1
|
Wildermuth E, Patton MS, Cortes-Gutierrez M, Jinwala Z, Grissom BH, Campbell RR, Kranzler HR, Lobo MK, Ament SA, Mathur BN. A single-cell genomic atlas for the effects of chronic ethanol exposure in the mouse dorsal striatum. Mol Psychiatry 2025:10.1038/s41380-025-03014-z. [PMID: 40240618 DOI: 10.1038/s41380-025-03014-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/22/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Alcohol use disorder (AUD) is characterized by compulsive drinking, which is thought to be mediated by effects of chronic intermittent ethanol exposure on the dorsal striatum, the input nucleus of the basal ganglia. Despite significant efforts to understand the impact of ethanol on the dorsal striatum, the rich diversity of striatal cell types and multitude of ethanol targets expressed by them necessitates an unbiased, discovery-based approach. In this study, we used single-nuclei RNA-sequencing (snRNA-seq; n = 86,715 cells) to examine the impact of chronic intermittent ethanol exposure on the dorsal striatum in C57BL/6 male and female mice. We detected 462 differentially expressed genes at FDR < 0.05, the majority of which were mapped to spiny projection neurons (SPNs), the most prominent cell type in the striatum. Gene co-expression network analysis and functional annotation of differentially expressed genes revealed down-regulation of postsynaptic intracellular signaling cascades in SPNs. Inflammation-related genes were down-regulated across many neuronal and non-neuronal cell types. Gene set enrichment analyses also pointed to altered states of rare cell types, including the induction of angiogenesis-related genes in vascular cells. A gene module down-regulated specifically in canonical SPNs was enriched for calcium-signaling genes and components of glutamatergic synapses, as well as for genes associated with genetic risk for AUD. Genetic perturbations of six of this module's hub genes - Foxp1, Bcl11b, Pde10a, Rarb, Rgs9, and Itgr1 - had causal effects on its expression in the mouse striatum and/or on the broader set of differentially expressed genes in alcohol-exposed mice. These data provide important clues as to the impact of ethanol on striatal biology and provide a key resource for future investigation.
Collapse
Affiliation(s)
- Erin Wildermuth
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Medical Scientist Training Program, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael S Patton
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcia Cortes-Gutierrez
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zeal Jinwala
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
| | - Benjamin H Grissom
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rianne R Campbell
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Henry R Kranzler
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
| | - Mary Kay Lobo
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Seth A Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Brian N Mathur
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Kahlert Institute for Addiction Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Bell KT, Hughes JM, Borman WA, Stoffel RD, Canfield SG. Alcohol diminishes barrier integrity in human stem cell-derived brain microvascular endothelial cells: Role of reactive oxygen species. Alcohol 2025; 125:55-66. [PMID: 40187584 DOI: 10.1016/j.alcohol.2025.03.005] [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: 12/18/2024] [Revised: 01/31/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
The World Health Organization has linked alcohol consumption to over 200 diseases including neurodegenerative diseases. A dysfunctional blood-brain barrier (BBB) has been found to be influential in a number of brain disorders. The BBB is critical in maintaining homeostasis between the brain vasculature and parenchyma and a loss in barrier integrity would enable otherwise impermeable immune cells, molecules, and inflammatory mediators to reach the brain parenchyma. A subset of studies demonstrated that alcohol could diminish BBB integrity, but it is unclear if this effect translates clinically. In this study, we utilize a human stem cell-derived BBB model with near in vivo properties to investigate the effects of alcohol on critical barrier properties. Barrier forming brain-like microvascular endothelial cells (BMECs) were derived from human induced pluripotent stem cells (iPSCs) and exposed to several alcohol concentrations. Alcohol decreased barrier integrity observed by a loss in trans-endothelial electrical resistance and an increase in sodium fluorescein permeability. Alcohol decreased expression and junctional localization of tight junction proteins, a critical component to barrier integrity. Additionally, alcohol did not affect efflux transporter activity or cell viability in BMECs. The detrimental effects of alcohol on BBB properties were due to in part elevated reactive oxygen species (ROS); as scavenging ROS improved barrier properties, including the restoration of tight junction expression and localization. These data suggest that excessive alcohol consumption could diminish the BBB and contribute to the development or exacerbation of brain disorders. CLINICAL TRIAL NUMBER AND REGISTRY URL: Not applicable.
Collapse
Affiliation(s)
- Kameron T Bell
- Indiana University School of Medicine, Terre Haute, IN, USA; Indiana State University, Terre Haute, IN, USA
| | - Jason M Hughes
- Indiana University School of Medicine, Terre Haute, IN, USA
| | | | - Ryan D Stoffel
- Indiana University School of Medicine, Terre Haute, IN, USA
| | | |
Collapse
|
3
|
Tong M, Homans C, Pelit W, Delikkaya B, de la Monte SM. Progressive Alcohol-Related Brain Atrophy and White Matter Pathology Are Linked to Long-Term Inhibitory Effects on mTOR Signaling. Biomolecules 2025; 15:413. [PMID: 40149949 PMCID: PMC11940526 DOI: 10.3390/biom15030413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Alcohol-related brain damage (ARBD) causes cognitive-behavioral impairments that can lead to dementia. White matter is a major target in ARBD. Additional research is needed to better understand the mechanisms of ARBD progression to advanced stages with permanent disability. Potential contributing factors include neuroinflammation and altered signaling through pathways that regulate cell survival, neuronal plasticity, myelin maintenance, and energy metabolism. OBJECTIVES This study characterizes the time course-related effects of chronic heavy ethanol feeding on white matter myelin protein expression, neuroinflammation, and molecules that mediate signaling through the mechanistic target of rapamycin (mTOR) pathways. METHODS Adult Long Evans rats (8-12/group) were fed with isocaloric liquid diets containing 0% (control) or 36% ethanol. Experimental endpoints spanned from 1 day to 8 weeks. The frontal lobes were used for histopathology and molecular and biochemical analyses. RESULTS Chronic ethanol feeding caused significant brain atrophy that was detected within 4 weeks and sustained over the course of the study. Early exposure time points, i.e., 2 weeks or less, were associated with global increases in the expression of non-myelinating, myelinating, and astrocyte markers, whereas at 6 or 8 weeks, white matter oligodendrocyte/myelin/glial protein expression was reduced. These effects were not associated with shifts in neuroinflammatory markers. Instead, the early stages of ARBD were accompanied by increases in several mTOR proteins and phosphoproteins, while later phases were marked by inhibition of downstream mTOR signaling through P70S6K. CONCLUSIONS Short-term versus long-term ethanol exposures differentially altered white matter glial protein expression and signaling through mTOR's downstream mediators that have known roles in myelin maintenance. These findings suggest that strategic targeting of mTOR signaling dysregulation may be critical for maintaining the functional integrity of white matter and ultimately preventing long-term ARBD-related cognitive impairment.
Collapse
Affiliation(s)
- Ming Tong
- Department of Medicine, Rhode Island Hospital, Brown University Health, and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Camilla Homans
- Molecular Pharmacology, Physiology, and Biotechnology Graduate Program, Brown University, Providence, RI 02903, USA
| | - William Pelit
- Department of Chemistry, Brown University, Providence, RI 02903, USA
| | - Busra Delikkaya
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Brown University Health, The Providence VA Medical Center, and the Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Suzanne M. de la Monte
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Brown University Health, The Providence VA Medical Center, and the Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
- Departments of Neurosurgery and Neurology, Rhode Island Hospital, Brown University Health, and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
4
|
Terracina S, Caronti B, Lucarelli M, Francati S, Piccioni MG, Tarani L, Ceccanti M, Caserta M, Verdone L, Venditti S, Fiore M, Ferraguti G. Alcohol Consumption and Autoimmune Diseases. Int J Mol Sci 2025; 26:845. [PMID: 39859557 PMCID: PMC11766456 DOI: 10.3390/ijms26020845] [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: 11/26/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Alcohol is the second-most misused substance after tobacco. It has been identified as a causal factor in more than 200 diseases and 5.3% of all deaths and is associated with significant behavioral, social, and economic difficulties. As alcohol consumption may modulate the immune system's regulatory mechanisms to avoid attacking the body's tissues, it has been proven to play a dichotomic role in autoimmune diseases (ADs) based on the quantity of consumption. In this review, we report updated evidence on the role of alcohol in ADs, with a focus on alcohol addiction and the human biological immune system and the relationship between them, with alcohol as a risk or protective factor. Then, in this narrative review, we report the main evidence on the most studied ADs where alcohol represents a key modulator, including autoimmune thyroiditis, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, diabetes, allergic rhinitis, and primary biliary cholangitis. Alcohol at low-moderate dosages seems mostly to have a protective role in these diseases, while at higher dosages, the collateral risks surpass possible benefits. The specific mechanisms by which low-to-moderate alcohol intake relieves AD symptoms are not yet fully understood; however, emerging studies suggest that alcohol may have a systemic immunomodulatory effect, potentially altering the balance of anti-inflammatory innate and adaptive immune cells, as well as cytokines (via the NF-κB or NLRP3 pathways). It might influence the composition of the gut microbiome (increasing amounts of beneficial gut microbes) and the production of their fatty acid metabolites, such as short-chain fatty acids (SCFAs) and polyunsaturated fatty acids (PUFAs), as well as elevated concentrations of acetate, high-density lipoprotein (HDL), and nitric oxide (NO). Unfortunately, a definite acceptable daily intake (ADI) of ethanol is complicated to establish because of the many mechanisms associated with alcohol consumption such that despite the interesting content of these findings, there is a limit to their applicability and risks should be weighed in cases of alcoholic drinking recommendations. The aim of future studies should be to modulate those beneficial pathways involved in the alcohol-protective role of ADs with various strategies to avoid the risks associated with alcohol intake.
Collapse
Affiliation(s)
- Sergio Terracina
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (S.T.); (M.L.); (S.F.)
| | - Brunella Caronti
- Department of Human Neurosciences, Sapienza University Hospital of Rome, 00185 Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (S.T.); (M.L.); (S.F.)
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy
| | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (S.T.); (M.L.); (S.F.)
| | - Maria Grazia Piccioni
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (M.G.P.); (L.T.)
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (M.G.P.); (L.T.)
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell’Alcolismo e le sue Complicanze, 00185 Rome, Italy;
| | - Micaela Caserta
- Institute of Molecular Biology and Pathology (IBPM-CNR), 00161 Rome, Italy; (M.C.); (L.V.)
| | - Loredana Verdone
- Institute of Molecular Biology and Pathology (IBPM-CNR), 00161 Rome, Italy; (M.C.); (L.V.)
| | - Sabrina Venditti
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00161 Rome, Italy;
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), c/o Department of Sensory Organs, Sapienza University of Rome, 00161 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (S.T.); (M.L.); (S.F.)
| |
Collapse
|
5
|
Friske MM, Torrico EC, Haas MJW, Borruto AM, Giannone F, Hade AC, Yu Y, Gao L, Sutherland GT, Hitzemann R, Philips MA, Fei SS, Sommer WH, Mayfield RD, Spanagel R. A systematic review and meta-analysis on the transcriptomic signatures in alcohol use disorder. Mol Psychiatry 2025; 30:310-326. [PMID: 39242950 PMCID: PMC11649567 DOI: 10.1038/s41380-024-02719-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
Currently available clinical treatments on alcohol use disorder (AUD) exhibit limited efficacy and new druggable targets are required. One promising approach to discover new molecular treatment targets involves the transcriptomic profiling of brain regions within the addiction neurocircuitry, utilizing animal models and postmortem brain tissue from deceased patients with AUD. Unfortunately, such studies suffer from large heterogeneity and small sample sizes. To address these limitations, we conducted a cross-species meta-analysis on transcriptome-wide data obtained from brain tissue of patients with AUD and animal models. We integrated 36 cross-species transcriptome-wide RNA-expression datasets with an alcohol-dependent phenotype vs. controls, following the PRISMA guidelines. In total, we meta-analyzed 964 samples - 502 samples from the prefrontal cortex (PFC), 282 nucleus accumbens (NAc) samples, and 180 from amygdala (AMY). The PFC had the highest number of differentially expressed genes (DEGs) across rodents, monkeys, and humans. Commonly dysregulated DEGs suggest conserved cross-species mechanisms for chronic alcohol consumption/AUD comprising MAPKs as well as STAT, IRF7, and TNF. Furthermore, we identified numerous unique gene sets that might contribute individually to these conserved mechanisms and also suggest novel molecular aspects of AUD. Validation of the transcriptomic alterations on the protein level revealed interesting targets for further investigation. Finally, we identified a combination of DEGs that are commonly regulated across different brain tissues as potential biomarkers for AUD. In summary, we provide a compendium of genes that are assessable via a shiny app, and describe signaling pathways, and physiological and cellular processes that are altered in AUD that require future studies for functional validation.
Collapse
Affiliation(s)
- Marion M Friske
- Institute of Psychopharmacology, Central Institute of Mental Health, Mannheim, University of Heidelberg, Heidelberg, Germany.
- Waggoner Center for Alcohol and Addiction Research and the Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA.
| | - Eva C Torrico
- Institute of Psychopharmacology, Central Institute of Mental Health, Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Maximilian J W Haas
- Institute of Psychopharmacology, Central Institute of Mental Health, Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Anna M Borruto
- Institute of Psychopharmacology, Central Institute of Mental Health, Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Francesco Giannone
- Institute of Psychopharmacology, Central Institute of Mental Health, Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Andreas-Christian Hade
- Department of Pathological Anatomy and Forensic Medicine, University of Tartu, Tartu, Estonia
- Forensic Medical Examination Department, Estonian Forensic Science Institute, Tallinn, Estonia
| | - Yun Yu
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Oregon Health & Science University West Campus, Portland, OR, USA
| | - Lina Gao
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Oregon Health & Science University West Campus, Portland, OR, USA
| | - Greg T Sutherland
- New South Wales Tissue Resource Center, University of Sydney, Camperdown, NSW, Australia
| | - Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Suzanne S Fei
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Oregon Health & Science University West Campus, Portland, OR, USA
| | - Wolfgang H Sommer
- Bethania Hospital for Psychiatry, Psychosomatics and Psychotherapy, Greifswald, Germany
- German Center for Mental Health (DZPG), Partner Site Mannheim-Heidelberg-Ulm, Mannheim, Germany
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research and the Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Mannheim, University of Heidelberg, Heidelberg, Germany.
- German Center for Mental Health (DZPG), Partner Site Mannheim-Heidelberg-Ulm, Mannheim, Germany.
| |
Collapse
|
6
|
Lubau NSA, Chengebroyen N, Subramaniyan V. Investigation of Uncovering Molecular Mechanisms of Alcohol-Induced Female Infertility-A Rational Approach. Reprod Sci 2024; 31:3660-3672. [PMID: 39485609 PMCID: PMC11611948 DOI: 10.1007/s43032-024-01692-8] [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: 04/11/2024] [Accepted: 09/06/2024] [Indexed: 11/03/2024]
Abstract
This study aimed to investigate the molecular mechanisms by which chronic alcohol consumption impacts female infertility, highlighting significant societal implications. By conducting a comprehensive literature review, we examined existing evidence on the association between long-term alcohol use and female reproductive health. Relevant studies were identified through systematic searches of electronic databases and key journals. We synthesized information on the molecular pathways affected by alcohol consumption, with particular emphasis on oxidative stress, inflammation, and hormonal disruptions. Additionally, we reviewed efforts to address alcohol-related health issues, including public health interventions, regulatory measures, and educational initiatives. Our study found strong evidence linking chronic alcohol consumption to increased mortality rates and a range of preventable diseases globally. Alcohol's effects extend beyond physiological consequences to psychological, social, and economic burdens. Chronic alcohol consumption disrupts hormonal balance and reproductive function, contributing to female infertility. Future research should focus on quantifying mortality risks associated with alcohol consumption, understanding gender-specific patterns in alcohol-related health outcomes, and elucidating the molecular mechanisms underlying female infertility. Addressing these gaps will inform strategies to mitigate the burden of alcohol-induced health issues and promote overall well-being. Collaborative efforts among diverse stakeholders are essential for advancing research agendas and translating findings into effective interventions.
Collapse
Affiliation(s)
- Natasha Sura Anak Lubau
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Neevashini Chengebroyen
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
7
|
Li Y, Yang J, Guo L. Role and mechanism of Lactobacillus casei in the modulation of alcohol preference in mice. Int Immunopharmacol 2024; 141:112902. [PMID: 39178519 DOI: 10.1016/j.intimp.2024.112902] [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: 04/18/2024] [Revised: 07/16/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Prolonged alcohol consumption may lead to gastrointestinal tract dysfunction and cause abnormalities in the associated nervous system activity, thereby increasing the body's craving for alcohol. Lactobacillus casei is a probiotic that has been shown to reduce the incidence of alcohol-related diseases. However, it is unclear whether Lactobacillus casei can delay the development of alcohol dependence. METHODS The chronic intermittent active drinking method was used to establish a mouse alcohol dependence model. The mice were randomly divided into 4 treatment groups, as follows: (1) Control group: two bottles of distilled water alternately, 0.2 mL/d saline gavage. (2) Alcohol group: alternating water and alcohol, 0.2 mL/d saline gavage. (3) Low group: alternating water and alcohol, 0.2 mL/d 1 × 108CFU of Lactobacillus casei by gavage. (4) High group: alternating water and alcohol, 0.2 mL/d 1 × 109CFU of Lactobacillus casei by gavage. The daily water consumption (mL), alcohol consumption (mL) and body weight of each mouse were recorded. After that, pathological changes in the intestines, brain tissues and serum of the experimental animals were detected, while changes in the intestinal flora of the mice were analysed by 16S rRNA sequencing. RESULTS The Lactobacillus casei intervention did not produce a significant effect on body weight in alcohol-exposed mice (P>0.05), but significantly reduced alcohol preference in alcohol-exposed mice (P<0.05). Subsequent analyses showed that Lactobacillus casei significantly ameliorated intestinal, brain tissue, and systemic inflammatory responses in alcohol-exposed mice (P<0.05). 16S rRNA sequencing showed that alcohol-exposed mice treated with Lactobacillus casei exhibited a richer composition of intestinal microorganisms, such as f__Rikenellaceae, g__Alistipes_A_871400, and g__Bacteroides_H genera showed relative enrichment in the High group. CONCLUSION By showing that Lactobacillus casei slows down alcohol preference and alleviates gut and brain tissue inflammation in alcohol-exposed mice, our findings provide a possible strategy: Lactobacillus casei may be able to serve as a potential target for the prevention and treatment of alcohol dependence.
Collapse
Affiliation(s)
- Yangchun Li
- Mudanjiang Medical University, Mudanjiang, China
| | - Jinyue Yang
- Mudanjiang Medical University, Mudanjiang, China
| | - Lishuang Guo
- Mudanjiang Medical University, Mudanjiang, China.
| |
Collapse
|
8
|
Yalcin EB, Tong M, Delikkaya B, Pelit W, Yang Y, de la Monte SM. Differential effects of moderate chronic ethanol consumption on neurobehavior, white matter glial protein expression, and mTOR pathway signaling with adolescent brain maturation. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2024; 50:492-516. [PMID: 38847790 PMCID: PMC11824867 DOI: 10.1080/00952990.2024.2355540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 09/06/2024]
Abstract
Background: Adolescent brains are highly vulnerable to heavy alcohol exposure. Increased understanding of how alcohol adversely impacts brain maturation may improve treatment outcomes.Objectives: This study characterizes short-term versus long-term effects of ethanol feeding on behavior, frontal lobe glial proteins, and mTOR signaling.Methods: Adolescent rats (8/group) were fed liquid diets containing 26% or 0% ethanol for 2 or 9 weeks, then subjected to novel object recognition (NOR) and open field (OF) tests. Frontal lobes were used for molecular assays.Results: Significant ethanol effects on OF performance occurred in the 2-week model (p < .0001). Further shifts in OF and NOR performance were unrelated to ethanol exposure in the 9-week models (p < .05 to p < .0001). Ethanol inhibited MAG1 (p < .01) and MBP (p < .0001) after 2 but not 9 weeks. However, both control and ethanol 9-week models had significantly reduced MAG1 (p < .001-0.0001), MBP (p < .0001), PDGFRA (p < .05-0.01), and PLP (p < .001-0.0001) relative to the 2-week models. GFAP was the only glial protein significantly inhibited by ethanol in both 2- (p < .01) and 9-week (p < .05) models. Concerning the mTOR pathway, ethanol reduced IRS-1 (p < .05) and globally inhibited mTOR (p < .01 or p < .001) in the 9- but not the 2-week model.Conclusions: Short-term versus long-term ethanol exposures differentially alter neurobehavioral function, glial protein expression, and signaling through IRS-1 and mTOR, which have known roles in myelination during adolescence. These findings suggest that strategies to prevent chronic alcohol-related brain pathology should consider the increased maturation-related vulnerability of adolescent brains.
Collapse
Affiliation(s)
- Emine B. Yalcin
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, The Providence VA Medical Center, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Busra Delikkaya
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, The Providence VA Medical Center, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - William Pelit
- Department of Chemistry, Brown University, Providence, RI, USA
| | - Yiwen Yang
- Biotechnology Graduate Program, Brown University, Providence, RI, USA
| | - Suzanne M. de la Monte
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, The Providence VA Medical Center, The Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, The Warren Alpert Medical School of Brown University, Providence, RI, USA
- Departments of Neurology and Neurosurgery, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
9
|
Shuvalova M, Dmitrieva A, Belousov V, Nosov G. The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers 2024:2361202. [PMID: 38808582 DOI: 10.1080/21688370.2024.2361202] [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: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The blood-brain barrier (BBB) regulates the exchange of metabolites and cells between the blood and brain, and maintains central nervous system homeostasis. Various factors affect BBB barrier functions, including reactive oxygen species (ROS). ROS can act as stressors, damaging biological molecules, but they also serve as secondary messengers in intracellular signaling cascades during redox signaling. The impact of ROS on the BBB has been observed in multiple sclerosis, stroke, trauma, and other neurological disorders, making blocking ROS generation a promising therapeutic strategy for BBB dysfunction. However, it is important to consider ROS generation during normal BBB functioning for signaling purposes. This review summarizes data on proteins expressed by BBB cells that can be targets of redox signaling or oxidative stress. It also provides examples of signaling molecules whose impact may cause ROS generation in the BBB, as well as discusses the most common diseases associated with BBB dysfunction and excessive ROS generation, open questions that arise in the study of this problem, and possible ways to overcome them.
Collapse
Affiliation(s)
- Margarita Shuvalova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiia Dmitrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Georgii Nosov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| |
Collapse
|
10
|
Togre NS, Melaka N, Bhoj PS, Mogadala N, Winfield M, Trivedi J, Grove D, Kotnala S, Rom SS, Sriram U, Persidsky Y. Neuroinflammatory Responses and Blood-Brain Barrier Injury in Chronic Alcohol Exposure: Role of Purinergic P2X7 Receptor Signaling. RESEARCH SQUARE 2024:rs.3.rs-4350949. [PMID: 38766082 PMCID: PMC11100971 DOI: 10.21203/rs.3.rs-4350949/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Alcohol consumption leads to neuroinflammation and blood-brain barrier (BBB) damage, resulting in neurological impairment. We previously demonstrated that ethanol-induced disruption of barrier function in human brain endothelial cells was associated with mitochondrial injury, increased ATP and extracellular vesicle (EV) release, and purinergic receptor P2X7R activation. Therefore, we aimed to evaluate the effect of P2X7r blockade on peripheral and neuro-inflammation in EtOH-exposed mice. In a chronic intermittent ethanol (CIE)-exposed mouse model, P2X7R was inhibited by two different methods: Brilliant Blue G (BBG) or gene knockout. We assessed blood ethanol concentration (BEC), plasma P2X7R and P-gp, number of extra-cellular vesicles (EV), serum ATP and EV-ATP levels. Brain microvessel gene expression and EV mtDNA copy numbers were measured by RT2 PCR array and digital PCR, respectively. A RT2 PCR array of brain microvessels revealed significant upregulation of proinflammatory genes involved in apoptosis, vasodilation, and platelet activation in CIE-exposed animals, which were decreased 15-50-fold in BBG-treated CIE-exposed animals. Plasma P-gp levels and serum P2X7R shedding were significantly increased in CIE-exposed animals. Pharmacological or genetic suppression of P2X7R decreased P2X7R shedding to levels equivalent to those in control group. The increase in EV number and EV-ATP content in the CIE-exposed mice was significantly reduced by P2X7R inhibition. CIE mice showed augmented EV-mtDNA copy numbers which were reduced in EVs after P2X7R inhibition or receptor knockout. These observations suggested that P2X7R signaling plays a critical role in ethanol-induced brain injury. Increased eATP, EV-ATP, EV numbers, and EV-mtDNA copy numbers highlight a new mechanism of brain injury during alcohol exposure via P2X7R and biomarkers of such damage. In this study, for the first time, we report the in vivo involvement of P2X7R signaling in CIE-induced brain injury.
Collapse
|
11
|
Seemiller LR, Flores-Cuadra J, Griffith KR, Smith GC, Crowley NA. Alcohol and stress exposure across the lifespan are key risk factors for Alzheimer's Disease and cognitive decline. Neurobiol Stress 2024; 29:100605. [PMID: 38268931 PMCID: PMC10806346 DOI: 10.1016/j.ynstr.2024.100605] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Alzheimer's Disease and related dementias (ADRD) are an increasing threat to global health initiatives. Efforts to prevent the development of ADRD require understanding behaviors that increase and decrease risk of neurodegeneration and cognitive decline, in addition to uncovering the underlying biological mechanisms behind these effects. Stress exposure and alcohol consumption have both been associated with increased risk for ADRD in human populations. However, our ability to understand causal mechanisms of ADRD requires substantial preclinical research. In this review, we summarize existing human and animal research investigating the connections between lifetime stress and alcohol exposures and ADRD.
Collapse
Affiliation(s)
- Laurel R. Seemiller
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Julio Flores-Cuadra
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Keith R. Griffith
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Grace C. Smith
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Nicole A. Crowley
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
12
|
Chaturvedi A, Rao G, Praharaj SK, Guruprasad KP, Pais V, Sadacharan CM. Decreased expression of CD200 on peripheral blood leukocytes in alcohol dependence. Alcohol 2023; 113:21-25. [PMID: 37595696 DOI: 10.1016/j.alcohol.2023.08.003] [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: 05/01/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Chronic alcohol use induces innate immune genes, which activate the innate immune system. Neuroimmune regulatory proteins [e.g., Cluster of Differentiation 200 (CD200)] are immune response regulators and are involved in balancing the immune response. This study aimed to investigate the expression of CD200 on the surface of peripheral blood leukocytes in patients with alcohol use disorder and compare them with controls. Fifty male patients with alcohol use disorder were included in the study. A baseline assessment was done, and alcohol use history, craving, and withdrawal scores were collected. A 2-mL venous blood sample was collected from cases and controls for immunophenotyping of CD200. The control group consisted of 50 participants with similar socio-economic backgrounds. The cellular expression of CD200 on total leukocytes (median ± IQR) [39.94 (28.85, 50.01)] in cases was significantly lower compared to controls [45.07 (37.70, 51.69)] (U = 896, p = 0.015). Expression of CD200 on lymphocytes in cases was negatively correlated with years of heavy drinking and this was statistically significant (r = -0.321, p = 0.023). The study indicates that cellular expression of CD200 on the surface of peripheral blood leukocytes is reduced in alcohol-dependent patients. This reduction can contribute to exaggerated immune activity, release of pro-inflammatory cytokines, chronic microglial activation, neuroinflammation, and neurodegeneration in alcohol dependence.
Collapse
Affiliation(s)
- Abhishek Chaturvedi
- Division of Biochemistry, Department of Basic Medical Sciences (DBMS), Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Guruprasad Rao
- Division of Biochemistry, Department of Basic Medical Sciences (DBMS), Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Samir Kumar Praharaj
- Department of Psychiatry, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India; Clinical Research Centre for Neuromodulation in Psychiatry, Department of Psychiatry, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Kanive Parashiva Guruprasad
- Department of Ageing Research, Manipal School of Life Sciences ((MSLS), Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vivek Pais
- Akhila Karnataka Jana Jagruthi Vedike, Shree Kshethra Dharmasthala Complex, Belthangady, 574214, Dakshina Kannada District, Karnataka, India
| | - Chakravarthy Marx Sadacharan
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, 5055 Medical Circle, Houston, TX, 77204, United States
| |
Collapse
|
13
|
Arruebarrena MA, Hawe CT, Lee YM, Branco RC. Mechanisms of Cadmium Neurotoxicity. Int J Mol Sci 2023; 24:16558. [PMID: 38068881 PMCID: PMC10706630 DOI: 10.3390/ijms242316558] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Cadmium is a heavy metal that increasingly contaminates food and drink products. Once ingested, cadmium exerts toxic effects that pose a significant threat to human health. The nervous system is particularly vulnerable to prolonged, low-dose cadmium exposure. This review article provides an overview of cadmium's primary mechanisms of neurotoxicity. Cadmium gains entry into the nervous system via zinc and calcium transporters, altering the homeostasis for these metal ions. Once within the nervous system, cadmium disrupts mitochondrial respiration by decreasing ATP synthesis and increasing the production of reactive oxygen species. Cadmium also impairs normal neurotransmission by increasing neurotransmitter release asynchronicity and disrupting neurotransmitter signaling proteins. Cadmium furthermore impairs the blood-brain barrier and alters the regulation of glycogen metabolism. Together, these mechanisms represent multiple sites of biochemical perturbation that result in cumulative nervous system damage which can increase the risk for neurological and neurodegenerative disorders. Understanding the way by which cadmium exerts its effects is critical for developing effective treatment and prevention strategies against cadmium-induced neurotoxic insult.
Collapse
Affiliation(s)
- Madelyn A. Arruebarrena
- Neuroscience and Behavior Program, University of Notre Dame, Notre Dame, IN 46556, USA; (M.A.A.); (Y.M.L.)
| | - Calvin T. Hawe
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA;
| | - Young Min Lee
- Neuroscience and Behavior Program, University of Notre Dame, Notre Dame, IN 46556, USA; (M.A.A.); (Y.M.L.)
| | - Rachel C. Branco
- Neuroscience and Behavior Program, University of Notre Dame, Notre Dame, IN 46556, USA; (M.A.A.); (Y.M.L.)
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA;
| |
Collapse
|
14
|
Hang Z, Zhou L, Xing C, Wen Y, Du H. The blood-brain barrier, a key bridge to treat neurodegenerative diseases. Ageing Res Rev 2023; 91:102070. [PMID: 37704051 DOI: 10.1016/j.arr.2023.102070] [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: 06/09/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
As a highly selective and semi-permeable barrier that separates the circulating blood from the brain and central nervous system (CNS), the blood-brain barrier (BBB) plays a critical role in the onset and treatment of neurodegenerative diseases (NDs). To delay or reverse the NDs progression, the dysfunction of BBB should be improved to protect the brain from harmful substances. Simultaneously, a highly efficient drug delivery across the BBB is indispensable. Here, we summarized several methods to improve BBB dysfunction in NDs, including knocking out risk geneAPOE4, regulating circadian rhythms, restoring the gut microenvironment, and activating the Wnt/β-catenin signaling pathway. Then we discussed the advances in BBB penetration techniques, such as transient BBB opening, carrier-mediated drug delivery, and nasal administration, which facilitates drug delivery across the BBB. Furthermore, various in vivo and in vitro BBB models and research methods related to NDs are reviewed. Based on the current research progress, the treatment of NDs in the long term should prioritize the integrity of the BBB. However, a treatment approach that combines precise control of transient BBB permeability and non-invasive targeted BBB drug delivery holds profound significance in improving treatment effectiveness, safety, and clinical feasibility during drug therapy. This review involves the cross application of biology, materials science, imaging, engineering and other disciplines in the field of BBB, aiming to provide multi-dimensional research directions and clinical ideas for the treating NDs.
Collapse
Affiliation(s)
- Zhongci Hang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Yongqiang Wen
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China.
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
15
|
Morphological and Functional Effects of Ultrasound on Blood-Brain Barrier Transitory Opening: An In Vitro Study on Rat Brain Endothelial Cells. Cells 2023; 12:cells12010192. [PMID: 36611987 PMCID: PMC9818236 DOI: 10.3390/cells12010192] [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: 10/16/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 01/05/2023] Open
Abstract
With the recent advances in medicine, human life expectancy is increasing; however, the extra years of life are not necessarily spent in good health or free from disability, resulting in a significantly higher incidence of age-associated pathologies. Among these disorders, neurodegenerative diseases have a significant impact. To this end, the presence of the protective blood-brain barrier (BBB) represents a formidable obstacle to the delivery of therapeutics. Thus, this makes it imperative to define strategies to bypass the BBB in order to successfully target the brain with the appropriate drugs. It has been demonstrated that targeting the BBB by ultrasound (US) can transiently make this anatomical barrier permeable and in so doing, allow the delivery of therapeutics. Thus, our aim was to carry out an in depth in vitro molecular and morphological study on the effects of US treatment on the BBB. The rat brain endothelial (RBE4) cell line was challenged with exposure to 12 MHz diagnostic US treatment for 10, 20, and 30 min. Cell viability assays, Western blotting analysis on the endoplasmic reticulum (ER), and oxidative stress marker evaluation were then performed, along with cytological and immunofluorescence staining, in order to evaluate the effects of US on the intercellular spaces and tight junction distribution of the brain endothelial cells. We observed that the US treatment exerted no toxic effects on either RBE4 cell viability or the upregulation/dislocation of the ER and oxidative stress marker (GRP78 and cytochrome C, respectively). Further, we observed that the application of US induced an increase in the intercellular spaces, as shown by Papanicolaou staining, mainly due to the altered distribution of the tight junction protein zonula occludens-1 (ZO-1). This latter US-dependent effect was transient and disappeared 20 min after the removal of the stimulus. In conclusion, our results show that US induces a transient alteration of the BBB, without altering the intracellular signaling pathways such as the ER and oxidative stress that could potentially be toxic for endothelial cells. These results suggested that US treatment could represent a potential strategy for improving drug delivery to the brain.
Collapse
|
16
|
Ruczaj A, Brzóska MM. Environmental exposure of the general population to cadmium as a risk factor of the damage to the nervous system: A critical review of current data. J Appl Toxicol 2023; 43:66-88. [PMID: 35304765 PMCID: PMC10084305 DOI: 10.1002/jat.4322] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/16/2022]
Abstract
Nowadays, more and more attention has been focused on the risk of the neurotoxic action of cadmium (Cd) under environmental exposure. Due to the growing incidence of nervous system diseases, including neurodegenerative changes, and suggested involvement of Cd in their aetiopathogenesis, this review aimed to discuss critically this element neurotoxicity. Attempts have been made to recognize at which concentrations in the blood and urine Cd may increase the risk of damage to the nervous system and compare it to the risk of injury of other organs and systems. The performed overview of the available literature shows that Cd may have an unfavourable impact on the human's nervous system at the concentration >0.8 μg Cd/L in the urine and >0.6 μg Cd/L in the blood. Because such concentrations are currently noted in the general population of industrialized countries, it can be concluded that environmental exposure to this xenobiotic may create a risk of damage to the nervous system and be involved in the aetiopathogenesis of neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, as well as worsening cognitive and behavioural functions. The potential mechanism of Cd neurotoxicity consists in inducing oxidative stress, disrupting the activity of enzymes essential to the proper functioning of the nervous system and destroying the homoeostasis of bioelements in the brain. Thus, further studies are necessary to recognize accurately both the risk of nervous system damage in the general population due to environmental exposure to Cd and the mechanism of this action.
Collapse
Affiliation(s)
- Agnieszka Ruczaj
- Department of ToxicologyMedical University of BialystokBialystokPoland
| | | |
Collapse
|
17
|
Dabhi N, Mastorakos P, Sokolowski JD, Kellogg RT, Park MS. Effect of drug use in the treatment of acute ischemic stroke: A scoping review. Surg Neurol Int 2022; 13:367. [PMID: 36128166 PMCID: PMC9479649 DOI: 10.25259/sni_561_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/27/2022] [Indexed: 11/04/2022] Open
Abstract
Background Drugs of abuse have been associated with ischemic stroke; however, the clinical presentation, outcomes, and treatment data in this population are limited. The overall safety and efficacy of thrombolytic therapy and thrombectomy in these patients remain unclear. This scoping review summarizes published complications and clinical outcomes in patients with recent abuse of cocaine, methamphetamine (MA), cannabis, decongestant, opioids, alcohol, and 3,4-methylenedioxymethamphetamine (MDMA) presenting with acute ischemic stroke. Methods We conducted a scoping review of the primary literature that assessed outcomes data of thrombolytic therapy or thrombectomy in drug users with acute ischemic stroke. We searched PubMed, Ovid Medline, and Web of Science. Demographic and stroke characteristics, treatment, complications, and clinical outcomes at last follow-up were collected and summarized. Results We identified 51 studies in this review. Drugs of abuse of interest were cocaine (14 studies), MDMA (one study), MA (eight studies), cannabis (23 studies), alcohol (two studies), decongestants (one study), and opioids (two studies). Clinical presentation and stroke presentation were most commonly described features. Thrombectomy outcomes were reported for four patients total (two studies), all with history of cocaine use. Thrombolysis treatment and outcomes were reported for 8851 patients (five studies) with history of cocaine, alcohol, or cannabis. Both treatments were pursued in three patients (three studies). Treatment complications included intracerebral hemorrhage, vasospasm, and cerebral edema. Conclusion Evidence for thrombolytic and thrombectomy treatment in drug users remains limited. Controlled studies are needed to examine complication profile and outcomes following thrombolytic and thrombectomy treatment in this population.
Collapse
Affiliation(s)
- Nisha Dabhi
- Department of Neurosurgery, University of Virginia, Charlottesville, United States
| | | | | | | | | |
Collapse
|
18
|
Chen X, Pang X, Yeo AJ, Xie S, Xiang M, Shi B, Yu G, Li C. The Molecular Mechanisms of Ferroptosis and Its Role in Blood-Brain Barrier Dysfunction. Front Cell Neurosci 2022; 16:889765. [PMID: 35663422 PMCID: PMC9160190 DOI: 10.3389/fncel.2022.889765] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
The blood-brain barrier (BBB) is a selective, semi-permeable layer of endothelial cells that protects the central nervous system from harmful substances circulating in blood. It is one of the important barriers of the nervous system. BBB dysfunction is an early pathophysiological change observed in nervous system diseases. There are few treatments for BBB dysfunction, so this motivates the review. Ferroptosis is a novel cell death mode caused by iron-mediated lipid peroxidation accumulation, which has recently attracted more attention due to its possible role in nervous system disorders. Studies have shown that lipid peroxidation and iron accumulation are related to the barrier dysfunction, especially the expression of tight junction proteins. Therefore, examination of the relationship between ferroptosis and BBB dysfunction may reveal new targets for the treatment of brain diseases.
Collapse
Affiliation(s)
- Xiaoshu Chen
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinru Pang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Abrey J. Yeo
- University of Queensland Centre for Clinical Research, Brisbane, QLD, Australia
| | - Siwen Xie
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Mengting Xiang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Bin Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Gongchang Yu
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Gongchang Yu,
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Chao Li,
| |
Collapse
|
19
|
Branca JJV, Carrino D, Paternostro F, Morucci G, Fiorillo C, Nicoletti C, Gulisano M, Ghelardini C, Di Cesare Mannelli L, Becatti M, Pacini A. The Protection of Zinc against Acute Cadmium Exposure: A Morphological and Molecular Study on a BBB In Vitro Model. Cells 2022; 11:cells11101646. [PMID: 35626683 PMCID: PMC9140137 DOI: 10.3390/cells11101646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Cadmium (Cd) is a well-known occupational and environmental pollutant worldwide, and its toxicity is widely recognised. Cd is reported to increase the permeability of the blood–brain barrier (BBB) and to penetrate and accumulate in the brain. Although many lines of evidence show that Cd toxicity is induced by different mechanisms, one of the best known is the Cd-dependent production of reactive oxygen species (ROS). Zinc is a trace element known as coenzyme and cofactor for many antioxidant proteins, such as metallothioneins and superoxide dismutase enzymes. To date, very little is known about the role of Zn in preventing Cd-induced blood–brain barrier (BBB) alterations. The goal of this study was to test the Zn antioxidant capacity against Cd-dependent alterations in a rat brain endothelial cell line (RBE4), as an in vitro model for BBB. In order to mimic acute Cd poisoning, RBE4 cells were treated with CdCl2 30 µM for 24 h. The protective role of ZnCl2 (50 µM) was revealed by evaluating the cell viability, reactive oxygen species (ROS) quantification, cytochrome C distribution, and the superoxide dismutase (SOD) protein activity. Additionally, the effectiveness of Zn in counteracting the Cd-induced damage was investigated by evaluating the expression levels of proteins already known to be involved in the Cd signalling pathway, such as GRP78 (an endoplasmic reticulum (ER) stress protein), caspase3 pro- and cleaved forms, and BAX. Finally, we evaluated if Zn was able to attenuate the alterations of zonula occludens-1 (ZO-1), one of the tight-junction (TJ) proteins involved in the formation of the BBB. Our data clearly demonstrate that Zn, by protecting from the SOD activity impairment induced by Cd, is able to prevent the triggering of the Cd-dependent signalling pathway that leads to ZO-1 dislocation and downregulation, and BBB damage.
Collapse
Affiliation(s)
- Jacopo J. V. Branca
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Donatello Carrino
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Ferdinando Paternostro
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Gabriele Morucci
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, University of Firenze, 50134 Firenze, Italy; (C.F.); (M.B.)
| | - Claudio Nicoletti
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Massimo Gulisano
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Firenze, 50139 Firenze, Italy; (C.G.); (L.D.C.M.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Firenze, 50139 Firenze, Italy; (C.G.); (L.D.C.M.)
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, University of Firenze, 50134 Firenze, Italy; (C.F.); (M.B.)
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
- Correspondence: ; Tel.: +39-055-2758067
| |
Collapse
|
20
|
Alcohol-Induced Alterations in the Vascular Basement Membrane in the Substantia Nigra of the Adult Human Brain. Biomedicines 2022; 10:biomedicines10040830. [PMID: 35453580 PMCID: PMC9028457 DOI: 10.3390/biomedicines10040830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/10/2022] Open
Abstract
The blood–brain barrier (BBB) represents a highly specialized interface that acts as the first line of defense against toxins. Herein, we investigated the structural and ultrastructural changes in the basement membrane (BM), which is responsible for maintaining the integrity of the BBB, in the context of chronic alcoholism. Human post-mortem tissues from the Substantia Nigra (SN) region were obtained from 44 individuals, then grouped into controls, age-matched alcoholics, and non-age-matched alcoholics and assessed using light and electron microscopy. We found significantly less CD31+ vessels in alcoholic groups compared to controls in both gray and white matter samples. Alcoholics showed increased expression levels of collagen-IV, laminin-111, and fibronectin, which were coupled with a loss of BM integrity in comparison with controls. The BM of the gray matter was found to be more disintegrated than the white matter in alcoholics, as demonstrated by the expression of both collagen-IV and laminin-111, thereby indicating a breakdown in the BM’s structural composition. Furthermore, we observed that the expression of fibronectin was upregulated in the BM of the white matter vasculature in both alcoholic groups compared to controls. Taken together, our findings highlight some sort of aggregation or clumping of BM proteins that occurs in response to chronic alcohol consumption.
Collapse
|
21
|
Chopra N, Menounos S, Choi JP, Hansbro PM, Diwan AD, Das A. Blood-Spinal Cord Barrier: Its Role in Spinal Disorders and Emerging Therapeutic Strategies. NEUROSCI 2022; 3:1-27. [PMID: 39484675 PMCID: PMC11523733 DOI: 10.3390/neurosci3010001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/14/2021] [Indexed: 11/03/2024] Open
Abstract
The blood-spinal cord barrier (BSCB) has been long thought of as a functional equivalent to the blood-brain barrier (BBB), restricting blood flow into the spinal cord. The spinal cord is supported by various disc tissues that provide agility and has different local immune responses compared to the brain. Though physiologically, structural components of the BSCB and BBB share many similarities, the clinical landscape significantly differs. Thus, it is crucial to understand the composition of BSCB and also to establish the cause-effect relationship with aberrations and spinal cord dysfunctions. Here, we provide a descriptive analysis of the anatomy, current techniques to assess the impairment of BSCB, associated risk factors and impact of spinal disorders such as spinal cord injury (SCI), amyotrophic lateral sclerosis (ALS), peripheral nerve injury (PNI), ischemia reperfusion injury (IRI), degenerative cervical myelopathy (DCM), multiple sclerosis (MS), spinal cavernous malformations (SCM) and cancer on BSCB dysfunction. Along with diagnostic and mechanistic analyses, we also provide an up-to-date account of available therapeutic options for BSCB repair. We emphasize the need to address BSCB as an individual entity and direct future research towards it.
Collapse
Affiliation(s)
- Neha Chopra
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Spiro Menounos
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
| | - Jaesung P Choi
- Centre for Inflammation, Faculty of Science, Centenary Institute, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2050, Australia; (J.P.C.); (P.M.H.)
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, Centenary Institute, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2050, Australia; (J.P.C.); (P.M.H.)
| | - Ashish D Diwan
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Abhirup Das
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia
| |
Collapse
|
22
|
Liu M, Guo S, Huang D, Hu D, Wu Y, Zhou W, Song W. Chronic Alcohol Exposure Alters Gene Expression and Neurodegeneration Pathways in the Brain of Adult Mice. J Alzheimers Dis 2022; 86:315-331. [PMID: 35034908 DOI: 10.3233/jad-215508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Chronic alcohol consumption can alter the structure of the central nervous system and disrupt cognitive function. Alcoholics are more likely to develop neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). However, the role of alcohol in promoting neurotoxicity and neurodegeneration remains unclear. OBJECTIVE In this study, we aimed at estimating the effects of chronic binge alcohol exposure on brain transcriptome and behavior changes in a chronic "Drinking in the Dark" (DID) mouse model. METHODS The adult C57BL/6J male mice were exposed to alcohol for 4 weeks. RNA-seq was applied to assess the effects of chronic alcohol exposure on transcriptome in brain. The open field test and novel object recognition test were used to assess the changes of anxiety level, locomotive function, and short-term memory induced by alcohol. RNA-seq analysis revealed that chronic alcohol exposure caused significant change in the brain transcriptome, especially in prefrontal cortex. RESULTS The gene dysregulation caused by chronic alcohol exposure includes pathways related to mitochondrial energy metabolism (such as oxidative phosphorylation) and multiple neurodegenerative diseases (such as AD and PD). Furthermore, the pathway and network analyses suggest that the genes involved in mitochondrial energy metabolism, ubiquitin-proteasome system, Wnt signaling pathway, and microtubules may attribute to the neurotoxicity and neurodegeneration caused by chronic alcohol consumption. Additionally, locomotive function was also significantly impaired. CONCLUSION This work provides gene transcriptional profile data for future research on alcohol-induced neurodegenerative diseases, especially AD and PD.
Collapse
Affiliation(s)
- Mingjing Liu
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China.,International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shipeng Guo
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China.,International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Daochao Huang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China.,International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dongjie Hu
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China.,International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yili Wu
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Weihui Zhou
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China.,International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihong Song
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China.,International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| |
Collapse
|
23
|
Guo X, Yan T, Chen M, Ma X, Li R, Li B, Yang A, Chen Y, Fang T, Yu H, Tian H, Chen G, Zhuo C. Differential effects of alcohol-drinking patterns on the structure and function of the brain and cognitive performance in young adult drinkers: A pilot study. Brain Behav 2022; 12:e2427. [PMID: 34808037 PMCID: PMC8785638 DOI: 10.1002/brb3.2427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION This study was aimed to determine how different patterns of alcohol consumption drive changes to brain structure and function and their correlation with cognitive impairments in young adult alcohol drinkers. METHODS In this study, we enrolled five groups participants and defined as: long-term abstinence from alcohol (LA), binge drinking (BD), long-term low dosage alcohol consumption but exceeding the safety drinking dosage (LD), long-term alcohol consumption of damaging dosage (LDD), and long-term heavy drinking (HD). All participants underwent magnetic resonance imaging (MRI) and functional MRI (fMRI) to acquire data on brain structure and function, including gray matter volume (GMV), fractional amplitude of low-frequency fluctuation (fALFF), regional homogeneity (ReHo), functional connectivity (FC), and brain network properties. The cognitive ability was evaluated with the California Verbal Learning Test (CVLT), intelligence quotient (IQ), and short delay free recall (SDFR). RESULTS Compared to LA, GMV significantly decreased in the brain regions in VN, SMN, and VAN in the alcohol-drinking groups (BD, LD, LDD, and HD). ReHo was significantly enhanced in the brain regions in VN, SMN, and VAN, while fALFF significantly increased in the brain regions in VN and SMN. The number of intra- and inter-modular connections within networks (VN, SMN, sensory control network [SCN], and VAN) and their connections to other modules were abnormally changed. These changes adversely affected cognition (e.g., IQ, CVLT, SDFR). CONCLUSION Despite the small sample size, this study provides new evidence supporting the need for young people to abstain from alcohol to protect their brains. These findings present strong reasoning for updating anti-alcohol slogans and guidelines for young people in the future.
Collapse
Affiliation(s)
- Xiaobing Guo
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Tongjun Yan
- Department of Psychiatry, 904th Hospital of PLA, Changzhou, Jiangsu, China
| | - Min Chen
- Institute of Mental Health, Jining Medical University, Jining, China
| | - Xiaoyan Ma
- Department of Alcohol Dependence Management, Tianjin Anding Hospital, Tianjin Medical University Clinical Hospital of Mental Health, Tianjin, China.,Tianjin Anding Hospital, Tianjin Mental Health Center, Key Laboratory of Psychiatry Neuroimaging-Genetics and Co-morbidity (PNGC_Lab) of Tianjin Medical University Clinical Hospital of Mental Health, Nankai University Affiliated Tianjin Anding Hospital, Tianjin, China
| | - Ranli Li
- Department of Alcohol Dependence Management, Tianjin Anding Hospital, Tianjin Medical University Clinical Hospital of Mental Health, Tianjin, China.,Tianjin Anding Hospital, Tianjin Mental Health Center, Key Laboratory of Psychiatry Neuroimaging-Genetics and Co-morbidity (PNGC_Lab) of Tianjin Medical University Clinical Hospital of Mental Health, Nankai University Affiliated Tianjin Anding Hospital, Tianjin, China
| | - Bo Li
- Department of Psychiatry, Tianjin Kangtai Mental Health Hospital, Tianjin, China
| | - Anqu Yang
- Department of Psychiatry, Tianjin Kangtai Mental Health Hospital, Tianjin, China
| | - Yuhui Chen
- Department of Psychiatry, Tianjin Kangtai Mental Health Hospital, Tianjin, China
| | - Tao Fang
- Key Laboratory of Real Time Brain Circuits Tracing of Neurology and Psychiatry (RTBNB_Lab), Tianjin Fourth Center Hospital, Tianjin Medical Affiliated Tianjin Fourth Central Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin, China
| | - Haiping Yu
- Department of Alcohol Dependence Management, Wenzhou Seventh Peoples Hospital, Wenzhou, China
| | - Hongjun Tian
- Key Laboratory of Real Time Brain Circuits Tracing of Neurology and Psychiatry (RTBNB_Lab), Tianjin Fourth Center Hospital, Tianjin Medical Affiliated Tianjin Fourth Central Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin, China
| | - Guangdong Chen
- Department of Alcohol Dependence Management, Wenzhou Seventh Peoples Hospital, Wenzhou, China
| | - Chuanjun Zhuo
- Key Laboratory of Real Time Brain Circuits Tracing of Neurology and Psychiatry (RTBNB_Lab), Tianjin Fourth Center Hospital, Tianjin Medical Affiliated Tianjin Fourth Central Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin, China.,Department of Alcohol Dependence Management, Wenzhou Seventh Peoples Hospital, Wenzhou, China
| |
Collapse
|
24
|
Branca JJV, Carrino D, Paternostro F, Gulisano M, Becatti M, Di Cesare Mannelli L, Pacini A. Antioxidant support to ameliorate the oxaliplatin-dependent microglial alteration: morphological and molecular study. Eur J Histochem 2021; 65. [PMID: 34755507 PMCID: PMC8607276 DOI: 10.4081/ejh.2021.3285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/20/2021] [Indexed: 12/24/2022] Open
Abstract
Oxaliplatin is a third-generation chemotherapy drug mainly used for colorectal cancer treatment. However, it is also known to trigger neuropathy whose underlying neurobiological mechanisms are still under investigation and currently available treatments show limited efficacy. It is now established that neurons are not the only cell type involved in chronic pain and that glial cells, mainly astrocytes and microglia, are involved in the initiation and maintenance of neuropathy. Among all the pathogenetic factors involved in neuropathic pain, an oxaliplatin-dependent oxidative stress plays a predominant role. In our study, the antioxidant properties of magnesium (Mg), manganese (Mn) and zinc (Zn) salts were evaluated in order to counteract microglial activation induced by oxaliplatin. The antioxidant efficacy of these metals was evaluated by the means of molecular and morphological assays on the BV-2 microglial cell line. Our data clearly show that Mg, Mn and Zn are able to prevent oxaliplatin-dependent microglial alterations by reducing both oxidative and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Jacopo J V Branca
- Department of Experimental and Clinical Medicine, Anatomy Section, University of Florence.
| | - Donatello Carrino
- Department of Experimental and Clinical Medicine, Anatomy Section, University of Florence.
| | - Ferdinando Paternostro
- Department of Experimental and Clinical Medicine, Anatomy Section, University of Florence.
| | - Massimo Gulisano
- Department of Experimental and Clinical Medicine, Anatomy Section, University of Florence.
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio, University of Florence.
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence.
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Anatomy Section, University of Florence.
| |
Collapse
|