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
| | - Yousef Baghcheghi
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran.
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
9
|
Gómez GI, García-Rodríguez C, Marillán JE, Vergara SA, Alvear TF, Farias-Pasten A, Sáez JC, Retamal MA, Rovegno M, Ortiz FC, Orellana JA. Acute activation of hemichannels by ethanol leads to Ca 2+-dependent gliotransmitter release in astrocytes. Front Cell Dev Biol 2024; 12:1422978. [PMID: 38974144 PMCID: PMC11224458 DOI: 10.3389/fcell.2024.1422978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024] Open
Abstract
Multiple studies have demonstrated that acute ethanol consumption alters brain function and cognition. Nevertheless, the mechanisms underlying this phenomenon remain poorly understood. Astrocyte-mediated gliotransmission is crucial for hippocampal plasticity, and recently, the opening of hemichannels has been found to play a relevant role in this process. Hemichannels are plasma membrane channels composed of six connexins or seven pannexins, respectively, that oligomerize around a central pore. They serve as ionic and molecular exchange conduits between the cytoplasm and extracellular milieu, allowing the release of various paracrine substances, such as ATP, D-serine, and glutamate, and the entry of ions and other substances, such as Ca2+ and glucose. The persistent and exacerbated opening of hemichannels has been associated with the pathogenesis and progression of several brain diseases for at least three mechanisms. The uncontrolled activity of these channels could favor the collapse of ionic gradients and osmotic balance, the release of toxic levels of ATP or glutamate, cell swelling and plasma membrane breakdown and intracellular Ca2+ overload. Here, we evaluated whether acute ethanol exposure affects the activity of astrocyte hemichannels and the possible repercussions of this phenomenon on cytoplasmatic Ca2+ signaling and gliotransmitter release. Acute ethanol exposure triggered the rapid activation of connexin43 and pannexin1 hemichannels in astrocytes, as measured by time-lapse recordings of ethidium uptake. This heightened activity derived from a rapid rise in [Ca2+]i linked to extracellular Ca2+ influx and IP3-evoked Ca2+ release from intracellular Ca2+ stores. Relevantly, the acute ethanol-induced activation of hemichannels contributed to a persistent secondary increase in [Ca2+]i. The [Ca2+]i-dependent activation of hemichannels elicited by ethanol caused the increased release of ATP and glutamate in astroglial cultures and brain slices. Our findings offer fresh perspectives on the potential mechanisms behind acute alcohol-induced brain abnormalities and propose targeting connexin43 and pannexin1 hemichannels in astrocytes as a promising avenue to prevent deleterious consequences of alcohol consumption.
Collapse
Affiliation(s)
- Gonzalo I. Gómez
- Faculty of Health Sciences, Institute of Biomedical Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Claudia García-Rodríguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Jesús E. Marillán
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio A. Vergara
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tanhia F. Alvear
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Arantza Farias-Pasten
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan C. Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Mauricio A. Retamal
- Programa de Comunicación Celular en Cancer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C. Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
10
|
Gómez GI, Alvear TF, Roa DA, Farias-Pasten A, Vergara SA, Mellado LA, Martinez-Araya CJ, Prieto-Villalobos J, García-Rodríguez C, Sánchez N, Sáez JC, Ortíz FC, Orellana JA. Cx43 hemichannels and panx1 channels contribute to ethanol-induced astrocyte dysfunction and damage. Biol Res 2024; 57:15. [PMID: 38576018 PMCID: PMC10996276 DOI: 10.1186/s40659-024-00493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Alcohol, a widely abused drug, significantly diminishes life quality, causing chronic diseases and psychiatric issues, with severe health, societal, and economic repercussions. Previously, we demonstrated that non-voluntary alcohol consumption increases the opening of Cx43 hemichannels and Panx1 channels in astrocytes from adolescent rats. However, whether ethanol directly affects astroglial hemichannels and, if so, how this impacts the function and survival of astrocytes remains to be elucidated. RESULTS Clinically relevant concentrations of ethanol boost the opening of Cx43 hemichannels and Panx1 channels in mouse cortical astrocytes, resulting in the release of ATP and glutamate. The activation of these large-pore channels is dependent on Toll-like receptor 4, P2X7 receptors, IL-1β and TNF-α signaling, p38 mitogen-activated protein kinase, and inducible nitric oxide (NO) synthase. Notably, the ethanol-induced opening of Cx43 hemichannels and Panx1 channels leads to alterations in cytokine secretion, NO production, gliotransmitter release, and astrocyte reactivity, ultimately impacting survival. CONCLUSION Our study reveals a new mechanism by which ethanol impairs astrocyte function, involving the sequential stimulation of inflammatory pathways that further increase the opening of Cx43 hemichannels and Panx1 channels. We hypothesize that targeting astroglial hemichannels could be a promising pharmacological approach to preserve astrocyte function and synaptic plasticity during the progression of various alcohol use disorders.
Collapse
Affiliation(s)
- Gonzalo I Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Tanhia F Alvear
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Daniela A Roa
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Arantza Farias-Pasten
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Sergio A Vergara
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Luis A Mellado
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Claudio J Martinez-Araya
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Juan Prieto-Villalobos
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Claudia García-Rodríguez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, 2360102, Chile
| | - Natalia Sánchez
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan C Sáez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, 2360102, Chile
| | - Fernando C Ortíz
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Mechanisms of Myelin Formation and Repair Laboratory, Chacabuco 675, of. 212, Santiago, 8350347, Chile.
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile.
| |
Collapse
|
11
|
Mellado S, Cuesta CM, Montagud S, Rodríguez‐Arias M, Moreno‐Manzano V, Guerri C, Pascual M. Therapeutic role of mesenchymal stem cell-derived extracellular vesicles in neuroinflammation and cognitive dysfunctions induced by binge-like ethanol treatment in adolescent mice. CNS Neurosci Ther 2023; 29:4018-4031. [PMID: 37381698 PMCID: PMC10651955 DOI: 10.1111/cns.14326] [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: 01/04/2023] [Revised: 04/10/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are heterogeneous membrane vesicles secreted by cells in extracellular spaces that play an important role in intercellular communication under both normal and pathological conditions. Mesenchymal stem cells (MSC) are anti-inflammatory and immunoregulatory cells capable of secreting EVs, which are considered promising molecules for treating immune, inflammatory, and degenerative diseases. Our previous studies demonstrate that, by activating innate immune receptors TLR4 (Toll-like receptor 4), binge-like ethanol exposure in adolescence causes neuroinflammation and neural damage. AIMS To evaluate whether the intravenous administration of MSC-derived EVs is capable of reducing neuroinflammation, myelin and synaptic alterations, and the cognitive dysfunction induced by binge-like ethanol treatment in adolescent mice. MATERIALS & METHODS MSC-derived EVs obtained from adipose tissue were administered in the tail vein (50 microg/dose, one weekly dose) to female WT adolescent mice treated intermittently with ethanol (3.0 g/kg) during two weeks. RESULTS MSC-derived EVs from adipose tissue ameliorate ethanol-induced up-regulation of inflammatory genes (e.g., COX-2, iNOS, MIP-1α, NF-κB, CX3CL1, and MCP-1) in the prefrontal cortex of adolescent mice. Notably, MSC-derived EVs also restore the myelin and synaptic derangements, and the memory and learning impairments, induced by ethanol treatment. Using cortical astroglial cells in culture, our results further confirm that MSC-derived EVs decrease inflammatory genes in ethanol-treated astroglial cells. This, in turn, confirms in vivo findings. CONCLUSION Taken together, these results provide the first evidence for the therapeutic potential of the MSC-derived EVs in the neuroimmune response and cognitive dysfunction induced by binge alcohol drinking in adolescence.
Collapse
Affiliation(s)
- Susana Mellado
- Department of Physiology, School of Medicine and DentistryUniversity of ValenciaValenciaSpain
| | - Carlos M. Cuesta
- Department of Physiology, School of Medicine and DentistryUniversity of ValenciaValenciaSpain
| | - Sandra Montagud
- Department of Psychobiology, Facultad de PsicologíaUniversitat de ValenciaValenciaSpain
| | - Marta Rodríguez‐Arias
- Department of Psychobiology, Facultad de PsicologíaUniversitat de ValenciaValenciaSpain
| | | | | | - María Pascual
- Department of Physiology, School of Medicine and DentistryUniversity of ValenciaValenciaSpain
| |
Collapse
|
12
|
Munshi S, Albrechet-Souza L, Dos-Santos RC, Stelly CE, Secci ME, Gilpin NW, Tasker JG. Acute Ethanol Modulates Synaptic Inhibition in the Basolateral Amygdala via Rapid NLRP3 Inflammasome Activation and Regulates Anxiety-Like Behavior in Rats. J Neurosci 2023; 43:7902-7912. [PMID: 37739795 PMCID: PMC10669756 DOI: 10.1523/jneurosci.1744-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 08/22/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023] Open
Abstract
Chronic alcohol exposure leads to a neuroinflammatory response involving activation of the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome and proinflammatory cytokine production. Acute ethanol (EtOH) exposure activates GABAergic synapses in the central and basolateral amygdala (BLA) ex vivo, but whether this rapid modulation of synaptic inhibition is because of an acute inflammatory response and alters anxiety-like behavior in male and female animals is not known. Here, we tested the hypotheses that acute EtOH facilitates inhibitory synaptic transmission in the BLA by activating the NLRP3 inflammasome-dependent acute inflammatory response, that the alcohol-induced increase in inhibition is cell type and sex dependent, and that acute EtOH in the BLA reduces anxiety-like behavior. Acute EtOH application at a binge-like concentration (22-44 mm) stimulated synaptic GABA release from putative parvalbumin (PV) interneurons onto BLA principal neurons in ex vivo brain slices from male, but not female, rats. The EtOH facilitation of synaptic inhibition was blocked by antagonists of the Toll-like receptor 4 (TLR4), the NLRP3 inflammasome, and interleukin-1 receptors, suggesting it was mediated by a rapid local neuroinflammatory response in the BLA. In vivo, bilateral injection of EtOH directly into the BLA produced an acute concentration-dependent reduction in anxiety-like behavior in male but not female rats. These findings demonstrate that acute EtOH in the BLA regulates anxiety-like behavior in a sex-dependent manner and suggest that this effect is associated with presynaptic facilitation of parvalbumin-expressing interneuron inputs to BLA principal neurons via a local NLRP3 inflammasome-dependent neuroimmune response.SIGNIFICANCE STATEMENT Chronic alcohol exposure produces a neuroinflammatory response, which contributes to alcohol-associated pathologies. Acute alcohol administration increases inhibitory synaptic signaling in the brain, but the mechanism for the rapid alcohol facilitation of inhibitory circuits is unknown. We found that acute ethanol at binge-like concentrations in the basolateral amygdala (BLA) facilitates GABA release from parvalbumin-expressing (PV) interneuron synapses onto principal neurons in ex vivo brain slices from male rats and that intra-BLA ethanol reduces anxiety-like behavior in vivo in male rats, but not female rats. The ethanol (EtOH) facilitation of inhibition in the BLA is mediated by Toll-like receptor 4 (TLR4) and nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome activation and proinflammatory IL-1β signaling, which suggests a rapid NLRP3 inflammasome-dependent neuroimmune cascade that plays a critical role in acute alcohol intoxication.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Lucas Albrechet-Souza
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | | | - Claire E Stelly
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Maria E Secci
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Southeast Louisiana Veterans Affairs Healthcare System (SLVHCS), New Orleans, Louisiana 70119
| | - Nicholas W Gilpin
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Southeast Louisiana Veterans Affairs Healthcare System (SLVHCS), New Orleans, Louisiana 70119
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana 70118
- Southeast Louisiana Veterans Affairs Healthcare System (SLVHCS), New Orleans, Louisiana 70119
| |
Collapse
|
13
|
Mukherjee S, Tarale P, Sarkar DK. Neuroimmune Interactions in Fetal Alcohol Spectrum Disorders: Potential Therapeutic Targets and Intervention Strategies. Cells 2023; 12:2323. [PMID: 37759545 PMCID: PMC10528917 DOI: 10.3390/cells12182323] [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: 07/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a set of abnormalities caused by prenatal exposure to ethanol and are characterized by developmental defects in the brain that lead to various overt and non-overt physiological abnormalities. Growing evidence suggests that in utero alcohol exposure induces functional and structural abnormalities in gliogenesis and neuron-glia interactions, suggesting a possible role of glial cell pathologies in the development of FASD. However, the molecular mechanisms of neuron-glia interactions that lead to the development of FASD are not clearly understood. In this review, we discuss glial cell pathologies with a particular emphasis on microglia, primary resident immune cells in the brain. Additionally, we examine the involvement of several neuroimmune molecules released by glial cells, their signaling pathways, and epigenetic mechanisms responsible for FASD-related alteration in brain functions. Growing evidence suggests that extracellular vesicles (EVs) play a crucial role in the communication between cells via transporting bioactive cargo from one cell to the other. This review emphasizes the role of EVs in the context of neuron-glia interactions during prenatal alcohol exposure. Finally, some potential applications involving nutritional, pharmacological, cell-based, and exosome-based therapies in the treatment of FASD are discussed.
Collapse
Affiliation(s)
- Sayani Mukherjee
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
- Hormone Laboratory Research Group, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Jonas Lies vei 91B, 5021 Bergen, Norway
| | - Prashant Tarale
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
| | - Dipak K. Sarkar
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
| |
Collapse
|
14
|
Chandrashekar DV, Steinberg RA, Han D, Sumbria RK. Alcohol as a Modifiable Risk Factor for Alzheimer's Disease-Evidence from Experimental Studies. Int J Mol Sci 2023; 24:9492. [PMID: 37298443 PMCID: PMC10253673 DOI: 10.3390/ijms24119492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment and memory loss. Epidemiological evidence suggests that heavy alcohol consumption aggravates AD pathology, whereas low alcohol intake may be protective. However, these observations have been inconsistent, and because of methodological discrepancies, the findings remain controversial. Alcohol-feeding studies in AD mice support the notion that high alcohol intake promotes AD, while also hinting that low alcohol doses may be protective against AD. Chronic alcohol feeding to AD mice that delivers alcohol doses sufficient to cause liver injury largely promotes and accelerates AD pathology. The mechanisms by which alcohol can modulate cerebral AD pathology include Toll-like receptors, protein kinase-B (Akt)/mammalian target of rapamycin (mTOR) pathway, cyclic adenosine monophosphate (cAMP) response element-binding protein phosphorylation pathway, glycogen synthase kinase 3-β, cyclin-dependent kinase-5, insulin-like growth factor type-1 receptor, modulation of β-amyloid (Aβ) synthesis and clearance, microglial mediated, and brain endothelial alterations. Besides these brain-centric pathways, alcohol-mediated liver injury may significantly affect brain Aβ levels through alterations in the peripheral-to-central Aβ homeostasis. This article reviews published experimental studies (cell culture and AD rodent models) to summarize the scientific evidence and probable mechanisms (both cerebral and hepatic) by which alcohol promotes or protects against AD progression.
Collapse
Affiliation(s)
- Devaraj V. Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA;
| | - Ross A. Steinberg
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (R.A.S.); (D.H.)
| | - Derick Han
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (R.A.S.); (D.H.)
| | - Rachita K. Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA;
- Department of Neurology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
15
|
Valles SL, Singh SK, Campos-Campos J, Colmena C, Campo-Palacio I, Alvarez-Gamez K, Caballero O, Jorda A. Functions of Astrocytes under Normal Conditions and after a Brain Disease. Int J Mol Sci 2023; 24:ijms24098434. [PMID: 37176144 PMCID: PMC10179527 DOI: 10.3390/ijms24098434] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
In the central nervous system (CNS) there are a greater number of glial cells than neurons (between five and ten times more). Furthermore, they have a greater number of functions (more than eight functions). Glia comprises different types of cells, those of neural origin (astrocytes, radial glia, and oligodendroglia) and differentiated blood monocytes (microglia). During ontogeny, neurons develop earlier (at fetal day 15 in the rat) and astrocytes develop later (at fetal day 21 in the rat), which could indicate their important and crucial role in the CNS. Analysis of the phylogeny reveals that reptiles have a lower number of astrocytes compared to neurons and in humans this is reversed, as there have a greater number of astrocytes compared to neurons. These data perhaps imply that astrocytes are important and special cells, involved in many vital functions, including memory, and learning processes. In addition, astrocytes are involved in different mechanisms that protect the CNS through the production of antioxidant and anti-inflammatory proteins and they clean the extracellular environment and help neurons to communicate correctly with each other. The production of inflammatory mediators is important to prevent changes in brain homeostasis. On the contrary, excessive, or continued production appears as a characteristic element in many diseases, such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and in neurodevelopmental diseases, such as bipolar disorder, schizophrenia, and autism. Furthermore, different drugs and techniques have been developed to reverse oxidative stress and/or excess of inflammation that occurs in many CNS diseases, but much remains to be investigated. This review attempts to highlight the functional relevance of astrocytes in normal and neuropathological conditions by showing the molecular and cellular mechanisms of their role in the CNS.
Collapse
Affiliation(s)
- Soraya L Valles
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Lucknow 226002, India
| | - Juan Campos-Campos
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Carlos Colmena
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Ignacio Campo-Palacio
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Kenia Alvarez-Gamez
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Oscar Caballero
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
16
|
Niedzwiedz-Massey VM, Douglas JC, Rafferty T, Johnson JW, Holloway KN, Berquist MD, Kane CJ, Drew PD. Effects of chronic and binge ethanol administration on mouse cerebellar and hippocampal neuroinflammation. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:345-358. [PMID: 36345683 PMCID: PMC10615135 DOI: 10.1080/00952990.2022.2128361] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 11/10/2022]
Abstract
Background: Hippocampal and cerebellar neuropathology occurs in individuals with alcohol use disorders (AUD), resulting in impaired cognitive and motor function.Objectives: Evaluate the effects of ethanol on the expression of pro- and anti-inflammatory molecules, as well as the effects of the anti-inflammatory PPAR-γ agonist pioglitazone in suppressing ethanol-induced neuroinflammation.Methods: Adult male and female mice were treated chronically with ethanol for just under a month followed by a single acute binge dose of ethanol. Animals were provided liquid diet in the absence of ethanol (Control; n = 18, 9 M/9F), liquid diet containing ethanol (ethanol; n = 22, 11 M/11F), or liquid diet containing ethanol plus gavage administration of 30.0 mg/kg pioglitazone (ethanol + pioglitazone; n = 20, 10 M/10F). The hippocampus and cerebellum were isolated 24 h following the binge dose of ethanol, mRNA was isolated, and pro- and anti-inflammatory molecules were quantified by qRT-PCR.Results: Ethanol significantly (p < .05) increased the expression of pro-inflammatory molecules IL-1β, TNF-α, CCL2, and COX2; increased the expression of inflammasome-related molecules NLRP3 and Casp1 but decreased IL-18; and altered the expression of anti-inflammatory molecules including TGFβR1 in the hippocampus and cerebellum, though some differences were observed between males and females and the two brain regions. The anti-inflammatory pioglitazone inhibited ethanol-induced alterations in the expression of most, but not all, inflammation-related molecules.Conclusion: Chronic plus binge administration of ethanol induced the expression of inflammatory molecules in adult mice and pioglitazone suppressed ethanol-induced neuroinflammation.
Collapse
Affiliation(s)
- Victoria M. Niedzwiedz-Massey
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - James C. Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Tonya Rafferty
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jennifer W. Johnson
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kalee N. Holloway
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Michael D. Berquist
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Cynthia J.M. Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul D. Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
17
|
Gano A, Deak T, Pautassi RM. A review on the reciprocal interactions between neuroinflammatory processes and substance use and misuse, with a focus on alcohol misuse. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:269-282. [PMID: 37148274 PMCID: PMC10524510 DOI: 10.1080/00952990.2023.2201944] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 05/08/2023]
Abstract
Background: The last decade has witnessed a surge of findings implicating neuroinflammatory processes as pivotal players in substance use disorders. The directionality of effects began with the expectation that the neuroinflammation associated with prolonged substance misuse contributes to long-term neuropathological consequences. As the literature grew, however, it became evident that the interactions between neuroinflammatory processes and alcohol and drug intake were reciprocal and part of a pernicious cycle in which disease-relevant signaling pathways contributed to an escalation of drug intake, provoking further inflammation-signaling and thereby exacerbating the neuropathological effects of drug misuse.Objectives: The goal of this review and its associated special issue is to provide an overview of the emergent findings relevant to understanding these reciprocal interactions. The review highlights the importance of preclinical and clinical studies in testing and validation of immunotherapeutics as viable targets for curtailing substance use and misuse, with a focus on alcohol misuse.Methods: A narrative review of the literature on drug and neuroinflammation was conducted, as well as articles published in this Special Issue on Alcohol- and Drug-induced Neuroinflammation: Insights from Pre-clinical Models and Clinical Research.Results: We argue that (a) demographic variables and genetic background contribute unique sensitivity to drug-related neuroinflammation; (b) co-morbidities between substance use disorders and affect dysfunction may share common inflammation-related signatures that predict the efficacy of immunotherapeutic drugs; and (c) examination of polydrug interactions with neuroinflammation is a critical area where greater research emphasis is needed.Conclusions: This review provides an accessible and example-driven review of the relationship between drug misuse, neuroinflammatory processes, and their resultant neuropathological outcomes.
Collapse
Affiliation(s)
- Anny Gano
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY 13902-6000, United States of America
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY 13902-6000, United States of America
| | - Ricardo Marcos Pautassi
- Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC – CONICET-Universidad Nacional de Córdoba), Córdoba, 5000, Argentina
| |
Collapse
|
18
|
Mondello JE, Gano A, Vore AS, Deak T. Cues associated with repeated ethanol exposure facilitate the corticosterone response to ethanol and immunological challenges in adult male Sprague Dawley rats: implications for neuroimmune regulation. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:359-369. [PMID: 36862971 PMCID: PMC10474242 DOI: 10.1080/00952990.2023.2169831] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 03/04/2023]
Abstract
Background: We previously found a conditioned increase in central neuroinflammatory markers (Interleukin 6; IL-6) following exposure to alcohol-associated cues. Recent studies suggest (unconditioned) induction of IL-6 is entirely dependent on ethanol-induced corticosterone.Objectives: The goals of these present studies were to test whether alcohol-paired cues facilitated the hypothalamic-pituitary-adrenal (HPA) axis response to either a subthreshold priming alcohol dose or an immune or psychological stress challengeMethods: In Experiment 1 (N = 64), adult male Sprague Dawley rats were trained (paired or unpaired, four pairings total) with either vehicle or 2 g/kg alcohol [intragastric (i.g.) or intraperitoneal (i.p.)] injections. In Experiments 2 (N = 28) and 3 (N = 30), male rats were similarly trained but with 4 g/kg alcohol i.g. intubations. On test day, all rats were either administered a 0.5 g/kg alcohol dose (i.p. or i.g. Experiment 1), a 100 µg/kg i.p. lipopolysaccharide (LPS) challenge (Experiment 2), or a restraint challenge (Experiment 3), and exposed to alcohol-associated cues. Blood plasma was collected for analysis.Results: Alcohol-associated cues facilitated the plasma corticosterone response to a subthreshold dose of alcohol (F1,28 = 4.85, p < .05) and an immune challenge (F8,80 = 6.23, p < .001), but not a restraint challenge (F2,27 = 0.18, p > .05).Conclusion: These findings reveal that the impact of the cues associated with alcohol intoxication on the HPA axis may be context-specific. This work illustrates how HPA axis learning processes form in the early stages of alcohol use and has important implications for how the HPA and neuroimmune conditioning may develop in alcohol use disorder in humans and facilitate the response to a later immune challenge.
Collapse
Affiliation(s)
- Jamie E. Mondello
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| | - Anny Gano
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| | - Andrew S. Vore
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| |
Collapse
|
19
|
Gruol DL, Calderon D, French K, Melkonian C, Huitron-Resendiz S, Cates-Gatto C, Roberts AJ. Neuroimmune interactions with binge alcohol drinking in the cerebellum of IL-6 transgenic mice. Neuropharmacology 2023; 228:109455. [PMID: 36775097 PMCID: PMC10029700 DOI: 10.1016/j.neuropharm.2023.109455] [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: 06/08/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
The neuroimmune system of the brain, which is comprised primarily of astrocytes and microglia, regulates a variety of homeostatic mechanisms that underlie normal brain function. Numerous conditions, including alcohol consumption, can disrupt this regulatory process by altering brain levels of neuroimmune factors. Alcohol and neuroimmune factors, such as proinflammatory cytokines IL-6 and TNF-alpha, act at similar targets in the brain, including excitatory and inhibitory synaptic transmission. Thus, alcohol-induced production of IL-6 and/or TNF-alpha could be important contributing factors to the effects of alcohol on the brain. Recent studies indicate that IL-6 plays a role in alcohol drinking and the effects of alcohol on the brain activity following the cessation of alcohol consumption (post-alcohol period), however information on these topics is limited. Here we used homozygous and heterozygous female and male transgenic mice with increased astrocyte expression of IL-6 to examined further the interactions between alcohol and IL-6 with respect to voluntary alcohol drinking, brain activity during the post-alcohol period, IL-6 signal transduction, and expression of synaptic proteins. Wildtype littermates (WT) served as controls. The transgenic mice model brain neuroimmune status with respect to IL-6 in subjects with a history of persistent alcohol use. Results showed a genotype dependent reduction in voluntary alcohol consumption in the Drinking in the Dark protocol and in frequency-dependent relationships between brain activity in EEG recordings during the post-alcohol period and alcohol consumption. IL-6, TNF-alpha, IL-6 signal transduction partners pSTAT3 and c/EBP beta, and synaptic proteins were shown to play a role in these genotypic effects.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Delilah Calderon
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Katharine French
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Claudia Melkonian
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
20
|
Perpiñá-Clérigues C, Mellado S, Català-Senent JF, Ibáñez F, Costa P, Marcos M, Guerri C, García-García F, Pascual M. Lipidomic landscape of circulating extracellular vesicles isolated from adolescents exposed to ethanol intoxication: a sex difference study. Biol Sex Differ 2023; 14:22. [PMID: 37085905 PMCID: PMC10120207 DOI: 10.1186/s13293-023-00502-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Lipids represent essential components of extracellular vesicles (EVs), playing structural and regulatory functions during EV biogenesis, release, targeting, and cell uptake. Importantly, lipidic dysregulation has been linked to several disorders, including metabolic syndrome, inflammation, and neurological dysfunction. Our recent results demonstrated the involvement of plasma EV microRNAs as possible amplifiers and biomarkers of neuroinflammation and brain damage induced by ethanol intoxication during adolescence. Considering the possible role of plasma EV lipids as regulatory molecules and biomarkers, we evaluated how acute ethanol intoxication differentially affected the lipid composition of plasma EVs in male and female adolescents and explored the participation of the immune response. METHODS Plasma EVs were extracted from humans and wild-type (WT) and Toll-like receptor 4 deficient (TLR4-KO) mice. Preprocessing and exploratory analyses were conducted after the extraction of EV lipids and data acquisition by mass spectrometry. Comparisons between ethanol-intoxicated and control human female and male individuals and ethanol-treated and untreated WT and TLR4-KO female and male mice were used to analyze the differential abundance of lipids. Annotation of lipids into their corresponding classes and a lipid set enrichment analysis were carried out to evaluate biological functions. RESULTS We demonstrated, for the first time, that acute ethanol intoxication induced a higher enrichment of distinct plasma EV lipid species in human female adolescents than in males. We observed a higher content of the PA, LPC, unsaturated FA, and FAHFA lipid classes in females, whereas males showed enrichment in PI. These lipid classes participate in the formation, release, and uptake of EVs and the activation of the immune response. Moreover, we observed changes in EV lipid composition between ethanol-treated WT and TLR4-KO mice (e.g., enrichment of glycerophosphoinositols in ethanol-treated WT males), and the sex-based differences in lipid abundance are more notable in WT mice than in TLR4-KO mice. All data and results generated have been made openly available on a web-based platform ( http://bioinfo.cipf.es/sal ). CONCLUSIONS Our results suggest that binge ethanol drinking in human female adolescents leads to a higher content of plasma EV lipid species associated with EV biogenesis and the propagation of neuroinflammatory responses than in males. In addition, we discovered greater differences in lipid abundance between sexes in WT mice compared to TLR4-KO mice. Our findings also support the potential use of EV-enriched lipids as biomarkers of ethanol-induced neuroinflammation during adolescence.
Collapse
Affiliation(s)
- Carla Perpiñá-Clérigues
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - José F Català-Senent
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Francesc Ibáñez
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - Pilar Costa
- Emergency Department, University Hospital of Salamanca-IBSAL, University of Salamanca, 37007, Salamanca, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007, Salamanca, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - Francisco García-García
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain.
| | - María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain.
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain.
| |
Collapse
|
21
|
Zhou H, Khan D, Gerdes N, Hagenbeck C, Rana M, Cornelius JF, Muhammad S. Colchicine Protects against Ethanol-Induced Senescence and Senescence-Associated Secretory Phenotype in Endothelial Cells. Antioxidants (Basel) 2023; 12:antiox12040960. [PMID: 37107335 PMCID: PMC10135532 DOI: 10.3390/antiox12040960] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Inflammaging is a potential risk factor for cardiovascular diseases. It results in the development of thrombosis and atherosclerosis. The accumulation of senescent cells in vessels causes vascular inflammaging and contributes to plaque formation and rupture. In addition to being an acquired risk factor for cardiovascular diseases, ethanol can induce inflammation and senescence, both of which have been implicated in cardiovascular diseases. In the current study, we used colchicine to abate the cellular damaging effects of ethanol on endothelial cells. Colchicine prevented senescence and averted oxidative stress in endothelial cells exposed to ethanol. It lowered the relative protein expression of aging and senescence marker P21 and restored expression of the DNA repair proteins KU70/KU80. Colchicine inhibited the activation of nuclear factor kappa B (NFκ-B) and mitogen activated protein kinases (MAPKs) in ethanol-treated endothelial cells. It reduced ethanol-induced senescence-associated secretory phenotype. In summary, we show that colchicine ameliorated the ethanol-caused molecular events, resulting in attenuated senescence and senescence-associated secretory phenotype in endothelial cells.
Collapse
Affiliation(s)
- Huakang Zhou
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Dilaware Khan
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Carsten Hagenbeck
- Clinic for Gynecology and Obstetrics, University Clinic, 40225 Düsseldorf, Germany
| | - Majeed Rana
- Department of Oral, Maxillofacial and Facial Plastic Surgery, University Hospital Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jan Frederick Cornelius
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
- Department of Neurosurgery, University Hospital Helsinki, Topeliuksenkatu 5, 00260 Helsinki, Finland
| |
Collapse
|
22
|
Holloway KN, Douglas JC, Rafferty TM, Majewska AK, Kane CJM, Drew PD. Ethanol-induced cerebellar transcriptomic changes in a postnatal model of fetal alcohol spectrum disorders: Focus on disease onset. Front Neurosci 2023; 17:1154637. [PMID: 37008214 PMCID: PMC10062483 DOI: 10.3389/fnins.2023.1154637] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a group of neurodevelopmental disorders caused by ethanol exposure in utero, which can result in neurocognitive and behavioral impairments, growth defects, and craniofacial anomalies. FASD affects up to 1-5% of school-aged children in the United States, and there is currently no cure. The underlying mechanisms involved in ethanol teratogenesis remain elusive and need greater understanding to develop and implement effective therapies. Using a third trimester human equivalent postnatal mouse model of FASD, we evaluate the transcriptomic changes induced by ethanol exposure in the cerebellum on P5 and P6, after only 1 or 2 days of ethanol exposure, with the goal of shedding light on the transcriptomic changes induced early during the onset and development of FASD. We have highlighted key pathways and cellular functions altered by ethanol exposure, which include pathways related to immune function and cytokine signaling as well as the cell cycle. Additionally, we found that ethanol exposure resulted in an increase in transcripts associated with a neurodegenerative microglia phenotype, and acute- and pan-injury reactive astrocyte phenotypes. Mixed effects on oligodendrocyte lineage cell associated transcripts and cell cycle associated transcripts were observed. These studies help to elucidate the underlying mechanisms that may be involved with the onset of FASD and provide further insights that may aid in identifying novel targets for interventions and therapeutics.
Collapse
Affiliation(s)
- Kalee N. Holloway
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - James C. Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Tonya M. Rafferty
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Ania K. Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
| | - Cynthia J. M. Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Paul D. Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
23
|
Carlson ER, Guerin SP, Nixon K, Fonken LK. The neuroimmune system - Where aging and excess alcohol intersect. Alcohol 2023; 107:153-167. [PMID: 36150610 PMCID: PMC10023388 DOI: 10.1016/j.alcohol.2022.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/21/2022] [Accepted: 08/31/2022] [Indexed: 01/05/2023]
Abstract
As the percentage of the global population over age 65 grows, and with it a subpopulation of individuals with alcohol use disorder (AUD), understanding the effect of alcohol on the aged brain is of utmost importance. Neuroinflammation is implicated in both natural aging as well as alcohol use, and its role in alterations to brain morphology and function may be exacerbated in aging individuals who drink alcohol to excess. The neuroimmune response to alcohol in aging is complex. The few studies investigating this issue have reported heightened basal activity and either hypo- or hyper-reactivity to an alcohol challenge. This review of preclinical research will first introduce key players of the immune system, then explore changes in neuroimmune function with aging or alcohol alone, with discussion of vulnerable brain regions, changes in cytokines, and varied reactions of microglia and astrocytes. We will then consider different levels of alcohol exposure, relevant animal models of AUD, and neuroimmune activation by alcohol across the lifespan. By identifying key findings, challenges, and targets for future research, we hope to bring more attention and resources to this underexplored area of inquiry.
Collapse
Affiliation(s)
- Erika R Carlson
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States
| | - Steven P Guerin
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States
| | - Kimberly Nixon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
24
|
Holloway KN, Pinson MR, Douglas JC, Rafferty TM, Kane CJM, Miranda RC, Drew PD. Cerebellar Transcriptomic Analysis in a Chronic plus Binge Mouse Model of Alcohol Use Disorder Demonstrates Ethanol-Induced Neuroinflammation and Altered Glial Gene Expression. Cells 2023; 12:745. [PMID: 36899881 PMCID: PMC10000476 DOI: 10.3390/cells12050745] [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: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Alcohol use disorder (AUD) is one of the most common preventable mental health disorders and can result in pathology within the CNS, including the cerebellum. Cerebellar alcohol exposure during adulthood has been associated with disruptions in proper cerebellar function. However, the mechanisms regulating ethanol-induced cerebellar neuropathology are not well understood. High-throughput next generation sequencing was performed to compare control versus ethanol-treated adult C57BL/6J mice in a chronic plus binge model of AUD. Mice were euthanized, cerebella were microdissected, and RNA was isolated and submitted for RNA-sequencing. Down-stream transcriptomic analyses revealed significant changes in gene expression and global biological pathways in control versus ethanol-treated mice that included pathogen-influenced signaling pathways and cellular immune response pathways. Microglial-associated genes showed a decrease in homeostasis-associated transcripts and an increase in transcripts associated with chronic neurodegenerative diseases, while astrocyte-associated genes showed an increase in transcripts associated with acute injury. Oligodendrocyte lineage cell genes showed a decrease in transcripts associated with both immature progenitors as well as myelinating oligodendrocytes. These data provide new insight into the mechanisms by which ethanol induces cerebellar neuropathology and alterations to the immune response in AUD.
Collapse
Affiliation(s)
- Kalee N. Holloway
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Marisa R. Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (M.R.P.); (R.C.M.)
| | - James C. Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Tonya M. Rafferty
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Cynthia J. M. Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
| | - Rajesh C. Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (M.R.P.); (R.C.M.)
| | - Paul D. Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.N.H.); (J.C.D.); (T.M.R.); (C.J.M.K.)
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
25
|
Alcohol-Induced Headache with Neuroinflammation: Recent Progress. FERMENTATION-BASEL 2023. [DOI: 10.3390/fermentation9020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Ethanol and other congeners in alcoholic beverages and foods are known triggers of alcohol-induced headaches (AIHs). Recent studies implicate AIHs as an important underlying factor for neuroinflammation. Studies show the relationship between alcoholic beverages, AIH agents, neuroinflammation, and the pathway they elicit. However, studies elucidating specific AIH agents’ pathways are scarce. Works reviewing their pathways can give invaluable insights into specific substances’ patterns and how they can be controlled. Hence, we reviewed the current understanding of how AIH agents in alcoholic beverages affect neuroinflammation and their specific roles. Ethanol upregulates transient receptor potential cation channel subfamily V member 1 (TRPV1) and Toll-like receptor 4 (TLR4) expression levels; both receptors trigger a neuroinflammation response that promotes AIH manifestation—the most common cause of AIHs. Other congeners such as histamine, 5-HT, and condensed tannins also upregulate TRPV1 and TLR4, neuroinflammatory conditions, and AIHs. Data elucidating AIH agents, associating pathways, and fermentation parameters can help reduce or eliminate AIH inducers and create healthier beverages.
Collapse
|
26
|
Akbari E, Hossaini D, Amiry GY, Ansari M, Haidary M, Beheshti F, Ahmadi-Soleimani SM. Vitamin B12 administration prevents ethanol-induced learning and memory impairment through re-establishment of the brain oxidant/antioxidant balance, enhancement of BDNF and suppression of GFAP. Behav Brain Res 2023; 438:114156. [PMID: 36243244 DOI: 10.1016/j.bbr.2022.114156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/26/2022] [Accepted: 10/08/2022] [Indexed: 12/03/2022]
Abstract
There are growing evidence indicating that the adolescent brain is persistently affected by the use of psychostimulant agents. In this regard, alcohol drinking has become rather common among the adolescents in many societies during the last decade. It is currently well known that long-term ethanol exposure deteriorates various cognitive functions such as learning and memory. Mechanistically, these adverse effects have been shown to be mediated by oxidative damage to central nervous system. On the other hand, Vit-B12 is known to improve cognitive performance by suppression of oxidative parameters. Thus, in the present study we aimed to test whether treatment by Vit-B12 could prevent ethanol-induced complications in mice using behavioral and biochemical methods. Different groups of male Syrian mice received ethanol, ethanol+Vit-B12, Vit-B12 alone, or saline during adolescence and then learning and memory functions were assessed by Morris water maze (MWM) and Passive Avoidance (PA) tests. Finally, mice were sacrificed for measurement of biochemical factors. Results indicated that, adolescent ethanol intake impairs learning and memory function through exacerbation of oxidative stress and Vit-B12 treatment improves these complications by re-establishment of oxidant/anti-oxidant balance in CNS. Moreover, we found that Vit-B12 prevents ethanol-induced reduction of BDNF and enhancement of GFAP and acetylcholinesterase (AChE) activity. In conclusion, it seems that Vit-B12 supplementation could be used as an effective therapeutic strategy to prevent learning and memory defects induced by chronic alcohol intake during adolescence.
Collapse
Affiliation(s)
- Elham Akbari
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran
| | - Dawood Hossaini
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran
| | - Ghulam Yahya Amiry
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran
| | - Mustafa Ansari
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran
| | - Murtaza Haidary
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran.
| | - S Mohammad Ahmadi-Soleimani
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran; Departments of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat, Heydariyeh, Iran.
| |
Collapse
|
27
|
Barkell GA, Parekh SV, Paniccia JE, Martin AJ, Reissner KJ, Knapp DJ, Robinson SL, Thiele TE, Lysle DT. Chronic ethanol consumption exacerbates future stress-enhanced fear learning, an effect mediated by dorsal hippocampal astrocytes. Alcohol Clin Exp Res 2022; 46:2177-2190. [PMID: 36349797 PMCID: PMC10187052 DOI: 10.1111/acer.14963] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/23/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD) are highly comorbid, yet there is a lack of preclinical research investigating how prior ethanol (EtOH) dependence influences the development of a PTSD-like phenotype. Furthermore, the neuroimmune system has been implicated in the development of both AUD and PTSD, but the extent of glial involvement in this context remains unclear. A rodent model was developed to address this gap in the literature. METHODS We used a 15-day exposure to the 5% w/v EtOH low-fat Lieber-DeCarli liquid diet in combination with the stress-enhanced fear learning (SEFL) paradigm to investigate the effects of chronic EtOH consumption on the development of a PTSD-like phenotype. Next, we used a reverse transcription quantitative real-time polymerase chain reaction to quantify mRNA expression of glial cell markers GFAP (astrocytes) and CD68 (microglia) following severe footshock stress in EtOH-withdrawn rats. Finally, we tested the functional contribution of dorsal hippocampal (DH) astrocytes in the development of SEFL in EtOH-dependent rats using astrocyte-specific Gi designer receptors exclusively activated by designer drugs (Gi -DREADD). RESULTS Results demonstrate that chronic EtOH consumption and withdrawal exacerbate future SEFL. Additionally, we found significantly increased GFAP mRNA expression in the dorsal and ventral hippocampus and amygdalar complex following the severe stressor in EtOH-withdrawn animals. Finally, the stimulation of the astroglial Gi -DREADD during EtOH withdrawal prevented the EtOH-induced enhancement of SEFL. CONCLUSIONS Collectively, results indicate that prior EtOH dependence and withdrawal combined with a severe stressor potentiate future enhanced fear learning. Furthermore, DH astrocytes significantly contribute to this change in behavior. Overall, these studies provide insight into the comorbidity of AUD and PTSD and the potential neurobiological mechanisms behind increased susceptibility to a PTSD-like phenotype in individuals with AUD.
Collapse
Affiliation(s)
- Gillian A Barkell
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shveta V Parekh
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jacqueline E Paniccia
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alia J Martin
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kathryn J Reissner
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Darin J Knapp
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stacey L Robinson
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Todd E Thiele
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Donald T Lysle
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
28
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
29
|
Yang J, Ran M, Li H, Lin Y, Ma K, Yang Y, Fu X, Yang S. New insight into neurological degeneration: Inflammatory cytokines and blood–brain barrier. Front Mol Neurosci 2022; 15:1013933. [PMID: 36353359 PMCID: PMC9637688 DOI: 10.3389/fnmol.2022.1013933] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Neurological degeneration after neuroinflammation, such as that resulting from Alzheimer’s disease (AD), stroke, multiple sclerosis (MS), and post-traumatic brain injury (TBI), is typically associated with high mortality and morbidity and with permanent cognitive dysfunction, which places a heavy economic burden on families and society. Diagnosing and curing these diseases in their early stages remains a challenge for clinical investigation and treatment. Recent insight into the onset and progression of these diseases highlights the permeability of the blood–brain barrier (BBB). The primary factor that influences BBB structure and function is inflammation, especially the main cytokines including IL-1β, TNFα, and IL-6, the mechanism on the disruption of which are critical component of the aforementioned diseases. Surprisingly, the main cytokines from systematic inflammation can also induce as much worse as from neurological diseases or injuries do. In this review, we will therefore discuss the physiological structure of BBB, the main cytokines including IL-1β, TNFα, IL-6, and their mechanism on the disruption of BBB and recent research about the main cytokines from systematic inflammation inducing the disruption of BBB and cognitive impairment, and we will eventually discuss the need to prevent the disruption of BBB.
Collapse
Affiliation(s)
- Jie Yang
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Mingzi Ran
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Anaesthesiology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Hongyu Li
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Ye Lin
- Department of Neurology, The First Medical Centre, PLA General Hospital, Beijing, China
| | - Kui Ma
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
| | - Yuguang Yang
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Xiaobing Fu,
| | - Siming Yang
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
- *Correspondence: Siming Yang,
| |
Collapse
|
30
|
Alcohol-Induced Oxidative Stress and the Role of Antioxidants in Alcohol Use Disorder: A Systematic Review. Antioxidants (Basel) 2022; 11:antiox11071374. [PMID: 35883865 PMCID: PMC9311529 DOI: 10.3390/antiox11071374] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder (AUD) is a highly prevalent, comorbid, and disabling disorder. The underlying mechanism of ethanol neurotoxicity and the involvement of oxidative stress is still not fully elucidated. However, ethanol metabolism has been associated with increased oxidative stress through alcohol dehydrogenase, the microsomal ethanol oxidation system, and catalase metabolic pathways. We searched the PubMed and genome-wide association studies (GWAS) catalog databases to review the literature systematically and summarized the findings focusing on AUD and alcohol abstinence in relation to oxidative stress. In addition, we reviewed the ClinicalTrials.gov resource of the US National Library of Medicine to identify all ongoing and completed clinical trials that include therapeutic interventions based on antioxidants. The retrieved clinical and preclinical studies show that oxidative stress impacts AUD through genetics, alcohol metabolism, inflammation, and neurodegeneration.
Collapse
|
31
|
Oyarce K, Cepeda MY, Lagos R, Garrido C, Vega-Letter AM, Garcia-Robles M, Luz-Crawford P, Elizondo-Vega R. Neuroprotective and Neurotoxic Effects of Glial-Derived Exosomes. Front Cell Neurosci 2022; 16:920686. [PMID: 35813501 PMCID: PMC9257100 DOI: 10.3389/fncel.2022.920686] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/06/2022] [Indexed: 12/19/2022] Open
Abstract
Exosomes derived from glial cells such as astrocytes, microglia, and oligodendrocytes can modulate cell communication in the brain and exert protective or neurotoxic effects on neurons, depending on the environmental context upon their release. Their isolation, characterization, and analysis under different conditions in vitro, in animal models and samples derived from patients has allowed to define the participation of other molecular mechanisms behind neuroinflammation and neurodegeneration spreading, and to propose their use as a potential diagnostic tool. Moreover, the discovery of specific molecular cargos, such as cytokines, membrane-bound and soluble proteins (neurotrophic factors, growth factors, misfolded proteins), miRNA and long-non-coding RNA, that are enriched in glial-derived exosomes with neuroprotective or damaging effects, or their inhibitors can now be tested as therapeutic tools. In this review we summarize the state of the art on how exosomes secretion by glia can affect neurons and other glia from the central nervous system in the context of neurodegeneration and neuroinflammation, but also, on how specific stress stimuli and pathological conditions can change the levels of exosome secretion and their properties.
Collapse
Affiliation(s)
- Karina Oyarce
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
| | - María Yamila Cepeda
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Raúl Lagos
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Camila Garrido
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ana María Vega-Letter
- Facultad de Medicina, Centro de Investigación Biomédica, Universidad de los Andes, Santiago, Chile
| | - María Garcia-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Patricia Luz-Crawford
- Facultad de Medicina, Centro de Investigación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- *Correspondence: Roberto Elizondo-Vega,
| |
Collapse
|
32
|
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: 3.0] [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
|
33
|
Liu J, Li JX, Wu R. Toll-Like Receptor 4: A Novel Target to Tackle Drug Addiction? Handb Exp Pharmacol 2022; 276:275-290. [PMID: 35434747 PMCID: PMC9829382 DOI: 10.1007/164_2022_586] [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: 01/12/2023]
Abstract
Drug addiction is a chronic brain disease characterized by compulsive drug-seeking and drug-taking behaviors despite the major negative consequences. Current well-established neuronal underpinnings of drug addiction have promoted the substantial progress in understanding this disorder. However, non-neuronal mechanisms of drug addiction have long been underestimated. Fortunately, increased evidence indicates that neuroimmune system, especially Toll-like receptor 4 (TLR4) signaling, plays an important role in the different stages of drug addiction. Drugs like opioids, psychostimulants, and alcohol activate TLR4 signaling and enhance the proinflammatory response, which is associated with drug reward-related behaviors. While extensive studies have shown that inhibition of TLR4 attenuated drug-related responses, there are conflicting findings implicating that TLR4 signaling may not be essential to drug addiction. In this chapter, preclinical and clinical studies will be discussed to further evaluate whether TLR4-based neuroimmune pharmacotherapy can be used to treat drug addiction. Furthermore, the possible mechanisms underlying the effects of TLR4 inhibition in modulating drug-related behaviors will also be discussed.
Collapse
Affiliation(s)
- Jianfeng Liu
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA,Corresponding authors: Dr. Jun-Xu Li, , Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY 14214. Tel: +1 716 829 2482; Fax: +1 716 829 2801 And Dr. Ruyan Wu, , School of Medicine, Yangzhou University, Yangzhou 225000, China
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA,School of Medicine, Yangzhou University, Yangzhou, China,Corresponding authors: Dr. Jun-Xu Li, , Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY 14214. Tel: +1 716 829 2482; Fax: +1 716 829 2801 And Dr. Ruyan Wu, , School of Medicine, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
34
|
Zhang G, Liu B, Zeng Z, Chen Q, Feng Y, Ning X. Oxymatrine hydrazone (OMTH) synthesis and its protective effect for rheumatoid arthritis through downregulation of MEK/NF-κB pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:2448-2453. [PMID: 34459104 DOI: 10.1002/tox.23357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/06/2021] [Accepted: 08/15/2021] [Indexed: 06/13/2023]
Abstract
Rheumatoid arthritis (RA) is one of the inflammatory diseases detected in more than 1% of the world population. In the present study, oxymatrine hydrazone (OMTH) was synthesized and investigated for treatment of RA in vitro in TNF-α induced fibroblast-like synoviocyte cell model. Cell viability and apoptosis were detected using MTT and flow cytometry assays, respectively. ELISA was used for determination of inflammatory cytokines and western blotting for evaluation of protein expression. Pretreatment of HFLS-RA cells with 0.5, 1.0, 1.5, 2.0, and 2.5 μM doses of OMTH suppressed TNF-α induced promotion of proliferative potential in dose-based manner. The OMTH pretreatment of TNF-α exposed HFLS-RA cells significantly increased apoptotic cell proportion. In TNF-α exposed HFLS-RA cells OMTH pretreatment elevated Bax and suppressed Bcl-2 expression. Treatment of HFLS-RA cells with OMTH prevented TNF-α mediated elevation of IL-1β, IL-6 and IL-8. Moreover, OMTH treatment of HFLS-RA cells effectively suppressed TNF-α mediated elevated levels of MMP-1 and MMP-13. Pretreatment of HFLS-RA cells with OMTH reversed TNF-α mediated promotion of iNOS and COX-2 levels. The MEK/1/2 and p65 phosphorylation in TNF-α exposed HFLS-RA cells was reduced by OMTH pre-treatment in dose-based manner. Thus, OMTH successfully inhibited TNF-α-mediated increased viability of RA synovial cells and activated apoptosis. Pretreatment of TNF-α exposed synovial cells with OMTH targeted phosphorylation of MEK/NF-κB. Therefore, OMTH may act as potential therapeutic agent for RA treatment.
Collapse
Affiliation(s)
- Guangwen Zhang
- Department of Arthroplasty Surgery, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Baojian Liu
- Department of Arthroplasty Surgery, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhaoyang Zeng
- Department of Arthroplasty Surgery, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Qinghai Chen
- Department of Arthroplasty Surgery, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Yiyun Feng
- Department of Arthroplasty Surgery, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xueqian Ning
- Department of Arthroplasty Surgery, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
35
|
Cuesta CM, Pascual M, Pérez-Moraga R, Rodríguez-Navarro I, García-García F, Ureña-Peralta JR, Guerri C. TLR4 Deficiency Affects the Microbiome and Reduces Intestinal Dysfunctions and Inflammation in Chronic Alcohol-Fed Mice. Int J Mol Sci 2021; 22:ijms222312830. [PMID: 34884634 PMCID: PMC8657603 DOI: 10.3390/ijms222312830] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/21/2022] Open
Abstract
Chronic alcohol abuse causes an inflammatory response in the intestinal tract with damage to the integrity of the mucosa and epithelium, as well as dysbiosis in the gut microbiome. However, the role of gut bacteria in ethanol effects and how these microorganisms interact with the immune system are not well understood. The aim of the present study was to evaluate if TLR4 alters the ethanol-induced intestinal inflammatory response, and whether the response of this receptor affects the gut microbiota profile. We analyzed the 16S rRNA sequence of the fecal samples from wild-type (WT) and TLR4-knockout (TLR4-KO) mice with and without ethanol intake for 3 months. The results demonstrated that chronic ethanol consumption reduces microbiota diversity and causes dysbiosis in WT mice. Likewise, ethanol upregulates several inflammatory genes (IL-1β, iNOS, TNF-α) and miRNAs (miR-155-5p, miR-146a-5p) and alters structural and permeability genes (INTL1, CDH1, CFTR) in the colon of WT mice. Our results further demonstrated that TLR4-KO mice exhibit a different microbiota that can protect against the ethanol-induced activation of the immune system and colon integrity dysfunctions. In short, our results reveal that TLR4 is a key factor for determining the gut microbiota, which can participate in dysbiosis and the inflammatory response induced by alcohol consumption.
Collapse
Affiliation(s)
- Carlos M. Cuesta
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
| | - María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 15 Avda. Blasco Ibanez, 46010 Valencia, Spain
| | - Raúl Pérez-Moraga
- Bioinformatics and Biostatistics Unit, Prince Felipe Research Center, 46012 Valencia, Spain; (R.P.-M.); (F.G.-G.)
| | - Irene Rodríguez-Navarro
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
| | - Francisco García-García
- Bioinformatics and Biostatistics Unit, Prince Felipe Research Center, 46012 Valencia, Spain; (R.P.-M.); (F.G.-G.)
| | - Juan R. Ureña-Peralta
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
- Correspondence: (J.R.U.-P.); (C.G.)
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
- Correspondence: (J.R.U.-P.); (C.G.)
| |
Collapse
|
36
|
Silva-Gotay A, Davis J, Tavares ER, Richardson HN. Alcohol drinking during early adolescence activates microglial cells and increases frontolimbic Interleukin-1 beta and Toll-like receptor 4 gene expression, with heightened sensitivity in male rats compared to females. Neuropharmacology 2021; 197:108698. [PMID: 34252404 PMCID: PMC8552486 DOI: 10.1016/j.neuropharm.2021.108698] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/13/2021] [Accepted: 06/15/2021] [Indexed: 12/31/2022]
Abstract
Adolescent drinking is risky because neural circuits in the frontal lobes are undergoing maturational processes important for cognitive function and behavioral control in adulthood. Previous studies have shown that myelinated axons in the medial prefrontal cortex (mPFC) are particularly sensitive to alcohol drinking, especially in males. Pro-inflammatory mediators like toll-like receptor 4 (TLR4) and interleukin-1 beta (IL1b) have been implicated in alcohol induced-inflammation and demyelination; thus, herein we test the hypothesis that voluntary alcohol drinking early in adolescence elicits a pro-inflammatory state that is more pronounced in the brain of males compared to females. Adolescent male and female Wistar rats self-administered sweetened alcohol or sweetened water from postnatal days 28-42 and separate sets of brains were processed for 1) immunolabeling for ionized calcium-binding adapter molecule 1 to analyze microglial cell morphology, or 2) qPCR analysis of gene expression of pro-inflammatory mediators. Binge drinking alcohol activated microglia in the mPFC and hippocampus of both males and females, suggesting that voluntary alcohol exposure initiates an inflammatory response. Il1b mRNA was upregulated in the mPFC of both sexes. Conversely, Tlr4 mRNA levels were elevated after drinking only in males, which could explain more robust effects of alcohol on myelin in this region in developing males compared to females. Il1b mRNA changes were not observed in the hippocampus, but alcohol elevated Tlr4 mRNA in both sexes, highlighting regional specificity in inflammatory responses to alcohol. Overall, these findings give insight into potential mechanisms by which low-to-moderate voluntary alcohol intake impacts the developing brain. This article is part of the special Issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- Andrea Silva-Gotay
- Neuroscience and Behavior Graduate Program, University of
Massachusetts, Amherst, MA 01003
| | - Jillian Davis
- Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| | - Elizabeth R. Tavares
- Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| | - Heather N. Richardson
- Neuroscience and Behavior Graduate Program, University of
Massachusetts, Amherst, MA 01003,Department of Psychological and Brain Sciences, University
of Massachusetts, Amherst, MA 01003
| |
Collapse
|
37
|
Ilješ AP, Plesničar BK, Dolžan V. Associations of NLRP3 and CARD8 gene polymorphisms with alcohol dependence and commonly related psychiatric disorders: a preliminary study. Arh Hig Rada Toksikol 2021; 72:191-197. [PMID: 34587665 PMCID: PMC8576752 DOI: 10.2478/aiht-2021-72-3432] [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: 05/01/2020] [Revised: 05/01/2020] [Accepted: 09/01/2021] [Indexed: 12/11/2022] Open
Abstract
We investigated two functional polymorphisms in NLRP3 inflammasome genes (NLRP3 rs35829419 and CARD8 rs2043211) and their association with alcohol dependence and related anxiety, depression, obsession-compulsion, or aggression in 88 hospitalised alcohol-dependent patients, 99 abstinent alcohol-dependent participants, and 94 controls, all male Caucasian. Alcohol dependence-related psychiatric disorders were assessed with the Zung Depression and Anxiety scale, Buss-Durkee Hostility Inventory, Alcohol Use Disorders Identification Test, Brief Social Phobia Scale, Obsessive Compulsive Drinking Scale, and Yale-Brown Obsessive-Compulsive Scale. For genotyping we used the allele-specific quantitative polymerase chain reaction-based methods. The three groups differed significantly in CARD8 rs2043211 distribution (P=0.049; chi-squared=9.557; df=4). The NLPR3 rs35829419 polymorphism was not significantly associated with alcohol dependence. In hospitalised alcohol-dependent patients the investigated polymorphisms were not associated with scores indicating alcohol consumption or comorbid symptoms. In abstinent alcohol-dependent subjects homozygotes for the polymorphic CARD8 allele presented with the highest scores on the Zung Anxiety Scale (p=0.048; df=2; F=3.140). Among controls, CARD8 genotype was associated with high scores on the Obsessive Compulsive Drinking Scale (P=0.027; df=2; F=3.744). In conclusion, our results reveal that CARD8 rs2043211 may play some role in susceptibility to alcohol dependence, expression of anxiety symptoms in abstinent alcohol-dependent subjects, and in obsessive compulsive drinking in healthy controls. However, further studies with larger cohorts are required to confirm these preliminary findings.
Collapse
Affiliation(s)
| | - Blanka Kores Plesničar
- University Psychiatric Clinic, Ljubljana, Slovenia
- University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Vita Dolžan
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Pharmacogenetics Laboratory, Ljubljana, Slovenia
| |
Collapse
|
38
|
Ibáñez F, Montesinos J, Area-Gomez E, Guerri C, Pascual M. Ethanol Induces Extracellular Vesicle Secretion by Altering Lipid Metabolism through the Mitochondria-Associated ER Membranes and Sphingomyelinases. Int J Mol Sci 2021; 22:ijms22168438. [PMID: 34445139 PMCID: PMC8395151 DOI: 10.3390/ijms22168438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Recent evidence pinpoints extracellular vesicles (EVs) as key players in intercellular communication. Given the importance of cholesterol and sphingomyelin in EV biology, and the relevance of mitochondria-associated endoplasmic reticulum membranes (MAMs) in cholesterol/sphingomyelin homeostasis, we evaluated if MAMs and sphingomyelinases (SMases) could participate in ethanol-induced EV release. EVs were isolated from the extracellular medium of BV2 microglia treated or not with ethanol (50 and 100 mM). Radioactive metabolic tracers combined with thin layer chromatography were used as quantitative methods to assay phospholipid transfer, SMase activity and cholesterol uptake/esterification. Inhibitors of SMase (desipramine and GW4869) and MAM (cyclosporin A) activities were also utilized. Our data show that ethanol increases the secretion and inflammatory molecule concentration of EVs. Ethanol also upregulates MAM activity and alters lipid metabolism by increasing cholesterol uptake, cholesterol esterification and SMase activity in microglia. Notably, the inhibition of either SMase or MAM activity prevented the ethanol-induced increase in EV secretion. Collectively, these results strongly support a lipid-driven mechanism, specifically via SMases and MAM, to explain the effect of ethanol on EV secretion in glial cells.
Collapse
Affiliation(s)
- Francesc Ibáñez
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012 Valencia, Spain; (F.I.); (C.G.)
| | - Jorge Montesinos
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA;
- Correspondence: (J.M.); (M.P.); Tel.: +34-961-625-635 (M.P.); Fax: +34-963-864-642 (M.P.)
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012 Valencia, Spain; (F.I.); (C.G.)
| | - María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012 Valencia, Spain; (F.I.); (C.G.)
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Correspondence: (J.M.); (M.P.); Tel.: +34-961-625-635 (M.P.); Fax: +34-963-864-642 (M.P.)
| |
Collapse
|
39
|
Mahajan SD, Homish GG, Quisenberry A. Multifactorial Etiology of Adolescent Nicotine Addiction: A Review of the Neurobiology of Nicotine Addiction and Its Implications for Smoking Cessation Pharmacotherapy. Front Public Health 2021; 9:664748. [PMID: 34291026 PMCID: PMC8287334 DOI: 10.3389/fpubh.2021.664748] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Nicotine is the primary pharmacologic component of tobacco, and its highly addictive nature is responsible for its widespread use and significant withdrawal effects that result in challenges to smoking cessation therapeutics. Nicotine addiction often begins in adolescence and this is at least partially attributed to the fact that adolescent brain is most susceptible to the neuro-inflammatory effects of nicotine. There is increasing evidence for the involvement of microglial cells, which are the brain's primary homeostatic sensor, in drug dependence and its associated behavioral manifestations particularly in the adolescent brain. A hallmark of neuro-inflammation is microglial activation and activation of microglia by nicotine during adolescent development, which may result in long-term addiction to nicotine. This non-systematic review examines multifactorial etiology of adolescent nicotine addiction, neurobiology of nicotine addiction and the potential mechanisms that underlie the effects of nicotine on inflammatory signaling in the microglia, understanding how nicotine affects the adolescent brain. We speculate, that modulating homeostatic balance in microglia, could have promising therapeutic potential in withdrawal, tolerance, and abstinence-related neural adaptations in nicotine addiction, in the adolescent brain. Further, we discuss nicotine addiction in the context of the sensitization-homeostasis model which provides a theoretical framework for addressing the potential role of microglial homeostasis in neural adaptations underlying nicotine abuse.
Collapse
Affiliation(s)
- Supriya D. Mahajan
- Department of Community Health and Health Behavior, School of Public Health, University at Buffalo, Buffalo, NY, United States
| | - Gregory G. Homish
- Department of Community Health and Health Behavior, School of Public Health, University at Buffalo, Buffalo, NY, United States
| | - Amanda Quisenberry
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
40
|
Studies of involvement of G-protein coupled receptor-3 in cannabidiol effects on inflammatory responses of mouse primary astrocytes and microglia. PLoS One 2021; 16:e0251677. [PMID: 33984046 PMCID: PMC8118243 DOI: 10.1371/journal.pone.0251677] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/29/2021] [Indexed: 12/04/2022] Open
Abstract
Cannabidiol (CBD) exhibits anti-inflammatory and neuroprotective properties and is suggested to be effective in the pre-clinical and clinical treatment of illnesses of the central nervous system (CNS). Two major types of CNS glial cells, astrocytes and microglia, play critical roles in the development and pathogenesis of CNS diseases. However, the mechanisms by which CBD plays an anti-inflammatory and neuroprotective role for these glial cells have not been fully elucidated. In this study, we examined the effects of CBD on the inflammatory response of mouse primary astrocytes and microglia. We also investigated whether the effect of CBD on cytokine release is mediated by the G protein coupled receptor 3 (GPR3), which was recently identified as a novel receptor for CBD. Our results showed that CBD inhibited inflammatory responses of astrocytes and microglia stimulated with lipopolysaccharide (LPS), a Toll-like receptor 4 (TLR4) ligand in vitro and in vivo. In addition, CBD reduced the phosphorylation of STAT3 and NF-κB signaling pathways in LPS-stimulated astrocytes. However, the inhibitory effect of CBD on pro-inflammatory cytokine production was independent of GPR3 expression in both types of glial cells. Thus, although CBD is effective in ameliorating the activation of astrocytes and microglia, its mechanism of action still requires further study. Our data support the concept that CBD may have therapeutic potential for neurological disorders that involve neuroinflammation.
Collapse
|
41
|
Lee JS, O’Connell EM, Pacher P, Lohoff FW. PCSK9 and the Gut-Liver-Brain Axis: A Novel Therapeutic Target for Immune Regulation in Alcohol Use Disorder. J Clin Med 2021; 10:1758. [PMID: 33919550 PMCID: PMC8074019 DOI: 10.3390/jcm10081758] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Alcohol use disorder (AUD) is a chronic relapsing disorder characterized by an impaired ability to control or stop alcohol intake and is associated with organ damage including alcohol-associated liver disease (ALD) and progressive neurodegeneration. The etiology of AUD is complex, but organ injury due to chronic alcohol use can be partially attributed to systemic and local inflammation along the gut-liver-brain axis. Excessive alcohol use can result in translocation of bacterial products into circulation, increased expression of pro-inflammatory cytokines, and activation of immune cells, including macrophages and/or microglia in the liver and brain. One potential mediator of this alcohol-induced inflammation is proprotein convertase subtilisin/kexin type 9 (PCSK9). PCSK9 is primarily known for its regulation of plasma low-density lipoprotein cholesterol but has more recently been shown to influence inflammatory responses in the liver and brain. In rodent and post-mortem brain studies, chronic alcohol use altered methylation of the PCSK9 gene and increased expression of PCSK9 in the liver and cerebral spinal fluid. Additionally, PCSK9 inhibition in a rat model of ALD attenuated liver inflammation and steatosis. PCSK9 may play an important role in alcohol-induced pathologies along the gut-liver-brain axis and may be a novel therapeutic target for AUD-related liver and brain inflammation.
Collapse
Affiliation(s)
- Ji Soo Lee
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (J.S.L.)
| | - Emma M. O’Connell
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (J.S.L.)
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20852, USA;
| | - Falk W. Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (J.S.L.)
| |
Collapse
|
42
|
Aryal SP, Fu X, Sandin JN, Neupane KR, Lakes JE, Grady ME, Richards CI. Nicotine induces morphological and functional changes in astrocytes via nicotinic receptor activity. Glia 2021; 69:2037-2053. [PMID: 33851731 DOI: 10.1002/glia.24011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 01/16/2023]
Abstract
Nicotine is a highly addictive compound present in tobacco, which causes the release of dopamine in different regions of the brain. Recent studies have shown that astrocytes express nicotinic acetylcholine receptors (nAChRs) and mediate calcium signaling. In this study, we examine the morphological and functional adaptations of astrocytes due to nicotine exposure. Utilizing a combination of fluorescence and atomic force microscopy, we show that nicotine-treated astrocytes exhibit time-dependent remodeling in the number and length of both proximal and fine processes. Blocking nAChR activity with an antagonist completely abolishes nicotine's influence on astrocyte morphology indicating that nicotine's action is mediated by these receptors. Functional studies show that 24-hr nicotine treatment induces higher levels of calcium activity in both the cell soma and the processes with a more substantial change observed in the processes. Nicotine does not induce reactive astrocytosis even at high concentrations (10 μM) as determined by cytokine release and glial fibrillary acidic protein expression. We designed tissue clearing experiments to test whether morphological changes occur in vivo using astrocyte specific Aldh1l1-tdTomato knock in mice. We find that nicotine induces a change in the volume of astrocytes in the prefrontal cortex, CA1 of the hippocampus, and the substantia nigra. These results indicate that nicotine directly alters the functional and morphological properties of astrocytes potentially contributing to the underlying mechanism of nicotine abuse.
Collapse
Affiliation(s)
- Surya P Aryal
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Xu Fu
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Joree N Sandin
- Department of Mechanical Engineering, University of Kentucky, Lexington, Kentucky, USA
| | - Khaga R Neupane
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Jourdan E Lakes
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Martha E Grady
- Department of Mechanical Engineering, University of Kentucky, Lexington, Kentucky, USA
| | | |
Collapse
|
43
|
Gupta H, Suk KT, Kim DJ. Gut Microbiota at the Intersection of Alcohol, Brain, and the Liver. J Clin Med 2021; 10:541. [PMID: 33540624 PMCID: PMC7867253 DOI: 10.3390/jcm10030541] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Over the last decade, increased research into the cognizance of the gut-liver-brain axis in medicine has yielded powerful evidence suggesting a strong association between alcoholic liver diseases (ALD) and the brain, including hepatic encephalopathy or other similar brain disorders. In the gut-brain axis, chronic, alcohol-drinking-induced, low-grade systemic inflammation is suggested to be the main pathophysiology of cognitive dysfunctions in patients with ALD. However, the role of gut microbiota and its metabolites have remained unclear. Eubiosis of the gut microbiome is crucial as dysbiosis between autochthonous bacteria and pathobionts leads to intestinal insult, liver injury, and neuroinflammation. Restoring dysbiosis using modulating factors such as alcohol abstinence, promoting commensal bacterial abundance, maintaining short-chain fatty acids in the gut, or vagus nerve stimulation could be beneficial in alleviating disease progression. In this review, we summarize the pathogenic mechanisms linked with the gut-liver-brain axis in the development and progression of brain disorders associated with ALD in both experimental models and humans. Further, we discuss the therapeutic potential and future research directions as they relate to the gut-liver-brain axis.
Collapse
Affiliation(s)
| | | | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24253, Korea; (H.G.); (K.T.S.)
| |
Collapse
|
44
|
Peng H, Nixon K. Microglia Phenotypes Following the Induction of Alcohol Dependence in Adolescent Rats. Alcohol Clin Exp Res 2021; 45:105-116. [PMID: 33164228 PMCID: PMC8296648 DOI: 10.1111/acer.14504] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Activation of the innate immune system may play a role in the development of alcohol use disorders (AUDs), which often originate with adolescent alcohol abuse. A key player in the innate immune system is microglia, the activation of which occurs along a spectrum from proinflammatory, or M1-like, to anti-inflammatory, or M2-like, phenotypes. METHODS Adolescent, male rats were gavaged with ethanol (EtOH) or isocaloric control diet every 8 hours for 4 days and then sacrificed at 0, 2, 7, and 14 days later. Microglia were isolated from the entorhinal cortex and hippocampus by Percoll gradient centrifugation, labeled with surface antigens for activation, and analyzed by flow cytometry. Polarization states of microglia, defined as CD11b+ CD45low cells, were determined by the expression of M1 surface markers, major histocompatibility complex (MHC) II, CD32, and CD86, and M2 surface marker, CD206 (mannose receptor). Cytokine gene expression was measured by reverse transcriptase polymerase chain reaction. RESULTS Isolated cells were a highly enriched population (>95% pure) of microglia/macrophages according to CD11b immunoreactivity. EtOH rats showed the most dramatic increases in microglia activation markers CD11b and CD45, and M1 (MHC-II) and M2 (CD206) markers at T2, when additional M1 markers CD86 and CD32 were also increased. Surprisingly, proinflammatory gene expression of CCL2, IL-1β, IL-6, and TNF-α generally was decreased at all time points in EtOH rats except for IL-6 which was increased at T0 and TNF-α which was not changed at T0 in either region. Simultaneously, BDNF expression was increased at T2 and T7, while IGF1 and TGF-β gene expression was decreased. Arginase was also increased at T0 in hippocampus, but not changed by alcohol otherwise. CONCLUSIONS These data show that microglia phenotype after alcohol dependence is not a simple M1 or M2 classification, though more indicators of an anti-inflammatory phenotype were observed. Determining microglia phenotype is critical for understanding their role in the development of AUDs.
Collapse
Affiliation(s)
- Hui Peng
- University of Kentucky, College of Pharmacy, Department of Pharmaceutical Sciences Lexington, KY 40536, USA
| | - Kimberly Nixon
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX USA
| |
Collapse
|
45
|
Cortez I, Rodgers SP, Kosten TA, Leasure JL. Sex and Age Effects on Neurobehavioral Toxicity Induced by Binge Alcohol. Brain Plast 2020; 6:5-25. [PMID: 33680843 PMCID: PMC7902983 DOI: 10.3233/bpl-190094] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Historically, most alcohol neurotoxicity studies were conducted in young adult males and focused on chronic intake. There has been a shift towards studying the effects of alcohol on the adolescent brain, due to alcohol consumption during this formative period disrupting the brain's developmental trajectory. Because the most typical pattern of adolescent alcohol intake is heavy episodic (binge) drinking, there has also been a shift towards the study of binge alcohol-induced neurobehavioral toxicity. It has thus become apparent that binge alcohol damages the adolescent brain and there is increasing attention to sex-dependent effects. Significant knowledge gaps remain in our understanding of the effects of binge alcohol on the female brain, however. Moreover, it is unsettling that population-level studies indicate that the prevalence of binge drinking is increasing among American women, particularly those in older age groups. Although study of adolescents has made it apparent that binge alcohol disrupts ongoing brain maturational processes, we know almost nothing about how it impacts the aging brain, as studies of its effects on the aged brain are relatively scarce, and the study of sex-dependent effects is just beginning. Given the rapidly increasing population of older Americans, it is crucial that studies address age-dependent effects of binge alcohol, and given the increase in binge drinking in older women who are at higher risk for cognitive decline relative to men, studies must encompass both sexes. Because adolescence and older age are both characterized by age-typical brain changes, and because binge drinking is the most common pattern of alcohol intake in both age groups, the knowledge that we have amassed on binge alcohol effects on the adolescent brain can inform our study of its effects on the aging brain. In this review, we therefore cover the current state of knowledge of sex and age-dependent effects of binge alcohol, as well as statistical and methodological considerations for studies aimed at addressing them.
Collapse
Affiliation(s)
- Ibdanelo Cortez
- Department of Psychology, University of Houston, Houston, TX, USA
| | | | | | - J. Leigh Leasure
- Department of Psychology, University of Houston, Houston, TX, USA
- Department of Biology & Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
46
|
Wu R, Li JX. Toll-Like Receptor 4 Signaling and Drug Addiction. Front Pharmacol 2020; 11:603445. [PMID: 33424612 PMCID: PMC7793839 DOI: 10.3389/fphar.2020.603445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
The emphasis of neuronal alterations and adaptations have long been the main focus of the studies of the mechanistic underpinnings of drug addiction. Recent studies have begun to appreciate the role of innate immune system, especially toll-like receptor 4 (TLR4) signaling in drug reward-associated behaviors and physiology. Drugs like opioids, alcohol and psychostimulants activate TLR4 signaling and subsequently induce proinflammatory responses, which in turn contributes to the development of drug addiction. Inhibition of TLR4 or its downstream effectors attenuated the reinforcing effects of opioids, alcohol and psychostimulants, and this effect is also involved in the withdrawal and relapse-like behaviors of different drug classes. However, conflicting results also argue that TLR4-related immune response may play a minimal part in drug addiction. This review discussed the preclinical evidence that whether TLR4 signaling is involved in multiple drug classes action and the possible mechanisms underlying this effect. Moreover, clinical studies which examined the potential efficacy of immune-base pharmacotherapies in treating drug addiction are also discussed.
Collapse
Affiliation(s)
- Ruyan Wu
- School of Medicine, Yangzhou University, Yangzhou, China.,Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
47
|
Lua YH, Ong WW, Wong HK, Chew CH. Ethanol-induced CYP2E1 Expression is Reduced by Lauric Acid via PI3K Pathway in HepG2 Cells. Trop Life Sci Res 2020; 31:63-75. [PMID: 33214856 PMCID: PMC7652244 DOI: 10.21315/tlsr2020.31.3.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The metabolism of alcohol involves cytochrome P450 2E1 (CYP2E1)-induced oxidative stress, with the association of phosphatidylinositol-3-kinases (PI3K) and nuclear factor kappa B (NFκB) signalling pathways. CYP2E1 is primarily involved in the microsomal ethanol oxidising system, which generates massive reactive oxygen species (ROS) and ultimately leads to oxidative stress and tissue damage. Lauric acid, a major fatty acid in palm kernel oil, has been shown as a potential antioxidant. Here, we aimed to evaluate the use of lauric acid as a potential antioxidant against ethanol-mediated oxidative stress by investigating its effect on CYP2E1 mRNA expression and the signalling pathway in ethanol-induced HepG2 cells. HepG2 cells were firstly treated with different concentrations of ethanol, and subsequently co-treated with different concentrations of lauric acid for 24 h. Total cellular RNA and total protein were extracted, and qPCR and Western blot was carried out. Ethanol induced the mRNA expression of CYP2E1 significantly, but lauric acid was able to downregulate the induced CYP2E1 expression in a dose-dependent manner. Similarly, Western blot analysis and densitometry analysis showed that the phosphorylated PI3K p85 (Tyr458) protein was significantly elevated in ethanol-treated HepG2 cells, but co-treatment with lauric acid repressed the activation of PI3K. However, there was no significant difference in NFκB pathway, in which the normalised NFκB p105 (Ser933) phosphorylation remained constant in any treatment conditions in this study. This suggests that ethanol induced CYP2E1 expression by activating PI3K p85 (Tyr458) pathway, but not the NFκB p105 (Ser933) pathway in HepG2 cells.
Collapse
Affiliation(s)
- Ying-Huan Lua
- Department of Allied Health Sciences, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Wei-Wah Ong
- Department of Allied Health Sciences, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Hong-Kin Wong
- Department of Allied Health Sciences, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Choy-Hoong Chew
- Department of Allied Health Sciences, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| |
Collapse
|
48
|
Kane CJM, Drew PD. Neuroinflammatory contribution of microglia and astrocytes in fetal alcohol spectrum disorders. J Neurosci Res 2020; 99:1973-1985. [PMID: 32959429 DOI: 10.1002/jnr.24735] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/03/2020] [Accepted: 09/05/2020] [Indexed: 12/12/2022]
Abstract
Ethanol exposure to the fetus during pregnancy can result in fetal alcohol spectrum disorders (FASD). These disorders vary in severity, can affect multiple organ systems, and can lead to lifelong disabilities. Damage to the central nervous system (CNS) is common in FASD, and can result in altered behavior and cognition. The incidence of FASD is alarmingly high, resulting in significant personal and societal costs. There are no cures for FASD. Alcohol can directly alter the function of neurons in the developing CNS. In addition, ethanol can alter the function of CNS glial cells including microglia and astrocytes which normally maintain homeostasis in the CNS. These glial cells can function as resident immune cells in the CNS to protect against pathogens and other insults. However, activation of glia can also damage CNS cells and lead to aberrant CNS function. Ethanol exposure to the developing brain can result in the activation of glia and neuroinflammation, which may contribute to the pathology associated with FASD. This suggests that anti-inflammatory agents may be effective in the treatment of FASD.
Collapse
Affiliation(s)
- Cynthia J M Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul D Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
49
|
Socodato R, Henriques JF, Portugal CC, Almeida TO, Tedim-Moreira J, Alves RL, Canedo T, Silva C, Magalhães A, Summavielle T, Relvas JB. Daily alcohol intake triggers aberrant synaptic pruning leading to synapse loss and anxiety-like behavior. Sci Signal 2020; 13:13/650/eaba5754. [PMID: 32963013 DOI: 10.1126/scisignal.aba5754] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alcohol abuse adversely affects the lives of millions of people worldwide. Deficits in synaptic transmission and in microglial function are commonly found in human alcohol abusers and in animal models of alcohol intoxication. Here, we found that a protocol simulating chronic binge drinking in male mice resulted in aberrant synaptic pruning and substantial loss of excitatory synapses in the prefrontal cortex, which resulted in increased anxiety-like behavior. Mechanistically, alcohol intake increased the engulfment capacity of microglia in a manner dependent on the kinase Src, the subsequent activation of the transcription factor NF-κB, and the consequent production of the proinflammatory cytokine TNF. Pharmacological blockade of Src activation or of TNF production in microglia, genetic ablation of Tnf, or conditional ablation of microglia attenuated aberrant synaptic pruning, thereby preventing the neuronal and behavioral effects of the alcohol. Our data suggest that aberrant pruning of excitatory synapses by microglia may disrupt synaptic transmission in response to alcohol abuse.
Collapse
Affiliation(s)
- Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Joana F Henriques
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Tiago O Almeida
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Tedim-Moreira
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Renata L Alves
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Teresa Canedo
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.,Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Cátia Silva
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Teresa Summavielle
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal. .,Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
50
|
Ureña-Peralta JR, Pérez-Moraga R, García-García F, Guerri C. Lack of TLR4 modifies the miRNAs profile and attenuates inflammatory signaling pathways. PLoS One 2020; 15:e0237066. [PMID: 32780740 PMCID: PMC7418977 DOI: 10.1371/journal.pone.0237066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 07/19/2020] [Indexed: 12/12/2022] Open
Abstract
TLR4 is a member of the toll-like receptors (TLR) immune family, which are activated by lipopolysaccharide, ethanol or damaged tissue, among others, by triggering proinflammatory cytokines release and inflammation. Lack of TLR4 protects against inflammatory processes and neuroinflammation linked with several neuropathologies. By considering that miRNAs are key post-transcriptional regulators of the proteins involved in distinct cellular processes, including inflammation, this study aimed to assess the impact of the miRNAs profile in mice cortices lacking the TLR4 response. Using mice cerebral cortices and next-generation sequencing (NGS), the findings showed that lack of TLR4 significantly reduced the quantity and diversity of the miRNAs expressed in WT mice cortices. The results also revealed a significant down-regulation of the miR-200 family, while cluster miR-99b/let-7e/miR-125a was up-regulated in TLR4-KO vs. WT. The bioinformatics and functional analyses demonstrated that TLR4-KO presented the systematic depletion of many pathways closely related to the immune system response, such as cytokine and interleukin signaling, MAPK and ion Channels routes, MyD88 pathways, NF-κβ and TLR7/8 pathways. Our results provide new insights into the molecular and biological processes associated with the protective effects of TLR-KO against inflammatory damage and neuroinflammation, and reveal the relevance of the TLR4 receptors response in many neuropathologies.
Collapse
Affiliation(s)
- Juan R. Ureña-Peralta
- Molecular and cellular pathology of Alcohol Laboratory, Prince Felipe Research Center, Valencia, Spain
| | - Raúl Pérez-Moraga
- Bioinformatics & Biostatistics Unit, Prince Felipe Research Center, Valencia, Spain
- Biomedical Imaging Unit FISABIO-CIPF, Prince Felipe Research Center, Valencia, Spain
| | | | - Consuelo Guerri
- Molecular and cellular pathology of Alcohol Laboratory, Prince Felipe Research Center, Valencia, Spain
- * E-mail:
| |
Collapse
|