1
|
Pich EM, Tarnanas I, Brigidi P, Collo G. Gut Microbiome-Liver-Brain axis in Alcohol Use Disorder. The role of gut dysbiosis and stress in alcohol-related cognitive impairment progression: possible therapeutic approaches. Neurobiol Stress 2025; 35:100713. [PMID: 40092632 PMCID: PMC11909761 DOI: 10.1016/j.ynstr.2025.100713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
The Gut Microbiome-Liver-Brain Axis is a relatively novel construct with promising potential to enhance our understanding of Alcohol Use Disorder (AUD), and its therapeutic approaches. Significant alterations in the gut microbiome occur in AUD even before any other systemic signs or symptoms manifest. Prolonged and inappropriate alcohol consumption, by affecting the gut microbiota and gut mucosa permeability, is thought to contribute to the development of behavioral and cognitive impairments, leading to Alcohol-Related Liver Disorders and potentially progressing into alcoholic cirrhosis, which is often associated with severe cognitive impairment related to neurodegeneration, such as hepatic encephalopathy and alcoholic dementia. The critical role of the gut microbiota is further supported by the efficacy of FDA-approved treatments for hepatic encephalopathy in alcoholic cirrhosis (i.e., lactulose and rifaximin). To stimulate new research, we hypothesize that interactions between a maladaptive stress response and a constitutional predisposition to neurodegeneration underlie the progression of AUD to conditions of Alcohol-Related Clinical Concerns with severe cognitive impairment, which represent a significant and costly burden to society. Early identification of AUD individuals at risk for developing these conditions could help to prioritize integrated therapeutic interventions targeting different substrates of the Gut Microbiome-Liver-Brain axis. Specifically, addiction medications, microbiome modulators, stress-reducing interventions, and, possibly soon, novel agents that reduce hepatic steatosis/fibrosis will be discussed in the context of digitally supported integrated therapeutic approaches. The explicit goal of this AUD treatment performed on the early stage of the disorder would be to reduce the transition from AUD to those conditions of Alcohol-Related Common Clinical Concerns associated with severe cognitive impairment, a strategy recommended for most neurological neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Ioannis Tarnanas
- Trinity College Dublin, Global Brain Health Institute, Dublin, Ireland
- Altoida Inc., Washington DC, USA
| | - Patrizia Brigidi
- Human Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Italy
| | - Ginetta Collo
- Human Neuropharmacology Unit, Department of Molecular & Translational Medicine, University of Brescia, Italy
| |
Collapse
|
2
|
Winters AD, Francescutti DM, Kracht DJ, Chaudhari DS, Zagorac B, Angoa-Perez M. The Gut Microbiome Regulates the Psychomotor Effects and Context-Dependent Rewarding Responses to Cocaine in Germ-Free and Antibiotic-Treated Animal Models. Microorganisms 2025; 13:77. [PMID: 39858845 PMCID: PMC11767876 DOI: 10.3390/microorganisms13010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Cocaine use disorder remains a major global health concern, with growing evidence that the gut microbiome modulates drug-related behaviors. This study examines the microbiome's role in cocaine-induced psychomotor activation and context-dependent reward responses using germ-free (GF) and antibiotic-treated (ABX) models. In GF mice, the absence of a microbiome blunted cocaine-induced psychomotor activation (p = 0.013), which was restored after conventionalization. GF mice also showed reduced cocaine-conditioned place preference (CPP) (p = 0.002), which normalized after conventionalization. Dopaminergic function, critical for psychomotor responses and reward, was microbiome-dependent, with increased dopamine levels (p = 0.009) and normalized turnover ratios after conventionalization. In the ABX model, microbiome depletion reduced both cocaine-induced locomotion and CPP responses (p ≤ 0.009), further supporting the role of gut microbes in modulating psychomotor and reward behaviors. ABX-treated mice also showed significant declines in microbial diversity, shifts in bacterial structure, and dysregulation in metabolic, immune, and neurotransmitter pathways (p ≤ 0.0001), including alterations in short-chain fatty acids and gamma-aminobutyric acid metabolism. These findings highlight the gut microbiome's critical role in regulating cocaine's psychomotor and rewarding effects, offering insights into potential therapeutic strategies for cocaine use disorder.
Collapse
Affiliation(s)
- Andrew D. Winters
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Dina M. Francescutti
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - David J. Kracht
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Diptaraj S. Chaudhari
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Branislava Zagorac
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Mariana Angoa-Perez
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| |
Collapse
|
3
|
Castro EM, Lotfipour S, Leslie FM. Neuroglia in substance use disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:347-369. [PMID: 40148055 DOI: 10.1016/b978-0-443-19102-2.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Substance use disorders (SUD) remain a major public health concern in which individuals are unable to control their use of substances despite significant harm and negative consequences. Drugs of abuse dysregulate major brain and behavioral functions. Glial cells, primarily microglia and astrocytes, play a crucial role in these drug-induced molecular and behavioral changes. This review explores preclinical and clinical studies of how neuroglia and their associated neuroinflammatory responses contribute to SUD and reward-related properties. We evaluate preclinical and clinical evidence for targeting neuroglia as therapeutic interventions. In addition, we evaluate the literature on the gut microbiome and its role in SUD. Clinical treatments are most effective for reducing drug cravings, and some have yielded promising results in other measures of drug use. N-Acetylcysteine, through modulation of cysteine-glutamate antiporter of glial cells, shows encouraging results across a variety of drug classes. Neuroglia and gut microbiome interactions are important factors to consider with regard to SUD and could lead to novel therapeutic avenues. Age- and sex-dependent properties of neuroglia, gut microbiome, and drug use behaviors are important areas in need of further investigation.
Collapse
Affiliation(s)
- Emily M Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
4
|
Han C, Manners MT, Robinson SA. Sex differences in opioid response: a role for the gut microbiome? Front Pharmacol 2024; 15:1455416. [PMID: 39268474 PMCID: PMC11390522 DOI: 10.3389/fphar.2024.1455416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Opioid drugs have been long known to induce different responses in males compared to females, however, the molecular mechanisms underlying these effects are yet to be fully characterized. Recent studies have established a link between the gut microbiome and behavioral responses to opioids. Chronic opioid use is associated with gut dysbiosis, or microbiome disruptions, which is thought to contribute to altered opioid analgesia and reward processing. Gut microbiome composition and functioning have also been demonstrated to be influenced by sex hormones. Despite this, there is currently very little work investigating whether sex differences in the gut microbiome mediate sex-dependent responses to opioids, highlighting a critical gap in the literature. Here, we briefly review the supporting evidence implicating a potential role for the gut microbiome in regulating sexually dimorphic opioid response and identify areas for future research.
Collapse
Affiliation(s)
- Caitlin Han
- Department of Psychology, Williams College, Williamstown, MA, United States
| | - Melissa T. Manners
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ, United States
| | - Shivon A. Robinson
- Department of Psychology, Williams College, Williamstown, MA, United States
| |
Collapse
|
5
|
Nohesara S, Mostafavi Abdolmaleky H, Thiagalingam S. Substance-Induced Psychiatric Disorders, Epigenetic and Microbiome Alterations, and Potential for Therapeutic Interventions. Brain Sci 2024; 14:769. [PMID: 39199463 PMCID: PMC11352452 DOI: 10.3390/brainsci14080769] [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: 06/23/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Substance use disorders (SUDs) are complex biopsychosocial diseases that cause neurocognitive deficits and neurological impairments by altering the gene expression in reward-related brain areas. Repeated drug use gives rise to alterations in DNA methylation, histone modifications, and the expression of microRNAs in several brain areas that may be associated with the development of psychotic symptoms. The first section of this review discusses how substance use contributes to the development of psychotic symptoms via epigenetic alterations. Then, we present more evidence about the link between SUDs and brain epigenetic alterations. The next section presents associations between paternal and maternal exposure to substances and epigenetic alterations in the brains of offspring and the role of maternal diet in preventing substance-induced neurological impairments. Then, we introduce potential therapeutic agents/approaches such as methyl-rich diets to modify epigenetic alterations for alleviating psychotic symptoms or depression in SUDs. Next, we discuss how substance use-gut microbiome interactions contribute to the development of neurological impairments through epigenetic alterations and how gut microbiome-derived metabolites may become new therapeutics for normalizing epigenetic aberrations. Finally, we address possible challenges and future perspectives for alleviating psychotic symptoms and depression in patients with SUDs by modulating diets, the epigenome, and gut microbiome.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Mental Health Research Center, Psychosocial Health Research Institute, Department of Psychiatry, School of Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Nutrition/Metabolism Laboratory, Department of Surgery, BIDMC, Harvard Medical School, Boston, MA 02215, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
6
|
Guo J, Zhao Y, Kang SG, Huang K, Tong T. Differential effects of four laboratory animal control diets on gut microbiota in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:4438-4452. [PMID: 38323712 DOI: 10.1002/jsfa.13331] [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: 07/13/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND The gut microbiota is intricate and susceptible to multiple factors, with diet being a major contributor. The present study aimed to investigate the impact of four commonly used laboratory animal control diets, namely Keao Xieli's maintenance diet (KX), HFK's 1025 (HF), Research Diets' D12450B (RD), and Lab Diet's 5CC4 (LD), on the gut microbiota of mice. RESULTS A total of 40 mice were randomly assigned to four groups, and each group was fed one of the four diets for a duration of 8 weeks. The assessment of gut microbiota was conducted using 16S rRNA sequencing both at the beginning of the study (week 0) and the end (week 8), which served as the baseline and endpoint samples, respectively. Following the 8-week feeding period, no significant differences were observed in physiological parameters, including body weight, visceral weight, and blood biochemical indices, across the four groups. Nonetheless, relative to the baseline, discernible alterations in the gut microbiota were observed in all groups, encompassing shifts in beta-diversity, hierarchical clustering, and key genera. Among the four diets, HF diet exhibited a significant influence on alpha-diversity, RD diet brought about notable changes in microbial composition at the phylum level, and LD diet demonstrated an interconnected co-occurrence network. Mantel analysis indicated no significant correlation between physiological parameters and gut microbiota in the four groups. CONCLUSION Overall, our study demonstrated that the four control diets had a minimal impact on physiological parameters, while exerting a distinct influence on the gut microbiota after 8 weeks. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Yuhan Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, P. R. China
- Beijing Laboratory for Food Quality and Safety, Beijing, P. R. China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, P. R. China
- Beijing Laboratory for Food Quality and Safety, Beijing, P. R. China
| |
Collapse
|
7
|
Browning BD, Kirkland AE, Green R, Engevik M, Alekseyenko AV, Leggio L, Tomko RL, Squeglia LM. The adolescent and young adult microbiome and its association with substance use: a scoping review. Alcohol Alcohol 2024; 59:agad055. [PMID: 37665023 PMCID: PMC10979412 DOI: 10.1093/alcalc/agad055] [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: 02/22/2023] [Revised: 07/18/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
AIMS The microbiome is a critical factor in health throughout human development. The aims of this scoping review are to (i) elucidate the differences between the youth (post-natal day 21-65 for rodents, 2-7 years for non-human primates, and 10-25 years for humans) microbiome with other life stages and (ii) identify youth-specific microbial changes associated with substance use. METHODS Peer-reviewed studies published up to May 2023 were identified in PubMed and SCOPUS and included gut and oral microbiome studies from rodents, non-human primates, and humans (N = 1733). Twenty-six articles were determined eligible based on inclusion criteria (aim 1: n = 19, aim 2: n = 7). RESULTS The adolescent and young adult oral and gut microbiomes are distinct compared to other life stages, within both non-human and human models. While there is limited research in this area, the microbiome appears to be vulnerable to substance use exposure earlier in life, including substances commonly initiated and escalated during adolescence and young adulthood (i.e. alcohol, cannabis, and tobacco). CONCLUSIONS Studies across the lifespan indicate that adolescence and young adulthood are distinct periods of development, where the microbiome is sensitive to exposures, including substance use. There is a need for more studies focused on the adolescent and young adult microbiome and substance use, as well as focused on the oral microbiome during this developmental period. Understanding the gut and oral microbiome during adolescence and young adulthood may provide insight into the pathophysiology of substance use disorders.
Collapse
Affiliation(s)
- Brittney D Browning
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., Charleston, SC 29425, United States
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 29425, United States
| | - Anna E Kirkland
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., Charleston, SC 29425, United States
| | - Rejoyce Green
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., Charleston, SC 29425, United States
| | - Melinda Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave., Charleston SC, 29425, United States
| | - Alexander V Alekseyenko
- Department of Public Health Sciences, Biomedical Informatics Center, Medical University of South Carolina, 135 Cannon St., Charleston, SC 29425, United States
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, Maryland, USA
| | - Rachel L Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., Charleston, SC 29425, United States
| | - Lindsay M Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., Charleston, SC 29425, United States
| |
Collapse
|
8
|
Li D, Liang W, Zhang W, Huang Z, Liang H, Liu Q. Fecal microbiota transplantation repairs intestinal permeability and regulates the expression of 5-HT to influence alcohol-induced depression-like behaviors in C57BL/6J mice. Front Microbiol 2024; 14:1241309. [PMID: 38249454 PMCID: PMC10797076 DOI: 10.3389/fmicb.2023.1241309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
The epidemic of alcohol abuse affects millions of people worldwide. Relevant evidence supports the notion that the gut microbiota (GM) plays a crucial role in central nervous system (CNS) function, and its composition undergoes changes following alcohol consumption. Therefore, the purpose of this study was to investigate the effect of reconstructing the gut microbiota by fecal microbiota transplantation (FMT) on alcohol dependence. Here, we established an alcohol dependence model with C57BL/6J mice and proved that FMT treatment improved anxiety-like behavior and alcohol-seeking behavior in alcohol-dependent mice. Additionally, we found that the expression of the intestinal intercellular tight junction structure proteins ZO-1 and occludin was significantly increased after FMT. FMT repaired intestinal permeability in alcohol-dependent mice and decreased the levels of lipopolysaccharide (LPS) and proinflammatory factors. Moreover, the serotonin (5-hydroxytryptamine, 5-HT) content was significantly increased in alcohol-dependent mouse intestinal and brain tissues after receiving the fecal microbiome from healthy mice. 16S rRNA sequencing demonstrated that FMT markedly reshaped the composition of the gut microbiota and elicited changes in the intestinal barrier and 5-HT levels. Collectively, our results revealed that FMT has a palliative effect on alcohol dependence and explored the underlying mechanisms, which provides new strategies for the treatment of alcohol dependence.
Collapse
Affiliation(s)
- Dezhi Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wei Liang
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Wentong Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Zhiqiang Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Haipeng Liang
- Qingyang City People's Hospital General Surgery, Qingyang, China
| | - Qing Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
9
|
Li Y, Wang Z, Li J, Yu Y, Wang Y, Jin X, Dong Y, Liu Q, Duan X, Yan N. Sodium Butyrate Ameliorates Fluorosis-Induced Neurotoxicity by Regulating Hippocampal Glycolysis In Vivo. Biol Trace Elem Res 2023; 201:5230-5241. [PMID: 36710293 DOI: 10.1007/s12011-023-03583-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/22/2023] [Indexed: 01/31/2023]
Abstract
Fluorosis can induce neurotoxicity. Sodium butyrate (SB), a histone deacetylase inhibitor, has important research potential in correcting glucose metabolism disorders and is widely used in a variety of neurological diseases and metabolic diseases, but it is not yet known whether it plays a role in combating fluoride-induced neurotoxicity. This study aims to evaluate the effect of SB on fluoride neurotoxicity and the possible associated mechanisms. The results of HE staining and Morris water maze showed that, in mice exposed to 100 mg/L fluoride for 3 months, the hippocampal cells arranged in loosely with large cell gaps and diminished in number. One thousand milligram per kilogram per day SB treatment improved fluoride-induced neuronal cell damage and spatial learning memory impairment. Western blot results showed that the abundance of malate dehydrogenase 2 (MDH2) and pyruvate dehydrogenase (PDH) in the hippocampus of fluorosis mice was increased, the abundance of pyruvate kinase M (PKM), lactate dehydrogenase (LDH), hexokinase (HK), phosphatidylinositol 3-kinase (PI3K), phosphorylated Akt (P-AKT), and hypoxia-inducible factor 1α (HIF-1α) was inhibited, and the content of lactate and ATP was decreased. SB treatment reversed the decreased glycolysis in the hippocampus of fluorosis mice. These results suggested that SB could ameliorate fluorosis-induced neurotoxicity, which might be linked with its function in regulating glycolysis as well as inhibition of the PI3K/AKT/HIF-1α pathway. Sodium butyrate ameliorates fluorosis-induced neurotoxicity by regulating hippocampal glycolysis in vivo (created with MedPeer (www.medpeer.cn)).
Collapse
Affiliation(s)
- Yangjie Li
- College of Basic Medicine, Shenyang Medical College, Shenyang, 110034, China
| | - Zhengdong Wang
- College of Basic Medicine, Shenyang Medical College, Shenyang, 110034, China
| | - Jing Li
- School of Pharmacy, Shenyang Medical College, Shenyang, 110034, China
| | - Yang Yu
- School of Medical Applied Technology, Shenyang Medical College, Shenyang, 110034, China
| | - Yuan Wang
- Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110034, China
| | - Xiaoxia Jin
- School of Public Health, Shenyang Medical College, Shenyang, 110034, China
| | - Yun Dong
- Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110034, China
| | - Qingsong Liu
- School of Public Health, Shenyang Medical College, Shenyang, 110034, China
| | - Xiaoxu Duan
- School of Public Health, Shenyang Medical College, Shenyang, 110034, China.
| | - Nan Yan
- School of Medical Applied Technology, Shenyang Medical College, Shenyang, 110034, China.
| |
Collapse
|
10
|
Huang S, Ghasem Ardabili N, Davidson TL, Riley AL. Western diet consumption does not impact the rewarding and aversive effects of morphine in male Sprague-Dawley rats. Physiol Behav 2023; 270:114317. [PMID: 37541607 DOI: 10.1016/j.physbeh.2023.114317] [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: 04/19/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/06/2023]
Abstract
The impacts of high-fat and/or high-sugar diets on opioid-induced effects are well documented; however, little is known about the effect of such diet on the affective responses to opiates. To address this issue, in the present experiment male Sprague-Dawley rats were given ad libitum access to a western-style diet (high in saturated fat and sugar) or a standard laboratory chow diet beginning in adolescence and continuing into adulthood at which point they were trained in a combined conditioned taste avoidance (CTA)/conditioned place preference (CPP) procedure to assess the aversive and rewarding effects of morphine, respectively. On four conditioning cycles, animals were given access to a novel saccharin solution, injected with morphine (1 mg/kg or 5 mg/kg), and then placed on one side of a place preference chamber. Animals were then tested for place preference and saccharin preference. All subjects injected with morphine displayed significant avoidance of the morphine-associated solution (CTA) and preferred the side associated with the drug (CPP). Furthermore, there were no differences between the two diet groups, indicating that chronic exposure to the western diet had no impact on the affective properties of morphine (despite increasing caloric intake, body weight, body fat and lean body mass). Given previously reported increases in drug self-administration in animals with a history of western-diet consumption, this study suggests that western-diet exposure may increase drug intake via mechanisms other than changes in the rewarding or aversive effects of the drug.
Collapse
Affiliation(s)
- Shihui Huang
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave, NW., Washington, D.C. 20016, United States.
| | - Negar Ghasem Ardabili
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave, NW., Washington, D.C. 20016, United States
| | - Terry L Davidson
- Laboratory for Behavioral and Neural Homeostasis, Department of Neuroscience, Center for Neuroscience and Behavior, American University, Washington, D.C. 20016, United States
| | - Anthony L Riley
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave, NW., Washington, D.C. 20016, United States.
| |
Collapse
|
11
|
Angoa-Pérez M, Zagorac B, Francescutti DM, Shaffer ZD, Theis KR, Kuhn DM. Cocaine hydrochloride, cocaine methiodide and methylenedioxypyrovalerone (MDPV) cause distinct alterations in the structure and composition of the gut microbiota. Sci Rep 2023; 13:13754. [PMID: 37612353 PMCID: PMC10447462 DOI: 10.1038/s41598-023-40892-1] [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: 02/27/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Cocaine is a highly addictive psychostimulant drug of abuse that constitutes an ongoing public health threat. Emerging research is revealing that numerous peripheral effects of this drug may serve as conditioned stimuli for its central reinforcing properties. The gut microbiota is emerging as one of these peripheral sources of input to cocaine reward. The primary objective of the present study was to determine how cocaine HCl and methylenedioxypyrovalerone, both of which powerfully activate central reward pathways, alter the gut microbiota. Cocaine methiodide, a quaternary derivative of cocaine that does not enter the brain, was included to assess peripheral influences on the gut microbiota. Both cocaine congeners caused significant and similar alterations of the gut microbiota after a 10-day course of treatment. Contrary to expectations, the effects of cocaine HCl and MDPV on the gut microbiota were most dissimilar. Functional predictions of metabolic alterations caused by the treatment drugs reaffirmed that the cocaine congeners were similar whereas MDPV was most dissimilar from the other two drugs and controls. It appears that the monoamine transporters in the gut mediate the effects of the treatment drugs. The effects of the cocaine congeners and MDPV on the gut microbiome may form the basis of interoceptive cues that can influence their abuse properties.
Collapse
Affiliation(s)
- Mariana Angoa-Pérez
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA.
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Branislava Zagorac
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dina M Francescutti
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zachary D Shaffer
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kevin R Theis
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Donald M Kuhn
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
12
|
Jew MH, Hsu CL. Alcohol, the gut microbiome, and liver disease. J Gastroenterol Hepatol 2023; 38:1205-1210. [PMID: 37096652 PMCID: PMC11272486 DOI: 10.1111/jgh.16199] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
The microorganisms inhabiting our gastrointestinal tract are critical for human health. Chronic heavy alcohol use can modulate the composition and function of the gut microbiota, thereby exacerbating end-organ damage via the gut-brain axis and the gut-liver axis. In this review, we summarize the bacterial, fungal, and viral gut microbial compositional changes associated with alcohol use and alcohol-associated liver disease and discuss the mechanisms of action by which gut dysbiosis reinforces alcohol use behavior and liver inflammation and injury. We also highlight important pre-clinical and clinical trials that target gut microbial-specific mechanisms for the treatment of alcohol use disorder and alcohol-associated liver disease.
Collapse
Affiliation(s)
- Michael H Jew
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Cynthia L Hsu
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
13
|
Kong D, Sun JX, Yang JQ, Li YS, Bi K, Zhang ZY, Wang KH, Luo HY, Zhu M, Xu Y. Ketogenic diet: a potential adjunctive treatment for substance use disorders. Front Nutr 2023; 10:1191903. [PMID: 37575322 PMCID: PMC10414993 DOI: 10.3389/fnut.2023.1191903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Substance use disorders (SUD) can lead to serious health problems, and there is a great interest in developing new treatment methods to alleviate the impact of substance abuse. In recent years, the ketogenic diet (KD) has shown therapeutic benefits as a dietary therapy in a variety of neurological disorders. Recent studies suggest that KD can compensate for the glucose metabolism disorders caused by alcohol use disorder by increasing ketone metabolism, thereby reducing withdrawal symptoms and indicating the therapeutic potential of KD in SUD. Additionally, SUD often accompanies increased sugar intake, involving neural circuits and altered neuroplasticity similar to substance addiction, which may induce cross-sensitization and increased use of other abused substances. Reducing carbohydrate intake through KD may have a positive effect on this. Finally, SUD is often associated with mitochondrial damage, oxidative stress, inflammation, glia dysfunction, and gut microbial disorders, while KD may potentially reverse these abnormalities and serve a therapeutic role. Although there is much indirect evidence that KD has a positive effect on SUD, the small number of relevant studies and the fact that KD leads to side effects such as metabolic abnormalities, increased risk of malnutrition and gastrointestinal symptoms have led to the limitation of KD in the treatment of SUD. Here, we described the organismal disorders caused by SUD and the possible positive effects of KD, aiming to provide potential therapeutic directions for SUD.
Collapse
Affiliation(s)
- Deshenyue Kong
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia-xue Sun
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ji-qun Yang
- Third People’s Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, China
| | - Yuan-sen Li
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ke Bi
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zun-yue Zhang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Kun-hua Wang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Hua-you Luo
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mei Zhu
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu Xu
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
14
|
Kargbo RB. Microbiome: The Next Frontier in Psychedelic Renaissance. J Xenobiot 2023; 13:386-401. [PMID: 37606422 PMCID: PMC10443327 DOI: 10.3390/jox13030025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 08/23/2023] Open
Abstract
The psychedelic renaissance has reignited interest in the therapeutic potential of psychedelics for mental health and well-being. An emerging area of interest is the potential modulation of psychedelic effects by the gut microbiome-the ecosystem of microorganisms in our digestive tract. This review explores the intersection of the gut microbiome and psychedelic therapy, underlining potential implications for personalized medicine and mental health. We delve into the current understanding of the gut-brain axis, its influence on mood, cognition, and behavior, and how the microbiome may affect the metabolism and bioavailability of psychedelic substances. We also discuss the role of microbiome variations in shaping individual responses to psychedelics, along with potential risks and benefits. Moreover, we consider the prospect of microbiome-targeted interventions as a fresh approach to boost or modulate psychedelic therapy's effectiveness. By integrating insights from the fields of psychopharmacology, microbiology, and neuroscience, our objective is to advance knowledge about the intricate relationship between the microbiome and psychedelic substances, thereby paving the way for novel strategies to optimize mental health outcomes amid the ongoing psychedelic renaissance.
Collapse
Affiliation(s)
- Robert B Kargbo
- Usona Institute, 2800 Woods Hollow Rd., Madison, WI 53711-5300, USA
| |
Collapse
|
15
|
Gerace E, Baldi S, Salimova M, Di Gloria L, Curini L, Cimino V, Nannini G, Russo E, Pallecchi M, Ramazzotti M, Bartolucci G, Occupati B, Lanzi C, Scarpino M, Lanzo G, Grippo A, Lolli F, Mannaioni G, Amedei A. Oral and fecal microbiota perturbance in cocaine users: Can rTMS-induced cocaine abstinence support eubiosis restoration? iScience 2023; 26:106627. [PMID: 37250301 PMCID: PMC10214473 DOI: 10.1016/j.isci.2023.106627] [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: 01/16/2023] [Revised: 03/09/2023] [Accepted: 03/31/2023] [Indexed: 05/31/2023] Open
Abstract
The effects of cocaine on microbiota have been scarcely explored. Here, we investigated the gut (GM) and oral (OM) microbiota composition of cocaine use disorder (CUD) patients and the effects of repetitive transcranial magnetic stimulation (rTMS). 16S rRNA sequencing was used to characterize GM and OM, whereas PICRUST2 assessed functional changes in microbial communities, and gas-chromatography was used to evaluate fecal short and medium chain fatty acids. CUD patients reported a significant decrease in alpha diversity and modification of the abundances of several taxa in both GM and OM. Furthermore, many predicted metabolic pathways were differentially expressed in CUD patients' stool and saliva samples, as well as reduced levels of butyric acid that appear restored to normal amounts after rTMS treatment. In conclusion, CUD patients showed a profound dysbiotic fecal and oral microbiota composition and function and rTMS-induced cocaine abstinence determined the restoration of eubiotic microbiota.
Collapse
Affiliation(s)
- Elisabetta Gerace
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, 50139 Florence, Italy
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, 50139 Florence, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Maya Salimova
- Azienda Ospedaliera Universitaria di Careggi, Clinical Toxicology and Poison Control Centre, 50134 Florence, Italy
| | - Leandro Di Gloria
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Lavinia Curini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Virginia Cimino
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, 50139 Florence, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Pallecchi
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, 50139 Florence, Italy
| | - Matteo Ramazzotti
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Gianluca Bartolucci
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, 50139 Florence, Italy
| | - Brunella Occupati
- Azienda Ospedaliera Universitaria di Careggi, Clinical Toxicology and Poison Control Centre, 50134 Florence, Italy
| | - Cecilia Lanzi
- Azienda Ospedaliera Universitaria di Careggi, Clinical Toxicology and Poison Control Centre, 50134 Florence, Italy
| | - Maenia Scarpino
- Azienda Ospedaliera Universitaria di Careggi, Neurophysiology Unit, 50134 Florence, Italy
| | - Giovanni Lanzo
- Azienda Ospedaliera Universitaria di Careggi, Neurophysiology Unit, 50134 Florence, Italy
| | - Antonello Grippo
- Azienda Ospedaliera Universitaria di Careggi, Neurophysiology Unit, 50134 Florence, Italy
| | - Francesco Lolli
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Guido Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, 50139 Florence, Italy
- Azienda Ospedaliera Universitaria di Careggi, Clinical Toxicology and Poison Control Centre, 50134 Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Interdisciplinary Internal Medicine Unit, Careggi University Hospital, 50134 Florence, Italy
| |
Collapse
|
16
|
Dickerson F, Dilmore AH, Godoy-Vitorino F, Nguyen TT, Paulus M, Pinto-Tomas AA, Moya-Roman C, Zuniga-Chaves I, Severance EG, Jeste DV. The Microbiome and Mental Health Across the Lifespan. Curr Top Behav Neurosci 2023; 61:119-140. [PMID: 35947353 DOI: 10.1007/7854_2022_384] [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] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The combined genetic material of the microorganisms in the human body, known as the microbiome, is being increasingly recognized as a major determinant of human health and disease. Although located predominantly on mucosal surfaces, these microorganisms have profound effects on brain functioning through the gut-brain axis. METHOD The content of the chapter is based on a study group session at the annual meeting of the American College of Neuropsychopharmacology (ACNP). The objective was to discuss the emerging relationship between the human microbiome and mental health as relevant to ACNP's interests in developing and evaluating novel neuropsychiatric treatment strategies. The focus is on specific brain disorders, such as schizophrenia, substance use, and Alzheimer's disease, as well as on broader clinical issues such as suicidality, loneliness and wisdom in old age, and longevity. RESULTS Studies of schizophrenia indicate that the microbiome of individuals with this disorder differs from that of non-psychiatric comparison groups in terms of diversity and composition. Differences are also found in microbial metabolic pathways. An early study in substance use disorders found that individuals with this disorder have lower levels of beta diversity in their oral microbiome than a comparison group. This measure, along with others, was used to distinguish individuals with substance use disorders from controls. In terms of suicidality, there is preliminary evidence that persons who have made a suicide attempt differ from psychiatric and non-psychiatric comparison groups in measures of beta diversity. Exploratory studies in Alzheimer's disease indicate that gut microbes may contribute to disease pathogenesis by regulating innate immunity and neuroinflammation and thus influencing brain function. In another study looking at the microbiome in older adults, positive associations were found between wisdom and alpha diversity and negative associations with subjective loneliness. In other studies of older adults, here with a focus on longevity, individuals with healthy aging and unusually long lives had an abundance of specific microorganisms which distinguished them from other individuals. DISCUSSION Future studies would benefit from standardizing methods of sample collection, processing, and analysis. There is also a need for the standardized collection of relevant demographic and clinical data, including diet, medications, cigarette smoking, and other potentially confounding factors. While still in its infancy, research to date indicates a role for the microbiome in mental health disorders and conditions. Interventions are available which can modulate the microbiome and lead to clinical improvements. These include microbiome-altering medications as well as probiotic microorganisms capable of modulating the inflammation in the brain through the gut-brain axis. This research holds great promise in terms of developing new methods for the prevention and treatment of a range of human brain disorders.
Collapse
Affiliation(s)
- Faith Dickerson
- Sheppard Pratt, Baltimore, MD, USA.
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Amanda Hazel Dilmore
- Biomedical Sciences Graduate Program, University of California, San Diego, CA, USA
- Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, CA, USA
| | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan, PR, USA
| | - Tanya T Nguyen
- Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Martin Paulus
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | | | | | - Ibrahim Zuniga-Chaves
- Department of Bacteriology, Microbial Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Emily G Severance
- Stanley Neurovirology Laboratory, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dilip V Jeste
- Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, CA, USA
| |
Collapse
|
17
|
Carrico AW, Cherenack EM, Rubin LH, McIntosh R, Ghanooni D, Chavez JV, Klatt NR, Paul RH. Through the Looking-Glass: Psychoneuroimmunology and the Microbiome-Gut-Brain Axis in the Modern Antiretroviral Therapy Era. Psychosom Med 2022; 84:984-994. [PMID: 36044613 PMCID: PMC9553251 DOI: 10.1097/psy.0000000000001133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/18/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Depression, substance use disorders, and other neuropsychiatric comorbidities are common in people with HIV (PWH), but the underlying mechanisms are not sufficiently understood. HIV-induced damage to the gastrointestinal tract potentiates residual immune dysregulation in PWH receiving effective antiretroviral therapy. However, few studies among PWH have examined the relevance of microbiome-gut-brain axis: bidirectional crosstalk between the gastrointestinal tract, immune system, and central nervous system. METHODS A narrative review was conducted to integrate findings from 159 articles relevant to psychoneuroimmunology (PNI) and microbiome-gut-brain axis research in PWH. RESULTS Early PNI studies demonstrated that neuroendocrine signaling via the hypothalamic-pituitary-adrenal axis and autonomic nervous system could partially account for the associations of psychological factors with clinical HIV progression. This review highlights the need for PNI studies examining the mechanistic relevance of the gut microbiota for residual immune dysregulation, tryptophan catabolism, and oxytocin release as key biological determinants of neuropsychiatric comorbidities in PWH (i.e., body-to-mind pathways). It also underscores the continued relevance of neuroendocrine signaling via the hypothalamic-pituitary-adrenal axis, autonomic nervous system, and oxytocin release in modifying microbiome-gut-brain axis functioning (i.e., mind-to-body pathways). CONCLUSIONS Advancing our understanding of PNI and microbiome-gut-brain axis pathways relevant to depression, substance use disorders, and other neuropsychiatric comorbidities in PWH can guide the development of novel biobehavioral interventions to optimize health outcomes. Recommendations are provided for biobehavioral and neurobehavioral research investigating bidirectional PNI and microbiome-gut-brain axis pathways among PWH in the modern antiretroviral therapy era.
Collapse
Affiliation(s)
- Adam W Carrico
- From the Department of Public Health Sciences (Carrico, Cherenack, Ghanooni, Chavez), University of Miami Miller School of Medicine, Miami, Florida; Departments of Neurology (Rubin) and Psychiatry and Behavioral Sciences (Rubin), Johns Hopkins University School of Medicine; Department of Epidemiology (Rubin), Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland; Department of Psychology (McIntosh), University of Miami College of Arts and Sciences, Coral Gables, Florida; Department of Surgery (Klatt), University of Minnesota School of Medicine, Minneapolis, Minnesota; and Department of Psychological Sciences (Paul), University of Missouri St. Louis, St. Louis, Missouri
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ma Q, Gao F, Zhou L, Fan Y, Zhao B, Xi W, Wang C, Zhu F, Ma X, Wang W, Wang Y. Characterizing serum amino acids in schizophrenic patients: Correlations with gut microbes. J Psychiatr Res 2022; 153:125-133. [PMID: 35810602 DOI: 10.1016/j.jpsychires.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/02/2022] [Accepted: 07/01/2022] [Indexed: 10/17/2022]
Abstract
Amino acid abnormalities have been suggested to be a key pathophysiological mechanism in schizophrenia (SZ). Recently, gut microbes were found to be critically involved in mental and metabolic diseases. However, the relationship between serum amino acid levels and gut microbes in SZ is rarely studied. Here, we analyzed serum amino acid levels in 76 untreated SZ patients and 79 healthy controls (HC). Serum levels of 10 amino acids were significantly altered in patients with SZ. We further classified the cut-off values for serum arginine, leucine, glutamine, and methionine levels to distinguish SZ patients from controls. These classifiers were shown to be effective in another validation cohort (49 SZ and 48 HC). The correlation between serum amino acids and clinical symptoms and cognitive functions was also analyzed. Arginine, leucine, glutamine, and methionine levels were significantly correlated with clinical symptoms and cognitive impairments in SZ patients. By metagenome shotgun sequencing of fecal samples, we found that patients with SZ with a low level of serum amino acids have higher richness and evenness of the gut microbiota. At the genus level, the abundances of Mitsuokella and Oscillibacter are significantly abnormal. At the mOTU level, 15 mOTUs in the low-level SZ group were significantly different from the HC group. In addition, Mitsuokella multacida was correlated with glutamine and methionine, respectively. Our research revealed that alterations in serum amino acid levels are critically related to changes in gut microbiota composition in SZ patients. These findings may shed light on new strategies for the diagnosis and treatment of SZ.
Collapse
Affiliation(s)
- Qingyan Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Fengjie Gao
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Lina Zhou
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yajuan Fan
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Binbin Zhao
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Wenyu Xi
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Chuyao Wang
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Feng Zhu
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiancang Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Wei Wang
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Clinical Research Center for Psychiatric Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China.
| | - Yunpeng Wang
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China.
| |
Collapse
|
19
|
Jia S, Guo X, Chen Z, Li S, Liu XA. The roles of the circadian hormone melatonin in drug addiction. Pharmacol Res 2022; 183:106371. [PMID: 35907435 DOI: 10.1016/j.phrs.2022.106371] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 10/16/2022]
Abstract
Given the devastating social and health consequences of drug addiction and the limitations of current treatments, a new strategy is needed. Circadian system disruptions are frequently associated with drug addiction. Correcting abnormal circadian rhythms and improving sleep quality may thus be beneficial in the treatment of patients with drug addiction. Melatonin, an essential circadian hormone that modulates the biological clock, has anti-inflammatory, analgesic, anti-depressive, and neuroprotective effects via gut microbiota regulation and epigenetic modifications. It has attracted scientists' attention as a potential solution to drug abuse. This review summarized scientific evidence on the roles of melatonin in substance use disorders at the cellular, circuitry, and system levels, and discussed its potential applications as an intervention strategy for drug addiction.
Collapse
Affiliation(s)
- Shuhui Jia
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xuantong Guo
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
20
|
Substance use, microbiome and psychiatric disorders. Pharmacol Biochem Behav 2022; 219:173432. [PMID: 35905802 DOI: 10.1016/j.pbb.2022.173432] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/29/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022]
Abstract
Accumulating evidence from several studies has shown association between substance use, dysregulation of the microbiome and psychiatric disorders such as depression, anxiety, and psychosis. Many of the abused substances such as cocaine and alcohol have been shown to alter immune signaling pathways and cause inflammation in both the periphery and the central nervous system (CNS). In addition, these substances of abuse also alter the composition and function of the gut microbiome which is known to play important roles such as the synthesis of neurotransmitters and metabolites, that affect the CNS homeostasis and consequent behavioral outcomes. The emerging interactions between substance use, microbiome and CNS neurochemical alterations could contribute to the development of psychiatric disorders. This review provides an overview of the associative effects of substance use such as alcohol, cocaine, methamphetamine, nicotine and opioids on the gut microbiome and psychiatric disorders involving anxiety, depression and psychosis. Understanding the relationship between substance use, microbiome and psychiatric disorders will provide insights for potential therapeutic targets, aimed at mitigating these adverse outcomes.
Collapse
|
21
|
Muchhala KH, Koseli E, Gade AR, Woods K, Minai S, Kang M, McQuiston AR, Dewey WL, Akbarali HI. Chronic Morphine Induces IL-18 in Ileum Myenteric Plexus Neurons Through Mu-opioid Receptor Activation in Cholinergic and VIPergic Neurons. J Neuroimmune Pharmacol 2022; 17:111-130. [PMID: 35106734 PMCID: PMC9343479 DOI: 10.1007/s11481-021-10050-3] [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: 12/02/2021] [Accepted: 12/26/2021] [Indexed: 12/29/2022]
Abstract
The gastrointestinal epithelium is critical for maintaining a symbiotic relationship with commensal microbiota. Chronic morphine exposure can compromise the gut epithelial barrier in mice and lead to dysbiosis. Recently, studies have implicated morphine-induced dysbiosis in the mechanism of antinociceptive tolerance and reward, suggesting the presence of a gut-brain axis in the pharmacological effects of morphine. However, the mechanism(s) underlying morphine-induced changes in the gut microbiome remains unclear. The pro-inflammatory cytokine, Interleukin-18 (IL-18), released by enteric neurons can modulate gut barrier function. Therefore, in the present study we investigated the effect of morphine on IL-18 expression in the mouse ileum. We observed that chronic morphine exposure in vivo induces IL-18 expression in the ileum myenteric plexus that is attenuated by naloxone. Given that mu-opioid receptors (MORs) are mainly expressed in enteric neurons, we also characterized morphine effects on the excitability of cholinergic (excitatory) and vasoactive intestinal peptide (VIP)-expressing (inhibitory) myenteric neurons. We found fundamental differences in the electrical properties of cholinergic and VIP neurons such that VIP neurons are more excitable than cholinergic neurons. Furthermore, MORs were primarily expressed in cholinergic neurons, although a subset of VIP neurons also expressed MORs and responded to morphine in electrophysiology experiments. In conclusion, these data show that morphine increases IL-18 in ileum myenteric plexus neurons via activation of MORs in a subset of cholinergic and VIP neurons. Thus, understanding the neurochemistry and electrophysiology of MOR-expressing enteric neurons can help to delineate mechanisms by which morphine perturbs the gut barrier.
Collapse
Affiliation(s)
- Karan H Muchhala
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA
| | - Eda Koseli
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA
| | - Aravind R Gade
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA
| | - Kareem Woods
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA
| | - Suha Minai
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA
| | - Minho Kang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA
| | - A Rory McQuiston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA, 23298, USA
| | - William L Dewey
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA
| | - Hamid I Akbarali
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1112 E. Clay Street, Richmond, VA, 23298, USA.
| |
Collapse
|
22
|
Yang J, Zhang Z, Xie Z, Bai L, Xiong P, Chen F, Zhu T, Peng Q, Wu H, Zhou Y, Ma Y, Zhang Y, Chen M, Gao J, Tian W, Shi K, Du Y, Duan Y, Wang H, Xu Y, Kuang YQ, Zhu M, Yu J, Wang K. Metformin modulates microbiota-derived inosine and ameliorates methamphetamine-induced anxiety and depression-like withdrawal symptoms in mice. Biomed Pharmacother 2022; 149:112837. [PMID: 35339829 DOI: 10.1016/j.biopha.2022.112837] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/16/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Metformin exhibits therapeutic potential in behavioural deficits induced by methamphetamine (METH) in rats. Emerging studies suggest gut microbiota may impact psychiatric symptoms, but there is no direct evidence supporting metformin's participation in the pathophysiology of withdrawal symptoms via modulation of gut microbiota. METHODS In order to define the functional impacts of gut microbiota and metformin to the behavioural deficits during METH withdrawal, we utilized a combination of fecal microbiota transplantation (FMT), high-throughput sequencing, and untargeted metabolomics technologies. RESULTS First, METH addicts exhibited higher α diversity and distinct microbial structures compared to healthy controls. In particular, the relative abundance of Rikenellaceae was positively correlated with the severity of anxiety and depression. Second, both human-to-mouse and mouse-to-mouse FMTs confirmed that METH-altered-microbiota transplantation is sufficient to promote anxiety and depression-like behaviours in recipient germ-free mice, and these behavioural disturbances could be ameliorated by metformin. In-depth analysis revealed that METH significantly altered the bacterial composition and structure as well as relative abundance of several bacterial taxa and metabolites, including Rikenellaceae and inosine, respectively, whereas add-on metformin could remodel these alterations. Finally, the inosine complementation successfully restored METH-induced anxiety and depression-like behaviours in mice. CONCLUSION This study demonstrates that METH withdrawal-induced anxiety and depression-like behaviours are reversible and transmissible via gut microbiota in a mouse model. The therapeutic effects of metformin on psychiatric manifestations are associated with microbiota-derived metabolites, highlighting the role of the gut microbiota in substance use disorders and the pathophysiology of withdrawal symptoms.
Collapse
Affiliation(s)
- Jiqing Yang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Zunyue Zhang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; School of Medicine, Yunnan University, Kunming 650091, China
| | - Zhenrong Xie
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Ling Bai
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Pu Xiong
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Fengrong Chen
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Tailin Zhu
- Ministry of Education-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qingyan Peng
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Hongjin Wu
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yong Zhou
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yuru Ma
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yongjin Zhang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Minghui Chen
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Jianyuan Gao
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Weiwei Tian
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Kai Shi
- College of Science, Guilin University of Technology, Guilin 541004, China
| | - Yan Du
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yong Duan
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Huawei Wang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yu Xu
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yi-Qun Kuang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Mei Zhu
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Juehua Yu
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Kunhua Wang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming 650500, China; School of Medicine, Yunnan University, Kunming 650091, China.
| |
Collapse
|
23
|
Yao J, Chen Y, Xu M. The critical role of short-chain fatty acids in health and disease: A subtle focus on cardiovascular disease-NLRP3 inflammasome-angiogenesis axis. Clin Immunol 2022; 238:109013. [DOI: 10.1016/j.clim.2022.109013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 12/13/2022]
|
24
|
Mousa WK, Chehadeh F, Husband S. Recent Advances in Understanding the Structure and Function of the Human Microbiome. Front Microbiol 2022; 13:825338. [PMID: 35185849 PMCID: PMC8851206 DOI: 10.3389/fmicb.2022.825338] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Trillions of microbes live within our bodies in a deep symbiotic relationship. Microbial populations vary across body sites, driven by differences in the environment, immunological factors, and interactions between microbial species. Major advances in genome sequencing enable a better understanding of microbiome composition. However, most of the microbial taxa and species of the human microbiome are still unknown. Without revealing the identity of these microbes as a first step, we cannot appreciate their role in human health and diseases. A shift in the microbial balance, termed dysbiosis, is linked to a broad range of diseases from simple colitis and indigestion to cancer and dementia. The last decade has witnessed an explosion in microbiome research that led to a better understanding of the microbiome structure and function. This understanding leads to potential opportunities to develop next-generation microbiome-based drugs and diagnostic biomarkers. However, our understanding is limited given the highly personalized nature of the microbiome and its complex and multidirectional interactions with the host. In this review, we discuss: (1) our current knowledge of microbiome structure and factors that shape the microbial composition, (2) recent associations between microbiome dysbiosis and diseases, and (3) opportunities of new microbiome-based therapeutics. We analyze common themes, promises, gaps, and challenges of the microbiome research.
Collapse
Affiliation(s)
- Walaa K. Mousa
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, United Arab Emirates
- Department of Biology, Whitman College, Walla Walla, WA, United States
- College of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Fadia Chehadeh
- Department of Biology, Whitman College, Walla Walla, WA, United States
| | - Shannon Husband
- Department of Biology, Whitman College, Walla Walla, WA, United States
| |
Collapse
|
25
|
Feng Y, Qin J, Zhou Y, Yue Q, Wei J. Spherical mesoporous Fe-N-C single-atom nanozyme for photothermal and catalytic synergistic antibacterial therapy. J Colloid Interface Sci 2022; 606:826-836. [PMID: 34425270 DOI: 10.1016/j.jcis.2021.08.054] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/22/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022]
Abstract
Nanozyme has been regarded as an efficient antibiotic to kill bacteria using the reactive oxygen species (ROS) generated by Fenton-like reaction. However, its activity is still unsatisfied and requires large amount of hydrogen peroxide with side effects toward normal tissues. Herein, spherical mesoporous Fe-N-C single-atom nanozyme (SAzyme) is designed for antibacterial therapy via photothermal treatment enhanced Fenton-like catalysis process. Due to the large pore size (4.0 nm), high specific surface area (413.9 m2 g-1) and uniform diameter (100 nm), the catalytic performance of Fe-N-C SAzyme is greatly improved. The Michaelis-Menten constant (Km) is 4.84 mmol L-1, which is similar with that of horseradish peroxidase (3.7 mmol L-1). Moreover, mesoporous Fe-N-C SAzyme shows high photothermal conversion efficiency (23.3 %) owing to the carbon framework. The catalytic activity can be enhanced under light irradiation due to the elevated reaction temperature. The bacteria can also be killed via physical heat effect. Due to the synergistic effect of nanozyme catalysis and photothermal treatment, the antibacterial performance is much higher than that using single antibacterial method. This work provides an alternative for combined antibacterial treatment via photothermal treatment assisted catalytic process using spherical mesoporous single-atom nanozyme as an antibiotic.
Collapse
Affiliation(s)
- Youyou Feng
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Jing Qin
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Yu Zhou
- Institute of Fundamental and Frontier Science, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - Qin Yue
- Institute of Fundamental and Frontier Science, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China.
| | - Jing Wei
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
26
|
Russell JT, Zhou Y, Weinstock GM, Bubier JA. The Gut Microbiome and Substance Use Disorder. Front Neurosci 2021; 15:725500. [PMID: 34531718 PMCID: PMC8439419 DOI: 10.3389/fnins.2021.725500] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/12/2021] [Indexed: 01/15/2023] Open
Abstract
Substance use disorders (SUDs) remain a significant public health challenge, affecting tens of millions of individuals worldwide each year. Often comorbid with other psychiatric disorders, SUD can be poly-drug and involve several different substances including cocaine, opiates, nicotine, and alcohol. SUD has a strong genetic component. Much of SUD research has focused on the neurologic and genetic facets of consumption behavior. There is now interest in the role of the gut microbiome in the pathogenesis of SUD. In this review, we summarize current animal and clinical evidence that the gut microbiome is involved in SUD, then address the underlying mechanisms by which the gut microbiome interacts with SUD through metabolomic, immune, neurological, and epigenetic mechanisms. Lastly, we discuss methods using various inbred and outbred mice models to gain an integrative understanding of the microbiome and host genetic controls in SUD.
Collapse
Affiliation(s)
- Jordan T Russell
- School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Yanjiao Zhou
- School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - George M Weinstock
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | |
Collapse
|
27
|
Butyrate and the Intestinal Epithelium: Modulation of Proliferation and Inflammation in Homeostasis and Disease. Cells 2021; 10:cells10071775. [PMID: 34359944 PMCID: PMC8304699 DOI: 10.3390/cells10071775] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The microbial metabolite butyrate serves as a link between the intestinal microbiome and epithelium. The monocarboxylate transporters MCT1 and SMCT1 are the predominant means of butyrate transport from the intestinal lumen to epithelial cytoplasm, where the molecule undergoes rapid β-oxidation to generate cellular fuel. However, not all epithelial cells metabolize butyrate equally. Undifferentiated colonocytes, including neoplastic cells and intestinal stem cells at the epithelial crypt base preferentially utilize glucose over butyrate for cellular fuel. This divergent metabolic conditioning is central to the phenomenon known as “butyrate paradox”, in which butyrate induces contradictory effects on epithelial proliferation in undifferentiated and differentiated colonocytes. There is evidence that accumulation of butyrate in epithelial cells results in histone modification and altered transcriptional activation that halts cell cycle progression. This manifests in the apparent protective effect of butyrate against colonic neoplasia. A corollary to this process is butyrate-induced inhibition of intestinal stem cells. Yet, emerging research has illustrated that the evolution of the crypt, along with butyrate-producing bacteria in the intestine, serve to protect crypt base stem cells from butyrate’s anti-proliferative effects. Butyrate also regulates epithelial inflammation and tolerance to antigens, through production of anti-inflammatory cytokines and induction of tolerogenic dendritic cells. The role of butyrate in the pathogenesis and treatment of intestinal neoplasia, inflammatory bowel disease and malabsorptive states is evolving, and holds promise for the potential translation of butyrate’s cellular function into clinical therapies.
Collapse
|