1
|
Novau-Ferré N, Papandreou C, Rojo-Marticella M, Canals-Sans J, Bulló M. Gut microbiome differences in children with Attention Deficit Hyperactivity Disorder and Autism Spectrum Disorder and effects of probiotic supplementation: A randomized controlled trial. RESEARCH IN DEVELOPMENTAL DISABILITIES 2025; 161:105003. [PMID: 40184961 DOI: 10.1016/j.ridd.2025.105003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Emerging evidence suggests a significant role of gut microbiota on neurodevelopmental disorders, including Attention Deficit Hyperactivity Disorder (ADHD) and Autism Spectrum Disorder (ASD). AIMS Our study aimed to compare gut microbiota composition between these disorders and evaluate the effect of probiotic supplementation. METHODS We conducted a 12-week randomized, double-blind, placebo-controlled trial with 80 children aged 5-14 years (39 with ADHD, 41 with ASD). Baseline and post-intervention fecal samples were analyzed using 16S rRNA gene sequencing to identify changes in gut microbiota composition. RESULTS We identified 22 taxa differentiating ADHD and ASD (AUC = 0.939), characterised by increased presence of Clostridia, Ruminococcaceae, and Lachnospiraceae in ADHD, and Bacteroides, Bacilli and Actinobacteria in ASD. These differences remained after accounting for potential confounders. ASD children receiving probiotics had significant increases in Chao 1, Fisher's alpha, and Shannon indices whereas no significant differences in α and β-diversity were found in ADHD. In ADHD, bacteria with potential adverse effects were under-represented. In ASD, the abundance of Eggerthellaceae, and other taxa associated with gastrointestinal problems and anxiety was decreased. CONCLUSION Variations in gut microbiota may influence responses in ADHD and ASD. Probiotic supplementation favorably altered gut microbiota composition, offering insights for future therapeutic strategies targeting the microbiome in neurodevelopmental disorders. WHAT THIS PAPER ADDS Recent research underscores the role of gut microbiota in ADHD and ASD, indicating that diet can significantly influence microbiota composition and potentially manage these neurodevelopmental disorders. This study reveals distinct differences in gut microbiota composition between children with ADHD and ASD and demonstrates that probiotic supplementation can modulate specific microbial genera in each disorder. These findings pave the way for the development of innovative microbiome-targeted therapies, offering a new avenue for the treatment of neurodevelopmental disorders. Understanding this relationship is crucial for designing future interventions.
Collapse
Affiliation(s)
- Nil Novau-Ferré
- Nutrition and Metabolic Health Research Group (NuMeH). Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain; Center of Environmental, Food and Toxicological Technology - TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Christopher Papandreou
- Nutrition and Metabolic Health Research Group (NuMeH). Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain; Center of Environmental, Food and Toxicological Technology - TecnATox, Rovira i Virgili University, 43201 Reus, Spain; Department of Nutrition and Dietetics Sciences, School of Health Sciences, Hellenic Mediterranean University (HMU), 72300 Siteia, Greece
| | - Meritxell Rojo-Marticella
- Nutrition and Mental Health Research Group (NutriSam), Department of Psychology, Rovira i Virgili University, 43007 Tarragona, Spain; Research Center for Behavior Assessment (CRAMC), Rovira i Virgili University, 43007 Tarragona, Spain
| | - Josefa Canals-Sans
- Nutrition and Mental Health Research Group (NutriSam), Department of Psychology, Rovira i Virgili University, 43007 Tarragona, Spain; Research Center for Behavior Assessment (CRAMC), Rovira i Virgili University, 43007 Tarragona, Spain.
| | - Mònica Bulló
- Nutrition and Metabolic Health Research Group (NuMeH). Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain; Center of Environmental, Food and Toxicological Technology - TecnATox, Rovira i Virgili University, 43201 Reus, Spain; CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain.
| |
Collapse
|
2
|
Kong L, Wang X, Chen G, Zhu Y, Wang L, Yan M, Zeng J, Zhou X, Lui SSY, Chan RCK. Gut microbiome characteristics in individuals across different stages of schizophrenia spectrum disorders: A systematic review and meta-analysis. Neurosci Biobehav Rev 2025; 173:106167. [PMID: 40250540 DOI: 10.1016/j.neubiorev.2025.106167] [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: 06/19/2024] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Schizophrenia (SCZ) is a complex neuropsychiatric disorder with unclear pathogenesis, limiting advances in early diagnosis and targeted interventions. Increasing evidence suggests that the gut microbiome contributed to SCZ pathophysiology, yet comprehensive characterization across illness stages remains lacking. This meta-analysis aimed to characterize gut microbial alterations across the SCZ spectrum disorder, including individuals at ultra-high risk for psychosis, first-episode psychosis (FEP) and chronic SCZ patients. A systematic search of 10 databases identified 91 case-control studies. Gut microbial outcome measures included relative abundance, alpha and beta diversity. Review Manager and R were used to analyze the data. The results showed that patients with SCZ exhibited significantly reduced alpha diversity, particularly in Shannon, Chao1, Observe and Evenness indices, compared to healthy controls. Beta diversity also differed significantly, with 88.5 % of studies reporting distinct microbial profiles across SCZ stages. Quantitative analysis revealed significantly increased relative abundance of Bacteroides and a decrease abundance of Bifidobacterium and Lactobacilli in FEP patients compared to healthy controls. Qualitative analysis further showed increasing abundance in Lactobacillus, Prevotella and Collinsella, but decreasing abundance in Faecalibacterium, Butyricicoccus, and Blautia in SCZ. Bifidobacterium exhibited stage-specific changes, decreasing in first-episode psychosis but increasing in chronic stages, while Bacteroides followed an opposite trajectory. Notably, Lactobacillus demonstrated an early upward tractor in high-risk individuals, persisting to chronic stages. This meta-analysis identified dynamic and consistent alterations in the gut microbial across the SCZ spectrum. These findings implicated the potentials of gut microbes as early indicators for identification and intervention of SCZ.
Collapse
Affiliation(s)
- Li Kong
- School of Psychology, Shanghai Normal University, Shanghai, China.
| | - Xingsong Wang
- School of Psychology, Shanghai Normal University, Shanghai, China
| | - Guanlin Chen
- School of Psychology, Shanghai Normal University, Shanghai, China
| | - Yikang Zhu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lina Wang
- School of Psychology, Shanghai Normal University, Shanghai, China
| | - Miaomiao Yan
- School of Psychology, Shanghai Normal University, Shanghai, China
| | - Jingwen Zeng
- School of Psychology, Shanghai Normal University, Shanghai, China
| | - Xiaoqi Zhou
- Center for Global Change and Ecological Forecasting, School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Simon S Y Lui
- Department of Psychiatry, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Raymond C K Chan
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, State Key Laboratory of Cognitive Science and Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, the University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Weng S, Zheng J, Lin Y, Fang H, Ko CY. Therapeutic effects of amisulpride in male schizophrenics: Role of short-chain fatty acids and gene expression changes. Physiol Behav 2025; 294:114864. [PMID: 40020893 DOI: 10.1016/j.physbeh.2025.114864] [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: 10/02/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Schizophrenia (SCZ) is a complex disorder characterized by acute symptom exacerbations. Amisulpride, an antipsychotic, has shown effects beyond its primary neurochemical actions, suggesting an influence on the gut microbiome, cytokine modulation, and short-chain fatty acid (SCFA) metabolism. This study aims to investigate these broader effects by examining changes in serum SCFA levels and gene expression profiles in peripheral blood mononuclear cells (PBMCs) following amisulpride treatment. Patients with SCZ undergoing a four-week amisulpride regimen were enrolled. Serum SCFA levels were quantified by gas chromatography, and gene expression profiling was performed in PBMCs using real-time quantitative polymerase chain reaction to assess treatment-associated changes. Results revealed that treatment with amisulpride resulted in a significant increase in serum acetate levels. Gene expression analysis revealed upregulation of G-protein coupled receptor 109a (GPR109a), histone deacetylase 1 (HDAC1), G-protein coupled receptor 43 (GPR43), Toll-like receptor 2 (TLR2), soluble CD14 (sCD14), and N-methyl-d-aspartate receptor (NMDAR), while Toll-like receptor 4 (TLR4) and pregnane X receptor (PXR) were downregulated. These findings suggest that amisulpride may modulate acetate metabolism and immune signaling pathways in SCZ, potentially contributing to anti-inflammatory effects and neuroimmune regulation. The observed increase in acetate, a key microbial metabolite, and the altered expression of immune-related genes suggest a possible link between metabolic shifts and immunomodulatory responses in SCZ pathophysiology. However, direct evidence linking these changes to gut-brain axis mechanisms remains insufficient. Further research is needed to elucidate the therapeutic implications of these metabolic and immunological alterations and their potential role in symptom modulation.
Collapse
Affiliation(s)
- Saizheng Weng
- Department of Psychiatry, Neuropsychiatric Hospital Affiliated to Fujian Medical University, 350008, PR China.
| | - Jinchi Zheng
- The third hospital of Quanzhou, Quanzhou 362000, PR China.
| | - Yichuan Lin
- The third hospital of Quanzhou, Quanzhou 362000, PR China.
| | - Hongqiao Fang
- The third hospital of Quanzhou, Quanzhou 362000, PR China.
| | - Chih-Yuan Ko
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, PR China.
| |
Collapse
|
4
|
Wang Y, Feng S, Huang Y, Peng R, Liang L, Wang W, Guo M, Zhu B, Zhang H, Liao J, Zhou J, Li H, Li X, Ning Y, Wu F, Wu K. Revealing multiple biological subtypes of schizophrenia through a data-driven approach. J Transl Med 2025; 23:505. [PMID: 40316994 PMCID: PMC12048963 DOI: 10.1186/s12967-025-06503-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/12/2025] [Indexed: 05/04/2025] Open
Abstract
INTRODUCTION The brain imaging subtypes of schizophrenia have been widely investigated using data-driven approaches. However, the heterogeneity of SZ in multiple biological data is largely unknown. METHODS A data-driven model was used to classify brain imaging, gut microbiota, and brain-gut fusion data obtained through a dot product fusion method, identifying significant subtypes and calculating their correlations with clinical symptoms and cognitive performance. RESULTS These subtypes remain relatively independent and demonstrate typical features and biomarkers, which are significantly associated with clinical symptoms and cognitive performance. Two brain subtypes with opposite structural and functional changes are identified: (1) a structural variant-dominant brain subtype with negative symptoms and cognitive deficits and (2) a functional alteration-dominant brain subtype with positive symptoms. The three gut subtypes include the following: (1) Collinsella-dominant; (2) Prevotella-dominant with positive symptoms; and (3) Streptococcus-dominant. Two brain-gut subtypes show different abnormalities in brain‒genus linkages: (1) strong connectivity of "brain function in the temporal and parietal lobes-Prevotella" with reduced attention scores and (2) strong connectivity of "brain structure and function in the frontal and parietal lobes-multiple genera" with positive symptoms. Notably, brain subtypes and brain-gut subtypes are most relevant to clinical symptoms, whereas gut subtypes reveal more cognitive biomarkers. CONCLUSION These findings show the potential to identify multiple biological subtypes with distinct biomarkers, thereby suggesting the possibility of personalized and precise treatment for SZ patients.
Collapse
Affiliation(s)
- Yuran Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Shixuan Feng
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
| | - Yuanyuan Huang
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Runlin Peng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Liqin Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Wei Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Minxin Guo
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Baoyuan Zhu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Heng Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Jianhao Liao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Jing Zhou
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Diagnosis and Rehabilitation of Dementia, Guangzhou, 510500, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Hehua Li
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Xiaobo Li
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Yuping Ning
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510370, China.
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, The Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China.
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China.
- Department of Nuclear Medicine and Radiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575, Japan.
| |
Collapse
|
5
|
Huang H, Yang N, Chen MM, Chen X, Chen W, Li X, Chen Y, Deng Z, Zhou W, Xu SX, Xie XH. Altered oral health and microbiota in drug-free patients with schizophrenia. BMC Psychiatry 2025; 25:274. [PMID: 40133801 PMCID: PMC11938765 DOI: 10.1186/s12888-025-06633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The oral microbiota is associated with neuro-psychiatric disorders. However, there is presently inadequate comprehension regarding the correlation between schizophrenia and the oral microbiota. Moreover, patients with schizophrenia frequently exhibit poor oral health, potentially influencing research outcomes. Therefore, this study aims to investigate changes in the oral microbiota and oral health status in drug-free schizophrenia patients. METHODS Oral microbiota samples were collected from 50 drug-free patients with schizophrenia and 50 healthy controls (HCs). The downstream microbiota analysis was based on Illumina sequencing of the V3-V4 hypervariable region of the 16 S rRNA gene. RESULTS The alpha diversity of SCZ group is increased, such as the Shannon index (p < 0.001) and Simpson index (p = 0.004), while the community structure also displays variance compared to the HC group (p < 0.001). Key discriminative taxa were found in LEfSe analysis, including the phyla Fusobacteriota, Firmicutes, and Actinobacteriota. The differential taxa and microbial functions showed a strong correlation with clinical oral conditions. Further analysis demonstrated that models based on the entire oral microbiota effectively distinguished SCZ patients from HC (AUC = 0.97). CONCLUSIONS The significant changes in the microbiota of Drug-free SCZ patients appear to be closely associated with the poor oral environment.
Collapse
Affiliation(s)
- Huawei Huang
- Department of Pharmacy, Second People's Hospital of Huizhou, Huizhou, China
| | - Naiyan Yang
- Department of Psychiatry, Second People's Hospital of Huizhou, Huizhou, China
| | - Mian-Mian Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoting Chen
- Department of Psychiatry, Second People's Hospital of Huizhou, Huizhou, China
| | - Wei Chen
- Department of Psychiatry, Second People's Hospital of Huizhou, Huizhou, China
| | - Xiaoping Li
- Department of Psychiatry, Second People's Hospital of Huizhou, Huizhou, China
| | - Yuchun Chen
- Department of Stomatology, Third People's Hospital of Huizhou, Huizhou, China
| | - Zhengang Deng
- Department of Psychiatry, Second People's Hospital of Huizhou, Huizhou, China
| | - Wenbing Zhou
- Department of Psychiatry, Second People's Hospital of Huizhou, Huizhou, China
| | - Shu-Xian Xu
- Department of Psychiatry, Second People's Hospital of Huizhou, Huizhou, China
| | - Xin-Hui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.
- Brain Function and Psychosomatic Medicine Institute, Second People's Hospital of Huizhou, Huizhou, Guangdong, China.
| |
Collapse
|
6
|
Scala M, Del Rocío González Soltero M, Bellido Esteban A, Biscaia Fernández JM, Romero-Ferreiro V, Serretti A, Fanelli G, Rodriguez-Jimenez R. Oropharyngeal microbiota in patients with psychotic disorders: A scoping review on compositional and functional alterations. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111288. [PMID: 39923913 DOI: 10.1016/j.pnpbp.2025.111288] [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: 07/27/2024] [Revised: 01/17/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUNDS Oropharyngeal microbiota may be implicated in the onset and progression of psychotic disorders. This scoping review aims to map the existing evidence concerning the composition, diversity, and metabolic pathways of the oropharyngeal microbiota in patients aged 18 to 65 with a main diagnosis of a psychotic disorder, including individuals at clinical high-risk for psychosis (CHRP) or experiencing first episode psychosis (FEP). METHODS The scoping review was performed according to the PRISMA-ScR checklist. The systematic literature search was conducted using PubMed, Web of Science, and CINAHL until February 2024. RESULTS Seven cross-sectional studies were included, comprising 43 individuals at CHRP, 13 with FEP, 85 with first-episode of schizophrenia (FES), 171 with schizophrenia, and 8 with another schizophrenia spectrum disorder. The oropharyngeal microbiota showed an increase in Lactobacillus gasseri abundance in schizophrenia, and in Firmicutes/Proteobacteria phylum ratio in patients experiencing CHR-P and FES. In schizophrenia, an altered β-diversity was observed alongside increased metabolic pathways related to metabolite transporters. In FES, higher α-diversity and disruptions in amino acid, carbohydrate, and xenobiotic metabolism pathways were found. Hydrogen sulfide (H2S)-producing bacteria were generally enriched in all the stages of disease. Correlations were observed between oropharyngeal microbiota and psychotic symptom domains. CONCLUSIONS Potential microbial signatures, such as Lactobacillus gasseri and H2S-producing bacteria, were identified in the oropharyngeal microbiota. Alterations in the oropharyngeal microbiota composition and function may be associated with different stages of psychotic disorders, with some overlap between CHR-P and FES.
Collapse
Affiliation(s)
- Mauro Scala
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Italy; Health Research Institute Hospital 12 de Octubre, (imas12), Madrid, Spain; Faculty of Biomedical and Health Sciences, European University of Madrid (UEM), Madrid, Spain; Complutense University of Madrid (UCM), Madrid, Spain.
| | - María Del Rocío González Soltero
- Faculty of Biomedical and Health Sciences, European University of Madrid (UEM), Madrid, Spain; Molecular Microbiology Group, Health Research Institute of the University Hospital La Paz (IdiPAZ), Hospital Universitario La Paz, Madrid, Spain.
| | - Alberto Bellido Esteban
- Faculty of Biomedical and Health Sciences, European University of Madrid (UEM), Madrid, Spain
| | | | - Verónica Romero-Ferreiro
- Health Research Institute Hospital 12 de Octubre, (imas12), Madrid, Spain; Faculty of Biomedical and Health Sciences, European University of Madrid (UEM), Madrid, Spain; CIBERSAM/ISCIII (Biomedical Research Networking Centre in Mental Health), Madrid, Spain
| | - Alessandro Serretti
- Department of Medicine and Surgery, Kore University of Enna, Italy; Oasi Research Institute-IRCCS, Troina, Italy
| | - Giuseppe Fanelli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Italy; Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Roberto Rodriguez-Jimenez
- Health Research Institute Hospital 12 de Octubre, (imas12), Madrid, Spain; Complutense University of Madrid (UCM), Madrid, Spain; CIBERSAM/ISCIII (Biomedical Research Networking Centre in Mental Health), Madrid, Spain
| |
Collapse
|
7
|
Yang Y, Hong J, Zhang Z, Zheng M, Zhao J, Fang X, Liang X, Liu J, Yang Y, Tian G, Fang C. Oral supplementation with lactic acid bacteria improve the intestinal epithelial barrier and gut microbiota of broiler chicks to alleviate Salmonella Enteritidis infection. Poult Sci 2024; 103:104385. [PMID: 39442198 PMCID: PMC11538865 DOI: 10.1016/j.psj.2024.104385] [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: 08/15/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
Lactic acid bacteria (LAB) play a key role in regulating the balance of gut microbiota and serve as a suitable alternative to antibiotics. This study aims to evaluate the characteristics of 2 LAB isolates Lactiplantibacillus plantarum Lp71 (L. plantarum Lp71) and Enterococcus faecium Ef72 (E. faecium Ef72), and their roles in alleviating Salmonella Enteritidis infection. Sixty 1-day-old chicks were randomly divided into 4 groups which treated with or without L. plantarum Lp71 and E. faecium Ef72 mixture for 21 d, and then intestinal samples were collected for gut microbiota analysis, pathological and immunohistochemical analysis at 24 h post infection with or without Salmonella Enteritidis on the 22nd d. The results showed that L. plantarum Lp71 and E. faecium Ef72 had the ability to anti-acid and anti-bile salt. Salmonella Enteritidis infection damaged the intestinal epithelial barrier and reduced the expression level of tight junction proteins (ZO-1, Claudin-1, Occludin). Oral supplementation with L. plantarum Lp71 and E. faecium Ef72 mixture could alleviated the damages to intestinal epithelial barrier by Salmonella Enteritidis infection. Salmonella Enteritidis could cause abnormal Akkermansia muciniphila proliferation and decrease the diversity of cecal microbiota in chicks. These conditions could have further led to reduce gut microbiota health index (GMHI), and improve microbial dysbiosis index (MDI). Moreover, oral supplementation with L. plantarum Lp71 and E. faecium Ef72 mixture could effectively prevent the aforementioned infection outcomes and increase the abundance proportions of the several key functions in metabolic pathways metabolic pathways such as transcription and signal transduction mechanisms. In summary, L. plantarum Lp71 and E. faecium Ef72 could be the probiotics candidates that used to prevent the damage from enteric pathogens such as Salmonella Enteritidis in broiler chicks.
Collapse
Affiliation(s)
- Yuting Yang
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Jiajun Hong
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Zheng Zhang
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Minghao Zheng
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Jingang Zhao
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Xiaowei Fang
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Xiongyan Liang
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Jing Liu
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China.
| | - Yuying Yang
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Guangming Tian
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Chun Fang
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China.
| |
Collapse
|
8
|
Wei X, Xi P, Chen M, Wen Y, Wu H, Wang L, Zhu Y, Ren Y, Gu Z. Capsule robots for the monitoring, diagnosis, and treatment of intestinal diseases. Mater Today Bio 2024; 29:101294. [PMID: 39483392 PMCID: PMC11525164 DOI: 10.1016/j.mtbio.2024.101294] [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: 06/04/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 11/03/2024] Open
Abstract
Current evidence suggests that the intestine as the new frontier for human health directly impacts both our physical and mental health. Therefore, it is highly desirable to develop the intelligent tool for the enhanced diagnosis and treatment of intestinal diseases. During the past 20 years, capsule robots have opened new avenues for research and clinical applications, potentially revolutionizing human health monitor, disease diagnosis and treatment. In this review, we summarize the research progress of edible multifunctional capsule robots in intestinal diseases. To begin, we introduce the correlation between the intestinal microbiome, intestinal gas and human diseases. After that, we focus on the technical structure of edible multifunctional robots. Subsequently, the biomedical applications in the monitoring, diagnosis and treatment of intestinal diseases are discussed in detail. Last but not least, the main challenges of multifunctional capsule robots during the development process are summarized, followed by a vision for future development opportunities.
Collapse
Affiliation(s)
- Xiangyu Wei
- Department of Rheumatology, Research Center of Immunology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Department of Rheumatology, Affiliated Municipal Hospital of Xuzhou Medical University, Xuzhou, 221100, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Peipei Xi
- Department of Emergency, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Minjie Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Ya Wen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Hao Wu
- Department of Otolaryngology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
| | - Li Wang
- Institutes of Biomedical Sciences and the Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yujuan Zhu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Yile Ren
- Department of Rheumatology, Affiliated Municipal Hospital of Xuzhou Medical University, Xuzhou, 221100, China
| | - Zhifeng Gu
- Department of Rheumatology, Research Center of Immunology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
| |
Collapse
|
9
|
Cheng W, Zhao M, Zhang X, Zhou X, Yan J, Li R, Shen H. Schizophrenia and antipsychotic medications present distinct and shared gut microbial composition: A meta-analysis. Schizophr Res 2024; 274:257-268. [PMID: 39388810 DOI: 10.1016/j.schres.2024.07.053] [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: 04/18/2024] [Revised: 07/09/2024] [Accepted: 07/26/2024] [Indexed: 10/12/2024]
Abstract
There are some conflicting results regarding alterations of gut microbial composition in schizophrenia (SZ), even a few meta-analysis studies have addressed this field. Ignoring of antipsychotic medication effects may cause the large heterogeneity and impact on study results. This study is a meta-analysis to systematically evaluate composition of gut microbiota in patients with SZ, to elucidate the impact of antipsychotic use and reveal distinct and shared gut bacteria in SZ and antipsychotic medications. We re-analyzed the publicly available 16S rRNA-gene amplicon datasets by a standardized pipeline in QIIME2, used the natural log of response ratios as an effect index to directly and quantitatively compare composition of gut microbiota by random-effects meta-analysis with resampling tests in Metawin, ultimately to evaluate distinct abundance of gut bacteria. A total of 19 studies with 1968 participants (1067 patients with SZ and 901 healthy controls (HCs)) were included in this meta-analysis. The alterations of alpha diversity indices occurred in SZ on antipsychotics but not in drug-naïve or -free patients, while variation of beta diversity metrics appeared in SZ regardless of antipsychotic use. After antipsychotic treatment, reversed Simpson index, decreased observed species index and significant difference of Bray-Curtis distance were observed in patients. Especially, risperidone treatment increased the Shannon and Simpson indices. Noteworthy, three differed genera, including Lactobacillus, Roseburia and Dialister, were identified in both states of antipsychotic use. This meta-analysis is to provide a novel insight that SZ and antipsychotic medications present distinct and shared gut microbial composition.
Collapse
Affiliation(s)
- Weirong Cheng
- Department of psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 264, Guangzhou Road, Nanjing, China
| | - Mengjie Zhao
- Neuro-psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 264, Guangzhou Road, Nanjing, China
| | - Xinyun Zhang
- School of Sports Medicine and Rehabilitation, Beijing Sport University, No. 48, Xinxi Road, Beijing, China.
| | - Xia Zhou
- Neuro-psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 264, Guangzhou Road, Nanjing, China.
| | - Jun Yan
- Department of Geriatrics, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 264, Guangzhou Road, Nanjing, China.
| | - Rui Li
- School of Pharmacy, Nanjing Medical University, No. 101, Longmian Avenue, Nanjing, China.
| | - Hong Shen
- Neuro-psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 264, Guangzhou Road, Nanjing, China.
| |
Collapse
|
10
|
Xie Y, Li Z, Fan Y, Liu X, Yi R, Gan Y, Yang Z, Liu S. Integrated gut microbiome and UHPLC-MS metabolomics to reveal the prevention mechanism of pidanjiangtang granules on IGT Rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156201. [PMID: 39531936 DOI: 10.1016/j.phymed.2024.156201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Pidanjiangtang (PDJT) is a traditional Chinese medicine formula empirically used to treat impaired glucose tolerance (IGT) based on the "Pidan" theory from the classic ancient book Nei Jing. However, the mechanism of PDJT intervention for IGT remains to be studied. OBJECTIVE This study aims to explore the mechanism of PDJT granules intervention in IGT by integrating gut microbiome and UHPLC-MS untargeted metabolomics. MATERIALS AND METHODS The IGT model was established in 6-week-old male Sprague-Dawley (SD) rats by feeding them a high-fat diet and using an STZ injection. The low, medium, and high doses of PDJT were used for six weeks. metformin (Glucophage) was used as the positive control drug. The efficacy of PDJT was evaluated using fasting blood glucose (FBG), blood glucose maximum (BGmax), blood lipid, and inflammatory factor levels. Finally, 16S rDNA gut microbiome sequencing with metabolomics analysis was used to explore the pharmacological mechanism of PDJT intervention in IGT. RESULTS PDJT could reverse the phenotype of IGT rats, reduce blood glucose levels, improve lipid metabolism disorder, and reduce inflammatory response. Gut microbiome analysis found that PDJT can improve gut microbiota composition and abundance of three phyla (Firmicutes, Bacteroidota, Desulfobacterota) and four genera (unclassified_f__Lachnospiraceae, Ruminococcus, Allobaculum, Desulfovibrio), which play an important role in the process of PDJT intervention on glucose metabolism and lipid metabolism in IGT rats. UHPLC-MS untargeted metabolomics showed that PDJT could regulate the levels of 258 metabolites in lipid metabolism pathways, inflammatory response pathways, fat and protein digestion, and absorption. The combined analysis of the two omics showed that improving the body's metabolism by gut microbes may be the possible mechanism of PDJT in treating IGT. Thus, this study provides a new method to integrate gut microbiome and UHPLC-MS untargeted metabolomics to evaluate the pharmacodynamics and mechanism of PDJT intervention in IGT, providing valuable ideas and insights for future research on the treatment of IGT with traditional Chinese medicine.
Collapse
Affiliation(s)
- Yu Xie
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zirong Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Yue Fan
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Liu
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ran Yi
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaoyao Gan
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zixuan Yang
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shangjian Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
11
|
Gui L, Zuo X, Feng J, Wang M, Chen Z, Sun Y, Qi J, Chen Z, Pathak JL, Zhang Y, Cui C, Zhang P, Guo X, Lv Q, Zhang X, Zhang Y, Gu J, Lin Z. Outgrowth of Escherichia is susceptible to aggravation of systemic lupus erythematosus. Arthritis Res Ther 2024; 26:191. [PMID: 39511594 PMCID: PMC11542361 DOI: 10.1186/s13075-024-03413-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is linked to host gut dysbiosis. Here we performed faecal gut microbiome sequencing to investigate SLE-pathogenic gut microbes and their potential mechanisms. METHODS There were 134 healthy controls (HCs) and 114 SLE cases for 16 S ribosomal RNA (rRNA) sequencing and 97 HCs and 124 SLE cases for shotgun metagenomics. Faecal microbial changes and associations with clinical phenotypes were evaluated, and SLE-associated microbial genera were identified in amplicon analysis. Next, metagenomic sequencing was applied for accurate identification of microbial species and discovery of their metabolic pathways and immunogenic peptides both relevant to SLE. Finally, contribution of specific taxa to disease development was confirmed by oral gavage into lupus-prone MRL/lpr mice. RESULTS SLE patients had gut microbiota richness reduction and composition alteration, particularly lupus nephritis and active patients. Proteobacteria/Bacteroidetes (P/B) ratio was remarkably up-regulated, and Escherichia was identified as the dominantly expanded genus in SLE, followed by metagenomics accurately located Escherichia coli and Escherichia unclassified species. Significant associations primarily appeared among Escherichia coli, metabolic pathways of purine nucleotide salvage or peptidoglycan maturation and SLE disease activity index (SLEDAI), and between multiple epitopes from Escherichia coli and disease activity or renal involvement phenotype. Finally, gavage with faecal Escherichia revealed that it upregulated lupus-associated serum traits and aggravated glomerular lesions in MRL/lpr mice. CONCLUSION We characterize a novel SLE exacerbating Escherichia outgrowth and suggest its contribution to SLE procession may be partially associated with metabolite changes and cross-reactivity of gut microbiota-associated epitopes and host autoantigens. The findings could provide a deeper insight into gut Escherichia in the procession of SLE.
Collapse
Affiliation(s)
- Lian Gui
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Zuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Junmei Feng
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mingbang Wang
- Department of Neonatology, Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Shenzhen, China
- Microbiome Therapy Center, Department of Experiment & Research, Medical School, South China Hospital, Shenzhen University, Shenzhen, China
| | - Zena Chen
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuhan Sun
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Qi
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhuanggui Chen
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Janak L Pathak
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yanli Zhang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunping Cui
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Pingping Zhang
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinghua Guo
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qing Lv
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xi Zhang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Zhiming Lin
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
12
|
Song P, Yuan X, Li X, Song X, Wang Y. Multi-Loss Disentangled Generative-Discriminative Learning for Multimodal Representation in Schizophrenia. IEEE J Biomed Health Inform 2024; 28:6395-6404. [PMID: 38117620 DOI: 10.1109/jbhi.2023.3337661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Schizophrenia (SCZ) is a multifactorial mental illness, thus it will be beneficial for exploring this disease using multimodal data, including functional magnetic resonance imaging (fMRI), genes, and the gut microbiome. Previous studies reported combining multimodal data can offer complementary information for better depicting the abnormalities of SCZ. However, the existing multimodal-based methods have multiple limitations. First, most approaches cannot fully use the relationships among different modalities for the downstream tasks. Second, representing multimodal data by the modality-common and modality-specific components can improve the performance of multimodal analysis but often be ignored. Third, most methods conduct the model for classification or regression, thus a unified model is needed for finishing these tasks simultaneously. To this end, a multi-loss disentangled generative-discriminative learning (MDGDL) model was developed to tackle these issues. Specifically, using disentangled learning method, the genes and gut microbial biomarkers were represented and separated into two modality-specific vectors and one modality-common vector. Then, a generative-discriminative framework was introduced to uncover the relationships between fMRI features and these three latent vectors, further producing the attentive vectors, which can help fMRI features for the downstream tasks. To validate the performance of MDGDL, an SCZ classification task and a cognitive score regression task were conducted. Results showed the MDGDL achieved superior performance and identified the most important multimodal biomarkers for the SCZ. Our proposed model could be a supplementary approach for multimodal data analysis. Based on this method, we could analyze the SCZ by combining multimodal data, and further obtain some interesting findings.
Collapse
|
13
|
Kang JW, Vemuganti V, Kuehn JF, Ulland TK, Rey FE, Bendlin BB. Gut microbial metabolism in Alzheimer's disease and related dementias. Neurotherapeutics 2024; 21:e00470. [PMID: 39462700 PMCID: PMC11585892 DOI: 10.1016/j.neurot.2024.e00470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
Multiple studies over the last decade have established that Alzheimer's disease and related dementias (ADRD) are associated with changes in the gut microbiome. These alterations in organismal composition result in changes in the abundances of functions encoded by the microbial community, including metabolic capabilities, which likely impact host disease mechanisms. Gut microbes access dietary components and other molecules made by the host and produce metabolites that can enter circulation and cross the blood-brain barrier (BBB). In recent years, several microbial metabolites have been associated with or have been shown to influence host pathways relevant to ADRD pathology. These include short chain fatty acids, secondary bile acids, tryptophan derivatives (such as kynurenine, serotonin, tryptamine, and indoles), and trimethylamine/trimethylamine N-oxide. Notably, some of these metabolites cross the BBB and can have various effects on the brain, including modulating the release of neurotransmitters and neuronal function, inducing oxidative stress and inflammation, and impacting synaptic function. Microbial metabolites can also impact the central nervous system through immune, enteroendocrine, and enteric nervous system pathways, these perturbations in turn impact the gut barrier function and peripheral immune responses, as well as the BBB integrity, neuronal homeostasis and neurogenesis, and glial cell maturation and activation. This review examines the evidence supporting the notion that ADRD is influenced by gut microbiota and its metabolites. The potential therapeutic advantages of microbial metabolites for preventing and treating ADRD are also discussed, highlighting their potential role in developing new treatments.
Collapse
Affiliation(s)
- Jea Woo Kang
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Vaibhav Vemuganti
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessamine F Kuehn
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Tyler K Ulland
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
14
|
Ioannou M, Borkent J, Andreu-Sánchez S, Wu J, Fu J, Sommer IEC, Haarman BCM. Reproducible gut microbial signatures in bipolar and schizophrenia spectrum disorders: A metagenome-wide study. Brain Behav Immun 2024; 121:165-175. [PMID: 39032544 DOI: 10.1016/j.bbi.2024.07.009] [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: 01/12/2024] [Revised: 05/30/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Numerous studies report gut microbiome variations in bipolar disorder (BD) and schizophrenia spectrum disorders (SSD) compared to healthy individuals, though, there is limited consensus on which specific bacteria are associated with these disorders. METHODS In this study, we performed a comprehensive metagenomic shotgun sequencing analysis in 103 Dutch patients with BD/SSD and 128 healthy controls matched for age, sex, body mass index and income, while accounting for diet quality, transit time and technical confounders. To assess the replicability of the findings, we used two validation cohorts (total n = 203), including participants from a distinct population with a different metagenomic isolation protocol. RESULTS The gut microbiome of the patients had a significantly different β-diversity, but not α-diversity nor neuroactive potential compared to healthy controls. Initially, twenty-six bacterial taxa were identified as differentially abundant in patients. Among these, the previously reported genera Lachnoclostridium and Eggerthella were replicated in the validation cohorts. Employing the CoDaCoRe learning algorithm, we identified two bacterial balances specific to BD/SSD, which demonstrated an area under the receiver operating characteristic curve (AUC) of 0.77 in the test dataset. These balances were replicated in the validation cohorts and showed a positive association with the severity of psychiatric symptoms and antipsychotic use. Last, we showed a positive association between the relative abundance of Klebsiella and Klebsiella pneumoniae with antipsychotic use and between the Anaeromassilibacillus and lithium use. CONCLUSIONS Our findings suggest that microbial balances could be a reproducible method for identifying BD/SSD-specific microbial signatures, with potential diagnostic and prognostic applications. Notably, Lachnoclostridium and Eggerthella emerge as frequently occurring bacteria in BD/SSD. Last, our study reaffirms the previously established link between Klebsiella and antipsychotic medication use and identifies a novel association between Anaeromassilibacillus and lithium use.
Collapse
Affiliation(s)
- Magdalini Ioannou
- University of Groningen and University Medical Center Groningen, Department of Psychiatry, Groningen, the Netherlands; University of Groningen and University Medical Center Groningen, Department of Biomedical Sciences, Groningen, the Netherlands.
| | - Jenny Borkent
- University of Groningen and University Medical Center Groningen, Department of Biomedical Sciences, Groningen, the Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen and University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands; University of Groningen and University Medical Center Groningen, Department of Pediatrics, Groningen, the Netherlands
| | - Jiafei Wu
- University of Groningen and University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands
| | - Jingyuan Fu
- University of Groningen and University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands; University of Groningen and University Medical Center Groningen, Department of Pediatrics, Groningen, the Netherlands
| | - Iris E C Sommer
- University of Groningen and University Medical Center Groningen, Department of Biomedical Sciences, Groningen, the Netherlands
| | - Bartholomeus C M Haarman
- University of Groningen and University Medical Center Groningen, Department of Psychiatry, Groningen, the Netherlands
| |
Collapse
|
15
|
Ling Z, Lan Z, Cheng Y, Liu X, Li Z, Yu Y, Wang Y, Shao L, Zhu Z, Gao J, Lei W, Ding W, Liao R. Altered gut microbiota and systemic immunity in Chinese patients with schizophrenia comorbid with metabolic syndrome. J Transl Med 2024; 22:729. [PMID: 39103909 PMCID: PMC11302365 DOI: 10.1186/s12967-024-05533-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) is highly prevalent in individuals with schizophrenia (SZ), leading to negative consequences like premature mortality. Gut dysbiosis, which refers to an imbalance of the microbiota, and chronic inflammation are associated with both SZ and MetS. However, the relationship between gut dysbiosis, host immunological dysfunction, and SZ comorbid with MetS (SZ-MetS) remains unclear. This study aims to explore alterations in gut microbiota and their correlation with immune dysfunction in SZ-MetS, offering new insights into its pathogenesis. METHODS AND RESULTS We enrolled 114 Chinese patients with SZ-MetS and 111 age-matched healthy controls from Zhejiang, China, to investigate fecal microbiota using Illumina MiSeq sequencing targeting 16 S rRNA gene V3-V4 hypervariable regions. Host immune responses were assessed using the Bio-Plex Pro Human Cytokine 27-Plex Assay to examine cytokine profiles. In SZ-MetS, we observed decreased bacterial α-diversity and significant differences in β-diversity. LEfSe analysis identified enriched acetate-producing genera (Megamonas and Lactobacillus), and decreased butyrate-producing bacteria (Subdoligranulum, and Faecalibacterium) in SZ-MetS. These altered genera correlated with body mass index, the severity of symptoms (as measured by the Scale for Assessment of Positive Symptoms and Scale for Assessment of Negative Symptoms), and triglyceride levels. Altered bacterial metabolic pathways related to lipopolysaccharide biosynthesis, lipid metabolism, and various amino acid metabolism were also found. Additionally, SZ-MetS exhibited immunological dysfunction with increased pro-inflammatory cytokines, which correlated with the differential genera. CONCLUSION These findings suggested that gut microbiota dysbiosis and immune dysfunction play a vital role in SZ-MetS development, highlighting potential therapeutic approaches targeting the gut microbiota. While these therapies show promise, further mechanistic studies are needed to fully understand their efficacy and safety before clinical implementation.
Collapse
Affiliation(s)
- Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250000, China.
| | - Zhiyong Lan
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250000, China
| | - Xia Liu
- Department of Intensive Care Unit, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Zhimeng Li
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Ying Yu
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Yuwei Wang
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China
| | - Li Shao
- School of Clinical Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, 310015, China
| | - Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Gao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250000, China
- Department of Basic Medicine, Shandong First Medical University, Jinan, Shandong, 250000, China
| | - Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Rongxian Liao
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, 324003, China.
| |
Collapse
|
16
|
Wang Y, Bi S, Li X, Zhong Y, Qi D. Perturbations in gut microbiota composition in schizophrenia. PLoS One 2024; 19:e0306582. [PMID: 38959253 PMCID: PMC11221673 DOI: 10.1371/journal.pone.0306582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/19/2024] [Indexed: 07/05/2024] Open
Abstract
Schizophrenia is a severe, complex and long-term psychiatric disorder with unclear etiology. Gut microbes influence the central nervous system via the gut-brain axis. Consequently, investigations of the relationship between gut microbes and schizophrenia are warranted. This study involved 29 patients with schizophrenia and 30 age-matched normal controls. After 16S rRNA gene sequencing and whole-genome shotgun metagenomic sequencing, we analyzed microbial diversity, composition, and function. According to 16S rRNA and metagenomic gene sequencing results, patients with schizophrenia had higher abundances of Clostridium and Megasphaera. Functional analysis showed that sphingolipid, phosphonates and phosphinates, as well as glutamine metabolism were associated with the occurrence and development of schizophrenia. Our data suggest that the gut microbiota exerts an effect on patients with schizophrenia, providing valuable insights into the potential regulation of in the context of this disorder.
Collapse
Affiliation(s)
- YiMeng Wang
- School of Public Health, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - SiGuo Bi
- Clinical Lab, Hulunbuir Third People’s Hospital (Hulunbuir Mental Health Center), Yakeshi, Inner Mongolia, China
| | - XiaoLong Li
- Clinical Lab, Hulunbuir Third People’s Hospital (Hulunbuir Mental Health Center), Yakeshi, Inner Mongolia, China
| | - YuTao Zhong
- Clinical Lab, Hulunbuir Third People’s Hospital (Hulunbuir Mental Health Center), Yakeshi, Inner Mongolia, China
| | - DongDong Qi
- School of Public Health, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Clinical Lab, Hulunbuir Third People’s Hospital (Hulunbuir Mental Health Center), Yakeshi, Inner Mongolia, China
| |
Collapse
|
17
|
Theleritis C, Stefanou MI, Demetriou M, Alevyzakis E, Triantafyllou K, Smyrnis N, Spandidos DA, Rizos E. Association of gut dysbiosis with first‑episode psychosis (Review). Mol Med Rep 2024; 30:130. [PMID: 38785152 PMCID: PMC11148526 DOI: 10.3892/mmr.2024.13254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
The gut‑microbiota‑brain axis is a complex bidirectional communication system linking the gastrointestinal tract to the brain. Changes in the balance, composition and diversity of the gut‑microbiota (gut dysbiosis) have been found to be associated with the development of psychosis. Early‑life stress, along with various stressors encountered in different developmental phases, have been shown to be associated with the abnormal composition of the gut microbiota, leading to irregular immunological and neuroendocrine functions, which are potentially responsible for the occurrence of first‑episode psychosis (FEP). The aim of the present narrative review was to summarize the significant differences of the altered microbiome composition in patients suffering from FEP vs. healthy controls, and to discuss its effects on the occurrence and intensity of symptoms in FEP.
Collapse
Affiliation(s)
- Christos Theleritis
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, School of Medicine, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Marina Demetriou
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Evangelos Alevyzakis
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, Second Department of Propaedeutic Internal Medicine, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Nikolaos Smyrnis
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Emmanouil Rizos
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
18
|
Chen Y, Yu H, Xue F, Bai J, Guo L, Peng Z. 16S rRNA gene sequencing reveals altered gut microbiota in young adults with schizophrenia and prominent negative symptoms. Brain Behav 2024; 14:e3579. [PMID: 38841824 PMCID: PMC11154826 DOI: 10.1002/brb3.3579] [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: 10/13/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Gut dysbiosis has been established as a characteristic of schizophrenia (SCH). However, the signatures regarding SCH patients with prominent negative symptoms (SCH-N) in young adults have been poorly elucidated. METHODS Stool samples were obtained from 30 young adults with SCH-N, 32 SCH patients with prominent positive symptoms (SCH-P) along with 36 healthy controls (HCs). Microbial diversity and composition were analyzed by 16S rRNA gene sequencing. Meanwhile, psychiatric symptoms were assessed by the positive and negative syndrome scale (PANSS). RESULTS There is a significant difference in β-diversity but not α-diversity indexes among the three groups. Moreover, we found a higher abundance of Fusobacteria and Proteobacteria phyla and a lower abundance of Firmicutes phyla in SCH-N when compared with HC. Besides, we identified a diagnostic potential panel comprising six genera (Coprococcus, Monoglobus, Prevotellaceae_NK3B31_group, Escherichia-Shigella, Dorea, and Butyricicoccus) that can distinguish SCH-N from HC (area under the curve = 0.939). However, the difference in microbial composition between the SCH-N and SCH-P is much less than that between SCH-N and the HC, and SCH-N and SCH-P cannot be effectively distinguished by gut microbiota. CONCLUSION The composition of gut microbiota was changed in the patients with SCH-N, which may help in further understanding of pathogenesis in young adults with SCH-N.
Collapse
Affiliation(s)
- Yi‐Huan Chen
- Department of PsychiatryXijing HospitalAir Force Medical UniversityXi'anChina
| | - Huan Yu
- Department of PsychiatryXijing HospitalAir Force Medical UniversityXi'anChina
| | - Fen Xue
- Department of PsychiatryXijing HospitalAir Force Medical UniversityXi'anChina
| | - Jie Bai
- Department of PsychiatryXijing HospitalAir Force Medical UniversityXi'anChina
- Department of PsychiatryGaoxin HospitalXi'anChina
| | - Li Guo
- Department of PsychiatryXijing HospitalAir Force Medical UniversityXi'anChina
| | - Zheng‐Wu Peng
- Department of PsychiatryXijing HospitalAir Force Medical UniversityXi'anChina
| |
Collapse
|
19
|
Dicks LMT. Gut Bacteria Provide Genetic and Molecular Reporter Systems to Identify Specific Diseases. Int J Mol Sci 2024; 25:4431. [PMID: 38674014 PMCID: PMC11050607 DOI: 10.3390/ijms25084431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
With genetic information gained from next-generation sequencing (NGS) and genome-wide association studies (GWAS), it is now possible to select for genes that encode reporter molecules that may be used to detect abnormalities such as alcohol-related liver disease (ARLD), cancer, cognitive impairment, multiple sclerosis (MS), diabesity, and ischemic stroke (IS). This, however, requires a thorough understanding of the gut-brain axis (GBA), the effect diets have on the selection of gut microbiota, conditions that influence the expression of microbial genes, and human physiology. Bacterial metabolites such as short-chain fatty acids (SCFAs) play a major role in gut homeostasis, maintain intestinal epithelial cells (IECs), and regulate the immune system, neurological, and endocrine functions. Changes in butyrate levels may serve as an early warning of colon cancer. Other cancer-reporting molecules are colibactin, a genotoxin produced by polyketide synthetase-positive Escherichia coli strains, and spermine oxidase (SMO). Increased butyrate levels are also associated with inflammation and impaired cognition. Dysbiosis may lead to increased production of oxidized low-density lipoproteins (OX-LDLs), known to restrict blood vessels and cause hypertension. Sudden changes in SCFA levels may also serve as a warning of IS. Early signs of ARLD may be detected by an increase in regenerating islet-derived 3 gamma (REG3G), which is associated with changes in the secretion of mucin-2 (Muc2). Pro-inflammatory molecules such as cytokines, interferons, and TNF may serve as early reporters of MS. Other examples of microbial enzymes and metabolites that may be used as reporters in the early detection of life-threatening diseases are reviewed.
Collapse
Affiliation(s)
- Leon M T Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
20
|
Guo C, Bai Y, Li P, He K. The emerging roles of microbiota-derived extracellular vesicles in psychiatric disorders. Front Microbiol 2024; 15:1383199. [PMID: 38650872 PMCID: PMC11033316 DOI: 10.3389/fmicb.2024.1383199] [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: 02/07/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
Major depressive disorder, schizophrenia, and bipolar disorder are three major psychiatric disorders that significantly impact the well-being and overall health of patients. Some researches indicate that abnormalities in the gut microbiota can trigger certain psychiatric diseases. Microbiota-derived extracellular vesicles have the ability to transfer bioactive compounds into host cells, altering signaling and biological processes, ultimately influencing the mental health and illness of the host. This review aims to investigate the emerging roles of microbiota-derived extracellular vesicles in these three major psychiatric disorders and discusses their roles as diagnostic biomarkers and therapies for these psychiatric disorders.
Collapse
Affiliation(s)
- Chuang Guo
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| | - Yulong Bai
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| | - Pengfei Li
- Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, China
| | - Kuanjun He
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| |
Collapse
|
21
|
Mosquera FEC, Guevara-Montoya MC, Serna-Ramirez V, Liscano Y. Neuroinflammation and Schizophrenia: New Therapeutic Strategies through Psychobiotics, Nanotechnology, and Artificial Intelligence (AI). J Pers Med 2024; 14:391. [PMID: 38673018 PMCID: PMC11051547 DOI: 10.3390/jpm14040391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
The prevalence of schizophrenia, affecting approximately 1% of the global population, underscores the urgency for innovative therapeutic strategies. Recent insights into the role of neuroinflammation, the gut-brain axis, and the microbiota in schizophrenia pathogenesis have paved the way for the exploration of psychobiotics as a novel treatment avenue. These interventions, targeting the gut microbiome, offer a promising approach to ameliorating psychiatric symptoms. Furthermore, advancements in artificial intelligence and nanotechnology are set to revolutionize psychobiotic development and application, promising to enhance their production, precision, and effectiveness. This interdisciplinary approach heralds a new era in schizophrenia management, potentially transforming patient outcomes and offering a beacon of hope for those afflicted by this complex disorder.
Collapse
Affiliation(s)
| | | | | | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia; (F.E.C.M.); (M.C.G.-M.); (V.S.-R.)
| |
Collapse
|
22
|
Aburto MR, Cryan JF. Gastrointestinal and brain barriers: unlocking gates of communication across the microbiota-gut-brain axis. Nat Rev Gastroenterol Hepatol 2024; 21:222-247. [PMID: 38355758 DOI: 10.1038/s41575-023-00890-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 02/16/2024]
Abstract
Crosstalk between gut and brain has long been appreciated in health and disease, and the gut microbiota is a key player in communication between these two distant organs. Yet, the mechanisms through which the microbiota influences development and function of the gut-brain axis remain largely unknown. Barriers present in the gut and brain are specialized cellular interfaces that maintain strict homeostasis of different compartments across this axis. These barriers include the gut epithelial barrier, the blood-brain barrier and the blood-cerebrospinal fluid barrier. Barriers are ideally positioned to receive and communicate gut microbial signals constituting a gateway for gut-microbiota-brain communication. In this Review, we focus on how modulation of these barriers by the gut microbiota can constitute an important channel of communication across the gut-brain axis. Moreover, barrier malfunction upon alterations in gut microbial composition could form the basis of various conditions, including often comorbid neurological and gastrointestinal disorders. Thus, we should focus on unravelling the molecular and cellular basis of this communication and move from simplistic framing as 'leaky gut'. A mechanistic understanding of gut microbiota modulation of barriers, especially during critical windows of development, could be key to understanding the aetiology of gastrointestinal and neurological disorders.
Collapse
Affiliation(s)
- María R Aburto
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
23
|
Bi K, Lei Y, Kong D, Li Y, Fan X, Luo X, Yang J, Wang G, Li X, Xu Y, Luo H. Progress in the study of intestinal microbiota involved in morphine tolerance. Heliyon 2024; 10:e27187. [PMID: 38533077 PMCID: PMC10963202 DOI: 10.1016/j.heliyon.2024.e27187] [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: 04/10/2023] [Revised: 01/09/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Morphine is a widely used opioid for treatment of pain. The attendant problems including morphine tolerance and morphine dependence pose a major public health challenge. In recent years, there has been increasing interest in the gastrointestinal microbiota in many physiological and pathophysiological processes. The connectivity network between the gut microbiota and the brain is involved in multiple biological systems, and bidirectional communication between them is critical in gastrointestinal tract homeostasis, the central nervous system, and the microbial system. Many research have previously shown that morphine has a variety of effects on the gastrointestinal tract, but none have determined the function of intestinal microbiota in morphine tolerance. This study reviewed the mechanisms of morphine tolerance from the perspective of dysregulation of microbiota-gut-brain axis homeostasis, by summarizing the possible mechanisms originating from the gut that may affect morphine tolerance and the improvement of morphine tolerance through the gut microbiota.
Collapse
Affiliation(s)
- Ke Bi
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Yi Lei
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Deshenyue Kong
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Yuansen Li
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Xuan Fan
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Xiao Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Jiqun Yang
- Third People's Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, 650041, China
| | - Guangqing Wang
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Xuejun Li
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Yu Xu
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Huayou Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
24
|
Li Z, Tao X, Wang D, Pu J, Liu Y, Gui S, Zhong X, Yang D, Zhou H, Tao W, Chen W, Chen X, Chen Y, Chen X, Xie P. Alterations of the gut microbiota in patients with schizophrenia. Front Psychiatry 2024; 15:1366311. [PMID: 38596637 PMCID: PMC11002218 DOI: 10.3389/fpsyt.2024.1366311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Schizophrenia is a complex psychiatric disorder, of which molecular pathogenesis remains largely unknown. Accumulating evidence suggest that gut microbiota may affect brain function via the complex gut-brain axis, which may be a potential contributor to schizophrenia. However, the alteration of gut microbiota showed high heterogeneity across different studies. Therefore, this study aims to identify the consistently altered gut microbial taxa associated with schizophrenia. Methods We conducted a systematic search and synthesis of the up-to-date human gut microbiome studies on schizophrenia, and performed vote counting analyses to identify consistently changed microbiota. Further, we investigated the effects of potential confounders on the alteration of gut microbiota. Results We obtained 30 available clinical studies, and found that there was no strong evidence to support significant differences in α-diversity and β-diversity between schizophrenic patients and healthy controls. Among 428 differential gut microbial taxa collected from original studies, we found that 8 gut microbial taxa were consistently up-regulated in schizophrenic patients, including Proteobacteria, Gammaproteobacteria, Lactobacillaceae, Enterobacteriaceae, Lactobacillus, Succinivibrio, Prevotella and Acidaminococcus. While 5 taxa were consistently down-regulated in schizophrenia, including Fusicatenibacter, Faecalibacterium, Roseburia, Coprococcus and Anaerostipes. Discussion These findings suggested that gut microbial changes in patients with schizophrenia were characterized by the depletion of anti-inflammatory butyrate-producing genera, and the enrichment of certain opportunistic bacteria genera and probiotics. This study contributes to further understanding the role of gut microbiota in schizophrenia, and developing microbiota-based diagnosis and therapy for schizophrenia.
Collapse
Affiliation(s)
- Zhuocan Li
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangkun Tao
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dongfang Wang
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Juncai Pu
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyun Liu
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Siwen Gui
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Xiaogang Zhong
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Dan Yang
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haipeng Zhou
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Tao
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyi Chen
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaopeng Chen
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Chen
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang Chen
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xie
- National Health Commission (NHC) Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- Chongqing Institute for Brain and Intelligence, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Wilson DR, Binford L, Hickson S. The Gut Microbiome and Mental Health. J Holist Nurs 2024; 42:79-87. [PMID: 37082808 DOI: 10.1177/08980101231170487] [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] [Indexed: 04/22/2023]
Abstract
The gut microbiome has been well researched in the past few years and may be a target for treating mental illness. Trillions of bacteria in the digestive system work with the brain, immune function, and endocrine pathways. This gut microbiome ecosystem mediates the interaction between the human being and the environment making its inclusion in holistic nursing essential. Changes in normal balance of the gut microbiome occur with diet, antibiotics and other medications, stress, cancer treatment, geography and environment, and current illnesses. When the microbiome is challenged a "dysbiotic" state leads to inadequate production of needed neurotransmitters such as serotonin and dopamine. Research has shown links between the dysbiosis, and the inflammatory response system that are known to contribute to depression, anxiety, and schizophrenia. Understanding the role of the gut microbiome can be beneficial to holistic nurses, providing a new tool to prevent, treat, or reduce symptoms of mental illness and improve general immune function. This innocuous holistic approach to mental wellness is becoming an important evidenced-based approach.
Collapse
Affiliation(s)
- Debra Rose Wilson
- Lenora C. Reuther Chair of Excellence. Austin Peay State University, Clarksville TN, Walden University, USA
| | | | | |
Collapse
|
26
|
Vasileva SS, Yang Y, Baker A, Siskind D, Gratten J, Eyles D. Associations of the Gut Microbiome With Treatment Resistance in Schizophrenia. JAMA Psychiatry 2024; 81:292-302. [PMID: 38294805 PMCID: PMC10831632 DOI: 10.1001/jamapsychiatry.2023.5371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/09/2023] [Indexed: 02/01/2024]
Abstract
Importance There is growing interest in the role of gut microbiome composition in schizophrenia. However, lifestyle factors are often neglected, and few studies have investigated microbiome composition in treatment-resistant schizophrenia. Objective To explore associations between the gut microbiome and schizophrenia diagnosis, treatment resistance, clozapine response, and treatment-related adverse effects while adjusting for demographic and lifestyle factors. Design, Setting, and Participants In this case-control study of adults aged 20 to 63 years, stool samples and data on demographic characteristics, lifestyle, and medication use were collected and gut microbiome measures obtained using shotgun metagenomics. Participants with a schizophrenia diagnosis were referred through psychiatric inpatient units and outpatient clinics. Data were collected for 4 distinct groups: control individuals without a psychiatric diagnosis (past or present), individuals with treatment-responsive schizophrenia taking nonclozapine antipsychotic medications, clozapine-responsive individuals with treatment-resistant schizophrenia, and clozapine-nonresponsive individuals with treatment-resistant schizophrenia. Participants were recruited between November 2020 and November 2021. Control individuals were recruited in parallel through posters and online advertisements and matched for age, sex, and body mass index (BMI) to the individuals with schizophrenia. Participants were excluded if taking antibiotics in the past 2 months, if unable to communicate in English or otherwise follow study instructions, were pregnant or planning to become pregnant, or had any concomitant disease or condition making them unsuited to the study per investigator assessment. Data were analyzed from January 2022 to March 2023. Main Outcomes and Measures Omics relationship matrices, α and β diversity, and relative abundance of microbiome features. Results Data were collected for 97 individuals (71 [74%] male; mean [SD] age, 40.4 [10.3] years; mean [SD] BMI, 32.8 [7.4], calculated as weight in kilograms divided by height in meters squared). Significant microbiome associations with schizophrenia were observed at multiple taxonomic and functional levels (eg, common species: b2, 30%; SE, 13%; adjusted P = .002) and treatment resistance (eg, common species: b2, 27%; SE, 16%; adjusted P = .03). In contrast, limited evidence was found for microbiome associations with clozapine response, constipation, or metabolic syndrome. Significantly decreased microbial richness was found in individuals with schizophrenia compared to control individuals (t95 = 4.25; P < .001; mean [SD] for control individuals, 151.8 [32.31]; mean [SD] for individuals with schizophrenia, 117.00 [36.2]; 95% CI, 18.6-51.0), which remained significant after a covariate and multiple comparison correction. However, limited evidence was found for differences in β diversity (weighted UniFrac) for schizophrenia diagnosis (permutational multivariate analysis of variance [PERMANOVA]: R2, 0.03; P = .02), treatment resistance (R2, 0.02; P = .18), or clozapine response (R2, 0.04; P = .08). Multiple differentially abundant bacterial species (19) and metabolic pathways (162) were found in individuals with schizophrenia, which were primarily associated with treatment resistance and clozapine exposure. Conclusions and Relevance The findings in this study are consistent with the idea that clozapine induces alterations to gut microbiome composition, although the possibility that preexisting microbiome differences contribute to treatment resistance cannot be ruled out. These findings suggest that prior reports of microbiome alterations in individuals with chronic schizophrenia may be due to medication or lifestyle factors and that future studies should incorporate these variables in their design and interpretation.
Collapse
Affiliation(s)
- Svetlina S. Vasileva
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Queensland Centre for Mental Health Research, Wacol, Queensland, Australia
| | - Yuanhao Yang
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Andrea Baker
- Queensland Centre for Mental Health Research, Wacol, Queensland, Australia
- Metro South Addiction and Mental Health Service, Metro South Health, Brisbane, Brisbane South, Queensland, Australia
| | - Dan Siskind
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Queensland Centre for Mental Health Research, Wacol, Queensland, Australia
- Metro South Addiction and Mental Health Service, Metro South Health, Brisbane, Brisbane South, Queensland, Australia
- University of Queensland School of Clinical Medicine, Brisbane, Queensland, Australia
| | - Jacob Gratten
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Darryl Eyles
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Queensland Centre for Mental Health Research, Wacol, Queensland, Australia
| |
Collapse
|
27
|
Sen P, Prandovszky E, Honkanen JK, Chen O, Yolken R, Suvisaari J. Dysregulation of Microbiota in Patients With First-Episode Psychosis Is Associated With Symptom Severity and Treatment Response. Biol Psychiatry 2024; 95:370-379. [PMID: 38061464 DOI: 10.1016/j.biopsych.2023.10.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/21/2023] [Accepted: 10/23/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND The gut microbiome has been implicated in the pathogenesis of mental disorders where the gut-brain axis acts as a bidirectional communication network. METHODS Herein, we investigated the compositional and functional differences of gut microbiome between patients with first-episode psychosis (FEP) (n = 26) and healthy control participants (n = 22) using whole-genome shotgun sequencing. In addition, we assessed the oral microbiome in patients with FEP (n = 13) and listed their taxonomic diversity. RESULTS Our findings suggest that there is a dysbiosis of gut microbiota in patients with FEP. Relative abundance of Bifidobacterium adolescentis, Prevotella copri, and Turicibacter sanguinis was markedly increased (linear discriminant analysis scores [log10] > 1, and Mann-Whitney U test; false discovery rate-adjusted p values < .05) in the FEP group compared with the healthy control participants. Pathway analysis indicated that several metabolic pathways, particularly deoxyribonucleotide biosynthesis, branched-chain amino acid biosynthesis, tricarboxylic acid cycle, and fatty acid elongation and biosynthesis, were dysregulated in the FEP group compared with the healthy control group. In addition, this preliminary study was able to identify specific gut microbes (at baseline) that were predictive of weight gain in the FEP group at a 1-year follow-up. Bacteroides dorei, Bifidobacterium adolescentis, Turicibacter sanguinis, Roseburia spp., and Ruminococcus lactaris were positively associated (eXtreme gradient boosting, XGBoost regression model, Shapley additive explanations, R2 = 0.82) with weight gain. CONCLUSIONS Our findings may suggest the involvement of gut microbiota in the pathogenesis of psychosis. The benefit of modulation of the gut microbiome in the treatment of psychotic disorders should be explored further.
Collapse
Affiliation(s)
- Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Emese Prandovszky
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jarno K Honkanen
- Translational Immunology Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Ou Chen
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Robert Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jaana Suvisaari
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Finnish Institute for Health and Welfare, Helsinki, Finland.
| |
Collapse
|
28
|
Zhang L, Yuan X, Li X, Zhang X, Mao Y, Hu S, Andreassen OA, Wang Y, Song X. Gut microbial diversity moderates polygenic risk of schizophrenia. Front Psychiatry 2024; 15:1275719. [PMID: 38362027 PMCID: PMC10868137 DOI: 10.3389/fpsyt.2024.1275719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/04/2024] [Indexed: 02/17/2024] Open
Abstract
Background Schizophrenia (SCZ) is a heritable disorder with a polygenic architecture, and the gut microbiota seems to be involved in its development and outcome. In this study, we investigate the interplay between genetic risk and gut microbial markers. Methods We included 159 first-episode, drug-naïve SCZ patients and 86 healthy controls. The microbial composition of feces was characterized using the 16S rRNA sequencing platform, and five microbial α-diversity indices were estimated [Shannon, Simpson, Chao1, the Abundance-based Eoverage Estimator (ACE), and a phylogenetic diversity-based estimate (PD)]. Polygenic risk scores (PRS) for SCZ were constructed using data from large-scale genome-wide association studies. Effects of microbial α-diversity, microbial abundance, and PRS on SCZ were evaluated via generalized linear models. Results We confirmed that PRS was associated with SCZ (OR = 2.08, p = 1.22×10-5) and that scores on the Shannon (OR = 0.29, p = 1.15×10-8) and Simpson (OR = 0.29, p = 1.25×10-8) indices were inversely associated with SCZ risk. We found significant interactions (p < 0.05) between PRS and α-diversity indices (Shannon, Simpson, and PD), with the effects of PRS being larger in those exhibiting higher diversity compared to those with lower diversity. Moreover, the PRS effects were larger in individuals with a high abundance of the genera Romboutsia, Streptococcus, and Anaerostipes than in those with low abundance (p < 0.05). All three of these genera showed protective effects against SCZ. Conclusion The current findings suggest an interplay between the gut microbiota and polygenic risk of SCZ that warrants replication in independent samples. Experimental studies are needed to determine the underpinning mechanisms.
Collapse
Affiliation(s)
- Liyuan Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xiuxia Yuan
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xue Li
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xiaoyun Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Yiqiao Mao
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Yunpeng Wang
- Centre for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Biological Psychiatry, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Wu H, Liu Y, Han Y, Liu B, Chen S, Ye Z, Li J, Xie L, Wu X. Integrated Analysis of Gut Microbiome, Inflammation, and Neuroimaging Features Supports the Role of Microbiome-Gut-Brain Crosstalk in Schizophrenia. SCHIZOPHRENIA BULLETIN OPEN 2024; 5:sgae026. [PMID: 39610873 PMCID: PMC11604084 DOI: 10.1093/schizbullopen/sgae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Background and Hypothesis Gut microbiota has been implicated in the pathogenesis of schizophrenia (SZ) and relevant changes in the brain, but the underlying mechanism remains elusive. This study aims to investigate the microbiota-gut-brain crosstalk centered on peripheral inflammation in SZ patients. Study Design We recruited a cohort of 182 SZ patients and 120 healthy controls (HC). Multi-omics data, including fecal 16S rRNA, cytokine data, and neuroimaging data, were collected and synthesized for analysis. Multi-omics correlations and mediation analyses were utilized to determine the associations of gut microbiome with inflammatory cytokines and neuroimaging characteristics. Additionally, machine learning models for effective SZ diagnosis were separately generated based on gut microbial and neuroimaging data. Study Results Gut microbial dysbiosis, characterized by a decrease in butyrate-producing bacteria and an increase in proinflammatory bacteria, has been identified in SZ patients. These key microbial taxa were associated with increased inflammatory cytokines, potentially through mediating lipid metabolic pathways such as steroid biosynthesis and linoleic acid metabolism. Further analysis revealed altered microbial genera to be correlated with disrupted gray matter volume and regional homogeneity in SZ patients. Importantly, certain inflammatory cytokines mediated the relationship between the SZ-enriched genus Succinivibrio and aberrant activity of anterior cingulate cortex and left inferior temporal gyrus in the SZ group. Moreover, the classification model based on gut microbial data showed comparable efficacy to the model based on brain functional signatures in SZ diagnosis. Conclusions This study presents evidence for the dysregulated microbiota-gut-brain axis in SZ and emphasizes the central role of peripheral inflammation.
Collapse
Affiliation(s)
- Hui Wu
- Psychiatry Department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Psychiatry Department, The First People’s Hospital of Kashi, Sun Yat-sen University, Kashi, China
- Radiology Department, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yaxi Liu
- Psychiatry Department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yunwu Han
- Psychiatry Department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bingdong Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of Microbiology, Guangdong Academy of Sciences, Guangdong, China
| | - Shengyun Chen
- Psychiatry Department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiye Ye
- Psychiatry Department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianbo Li
- Psychiatry Department, The First People’s Hospital of Kashi, Sun Yat-sen University, Kashi, China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of Microbiology, Guangdong Academy of Sciences, Guangdong, China
| | - Xiaoli Wu
- Psychiatry Department, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Psychiatry Department, The First People’s Hospital of Kashi, Sun Yat-sen University, Kashi, China
| |
Collapse
|
30
|
Ling Z, Cheng Y, Liu X, Yan X, Wu L, Shao L, Gao J, Lei W, Song Q, Zhao L, Jin G. Altered oral microbiota and immune dysfunction in Chinese elderly patients with schizophrenia: a cross-sectional study. Transl Psychiatry 2023; 13:383. [PMID: 38071192 PMCID: PMC10710460 DOI: 10.1038/s41398-023-02682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Schizophrenia (SZ) is a complex psychiatric neurodevelopmental disorder with uncertain etiology and pathogenesis. Increasing evidence has recognized the key role of the gut microbiota in SZ. However, few studies have investigated the potential link between oral microbiota and SZ. We studied the tongue coating microbiota and inflammatory profiles of 118 elderly SZ patients and 97 age-matched healthy controls using Illumina MiSeq sequencing and multiplex immunoassays, respectively. Reduced α-diversity, along with a significant difference in β-diversity, were observed in patients with SZ. We have identified SZ-associated oral dysbiosis, characterized by increased Streptococcus and Fusobacterium, as well as decreased Prevotella and Veillonella. These differential genera could potentially serve as biomarkers for SZ, either alone or in combination. Additionally, an elevated Streptococcus/Prevotella ratio could indicate oral dysbiosis. These differential genera formed two distinct clusters: Streptococcus-dominated and Prevotella-dominated, which exhibited different correlations with the altered immunological profiles. Furthermore, we also observed disruptions in the inferred microbiota functions in SZ-associated microbiota, particularly in lipid and amino acid metabolism. Our study provides novel insights into the characteristics of tongue coating microbiota and its associations with immunological disturbances in elderly SZ patients, which offer new targets for the diagnosis and treatment of SZ in the elderly.
Collapse
Affiliation(s)
- Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Jinan Microecological Biomedicine Shandong Laboratory, 250000, Jinan, Shandong, China.
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, 250000, Jinan, Shandong, China
| | - Xia Liu
- Department of Intensive Care Unit, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
| | - Xiumei Yan
- Department of Laboratory Medicine, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China
| | - Lingbin Wu
- Department of Laboratory Medicine, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China
| | - Li Shao
- School of Clinical Medicine, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, 310015, Hangzhou, Zhejiang, China
| | - Jie Gao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, 250000, Jinan, Shandong, China
- School of Basic Medicine, Shandong First Medical University, 250000, Jinan, Shandong, China
| | - Qinghai Song
- Department of Psychiatry, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China
| | - Longyou Zhao
- Department of Laboratory Medicine, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China.
| | - Guolin Jin
- Department of Psychiatry, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China.
| |
Collapse
|
31
|
Dubey H, Roychoudhury R, Alex A, Best C, Liu S, White A, Carlson A, Azcarate-Peril MA, Mansfield LS, Knickmeyer R. Effect of Human Infant Gut Microbiota on Mouse Behavior, Dendritic Complexity, and Myelination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563309. [PMID: 37961091 PMCID: PMC10634763 DOI: 10.1101/2023.10.24.563309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The mammalian gut microbiome influences numerous developmental processes. In human infants it has been linked with cognition, social skills, hormonal responses to stress, and brain connectivity. Yet, these associations are not necessarily causal. The present study tested whether two microbial stool communities, common in human infants, affected behavior, myelination, dendritic morphology, and spine density when used to colonize mouse models. Humanized animals were more like specific-pathogen free mice than germ-free mice for most phenotypes, although in males, both humanized groups were less social. Both humanized groups had thinner myelin sheaths in the hippocampus, than did germ-free animals. Humanized animals were similar to each other except for dendritic morphology and spine density where one group had greater dendritic length in the prefrontal cortex, greater dendritic volume in the nucleus accumbens, and greater spine density in both regions, compared to the other. Results add to a body of literature suggesting the gut microbiome impacts brain development. Teaser Fecal transplants from human infants with highly abundant Bifidobacterium , an important inhabitant of the intestinal tract of breastfed newborns, may promote brain connectivity in mice.
Collapse
|
32
|
Murray N, Al Khalaf S, Bastiaanssen TFS, Kaulmann D, Lonergan E, Cryan JF, Clarke G, Khashan AS, O’Connor K. Compositional and Functional Alterations in Intestinal Microbiota in Patients with Psychosis or Schizophrenia: A Systematic Review and Meta-analysis. Schizophr Bull 2023; 49:1239-1255. [PMID: 37210594 PMCID: PMC10483467 DOI: 10.1093/schbul/sbad049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
BACKGROUND AND HYPOTHESIS Intestinal microbiota is intrinsically linked to human health. Evidence suggests that the composition and function of the microbiome differs in those with schizophrenia compared with controls. It is not clear how these alterations functionally impact people with schizophrenia. We performed a systematic review and meta-analysis to combine and evaluate data on compositional and functional alterations in microbiota in patients with psychosis or schizophrenia. STUDY DESIGN Original studies involving humans and animals were included. The electronic databases PsycINFO, EMBASE, Web of Science, PubMed/MEDLINE, and Cochrane were systematically searched and quantitative analysis performed. STUDY RESULTS Sixteen original studies met inclusion criteria (1376 participants: 748 cases and 628 controls). Ten were included in the meta-analysis. Although observed species and Chao 1 show a decrease in diversity in people with schizophrenia compared with controls (SMD = -0.14 and -0.66 respectively), that did not reach statistical significance. We did not find evidence for variations in richness or evenness of microbiota between patients and controls overall. Differences in beta diversity and consistent patterns in microbial taxa were noted across studies. We found increases in Bifidobacterium, Lactobacillus, and Megasphaera in schizophrenia groups. Variations in brain structure, metabolic pathways, and symptom severity may be associated with compositional alterations in the microbiome. The heterogeneous design of studies complicates a similar evaluation of functional readouts. CONCLUSIONS The microbiome may play a role in the etiology and symptomatology of schizophrenia. Understanding how the implications of alterations in microbial genes for symptomatic expression and clinical outcomes may contribute to the development of microbiome targeted interventions for psychosis.
Collapse
Affiliation(s)
- Nuala Murray
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Sukainah Al Khalaf
- School of Public Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - David Kaulmann
- School of Public Health, University College Cork, Cork, Ireland
| | - Edgar Lonergan
- RISE, Early Intervention in Psychosis Service, South Lee Mental Health Services, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| | - Karen O’Connor
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
- RISE, Early Intervention in Psychosis Service, South Lee Mental Health Services, Cork, Ireland
| |
Collapse
|
33
|
Xing M, Gao H, Yao L, Wang L, Zhang C, Zhu L, Cui D. Profiles and diagnostic value of intestinal microbiota in schizophrenia patients with metabolic syndrome. Front Endocrinol (Lausanne) 2023; 14:1190954. [PMID: 37576972 PMCID: PMC10415044 DOI: 10.3389/fendo.2023.1190954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/26/2023] [Indexed: 08/15/2023] Open
Abstract
Aims/hypothesis It is widely thought that the intestinal microbiota plays a significant role in the pathogenesis of metabolic disorders. However, the gut microbiota composition and characteristics of schizophrenia patients with metabolic syndrome (MetS) have been largely understudied. Herein, we investigated the association between the metabolic status of mainland Chinese schizophrenia patients with MetS and the intestinal microbiome. Methods Fecal microbiota communities from 115 male schizophrenia patients (57 with MetS and 58 without MetS) were assessed by 16S ribosomal RNA gene sequencing. We assessed the variations of gut microbiome between both groups and explored potential associations between intestinal microbiota and parameters of MetS. In addition, the Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) based on the KEGG database was used to predict the function of intestinal microbiota. We also conducted Decision Tree Analysis to develop a diagnostic model for the MetS in patients with schizophrenia based on the composition of intestinal microbiota. Results The fecal microbial diversity significantly differed between groups with or without MetS (α-diversity (Shannon index and Simpson index): p=0.0155, p=0.0089; β-diversity: p=0.001). Moreover, the microbial composition was significantly different between the two groups, involving five phyla and 38 genera (p<0.05). In addition, a significant correlation was observed between the metabolic-related parameters and abundance of altered microbiota including HDL-c (r2 = 0.203, p=0.0005), GLU (r2 = 0.286, p=0.0005) and WC (r2 = 0.061, p=0.037). Furthermore, KEGG pathway analysis showed that 16 signaling pathways were significantly enriched between the two groups (p<0.05). Importantly, our diagnostic model based on five microorganisms established by decision tree analysis could effectively distinguish between patients with and without MetS (AUC = 0.94). Conclusions/interpretation Our study established the compositional and functional characteristics of intestinal microbiota in schizophrenia patients with MetS. These new findings provide novel insights into a better understanding of this disease and provide the theoretical basis for implementing new interventional therapies in clinical practice.
Collapse
Affiliation(s)
- Mengjuan Xing
- Department of General Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hui Gao
- The First Minzheng Mental Health Center, Shanghai, China
| | - Lili Yao
- The First Minzheng Mental Health Center, Shanghai, China
| | - Li Wang
- The First Minzheng Mental Health Center, Shanghai, China
| | - Chengfang Zhang
- Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, China
| | - Liping Zhu
- Department of General Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Donghong Cui
- Department of General Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
34
|
Nuncio-Mora L, Lanzagorta N, Nicolini H, Sarmiento E, Ortiz G, Sosa F, Genis-Mendoza AD. The Role of the Microbiome in First Episode of Psychosis. Biomedicines 2023; 11:1770. [PMID: 37371865 DOI: 10.3390/biomedicines11061770] [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: 03/11/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The relationship between the gut-brain-microbiome axis has gained great importance in the study of psychiatric disorders, as it may represent a new target for their treatment. To date, the available literature suggests that the microbiota may influence the pathophysiology of several diseases, including psychosis. The aim of this review is to summarize the clinical and preclinical studies that have evaluated the differences in microbiota as well as the metabolic consequences related to psychosis. Current data suggest that the genera Lactobacillus and Megasphaera are increased in schizophrenia (SZ), as well as alterations in the glutamate-glutamine-GABA cycle, serum levels of tryptophan, kynurenic acid (KYNA), and short-chain fatty acids (SCFAs). There are still very few studies on early-onset psychosis, thus more studies are needed to be able to propose targeted therapies for a point when the disease has just started or has not yet progressed.
Collapse
Affiliation(s)
- Lucero Nuncio-Mora
- Laboratory of Genomics of Psychiatric and Neurodegenerative Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Posgraduate Studies in Biological Sciences, Posgraduate Unit, Posgraduate Circuit, Universitary City, Building D, 1st Floor, Coyoacan, Mexico City 04510, Mexico
| | | | - Humberto Nicolini
- Laboratory of Genomics of Psychiatric and Neurodegenerative Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Carraci Medical Group, Mexico City 03740, Mexico
| | - Emmanuel Sarmiento
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| | - Galo Ortiz
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| | - Fernanda Sosa
- Carraci Medical Group, Mexico City 03740, Mexico
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| | - Alma Delia Genis-Mendoza
- Laboratory of Genomics of Psychiatric and Neurodegenerative Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| |
Collapse
|
35
|
Yu L, Gao Y, Ye Z, Duan H, Zhao J, Zhang H, Narbad A, Tian F, Zhai Q, Chen W. Interaction of beta-glucans with gut microbiota: Dietary origins, structures, degradation, metabolism, and beneficial function. Crit Rev Food Sci Nutr 2023; 64:9884-9909. [PMID: 37272431 DOI: 10.1080/10408398.2023.2217727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Beta-glucan (BG), a polysaccharide comprised of interfacing glucose monomers joined via beta-glycosidic linkages, can be defined as a type of dietary fiber with high specificity based on its interaction with the gut microbiota. It can induce similar interindividual microbiota responses, thereby having beneficial effects on the human body. In this paper, we review the four main sources of BG (cereals, fungi, algae, and bacteria) and their differences in structure and content. The interaction of BG with gut microbiota and the resulting health effects have been highlighted, including immune enhancement, regulation of serum cholesterol and insulin levels, alleviation of obesity and improvement of cognitive disorders. Finally, the application of BG in food products and its beneficial effects on the gut microbiota of consumers were discussed. Although some of the mechanisms of action remain unclear, revealing the beneficial functions of BG from the perspective of gut microbiota can help provide theoretical support for the development of diets that target the regulation of microbiota.
Collapse
Affiliation(s)
- Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuhang Gao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zi Ye
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hui Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Arjan Narbad
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- Gut Health and Microbiome Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
36
|
Amdanee N, Shao M, Hu X, Fang X, Zhou C, Chen J, Ridwan Chattun M, Wen L, Pan X, Zhang X, Xu Y. Serum Metabolic Profile in Schizophrenia Patients With Antipsychotic-Induced Constipation and Its relationship With Gut Microbiome. Schizophr Bull 2023; 49:646-658. [PMID: 36723169 PMCID: PMC10154739 DOI: 10.1093/schbul/sbac202] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND HYPOTHESIS Antipsychotics (APs), the cornerstone of schizophrenia treatment, confer a relatively high risk of constipation. However, the mechanisms underpinning AP-induced constipation are poorly understood. Thus, we hypothesized that (1) schizophrenia patients with AP-induced constipation have distinct metabolic patterns; (2) there is more than one mechanism at play in producing this adverse drug effect; and (3) AP-associated changes in the gut microbiome are related to the altered metabolic profiles. STUDY DESIGN Eighty-eight schizophrenia patients, including 44 with constipation (C) and 44 matched patients without constipation (NC), were enrolled in this study. Constipation was diagnosed by Rome IV criteria for constipation and colonic transit time using radiopaque markers (ROMs) while severity was evaluated with the Bristol Stool Form Scale (BSS) and Constipation Assessment Scale (CAS). Fasting blood samples were drawn from all participants and were subjected to non-targeted liquid chromatography-mass spectrometry (LC-MS) metabolomic analysis. STUDY RESULTS Eleven metabolites were significantly altered in AP-induced constipation which primarily disturbed sphingolipid metabolism, choline metabolism, and sphingolipid signaling pathway (P value < .05, FDR < 0.05). In the C group, changes in the gut bacteria showed a certain degree of correlation with 2 of the significantly altered serum metabolites and were associated with alterations in choline metabolism. CONCLUSIONS Our findings indicated that there were disturbances in distinct metabolic pathways that were associated with AP-induced constipation. In addition, this study presents evidence of a link between alterations in the gut microbiome and host metabolism which provides additional mechanistic insights on AP-induced constipation.
Collapse
Affiliation(s)
- Nousayhah Amdanee
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Miaomiao Shao
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Psychiatry, The Second People’s Hospital of Jiangning District, Nanjing, Jiangsu, China
| | - Xiuxiu Hu
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Psychiatry, The Second People’s Hospital of Jiangning District, Nanjing, Jiangsu, China
| | - Xinyu Fang
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chao Zhou
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Mohammad Ridwan Chattun
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Lu Wen
- Department of Psychiatry, The Second People’s Hospital of Jiangning District, Nanjing, Jiangsu, China
| | - Xinming Pan
- Department of Psychiatry, The Second People’s Hospital of Jiangning District, Nanjing, Jiangsu, China
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yue Xu
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Li Z, Qing Y, Cui G, Li M, Liu T, Zeng Y, Zhou C, Hu X, Jiang J, Wang D, Gao Y, Zhang J, Cai C, Wang T, Wan C. Shotgun metagenomics reveals abnormal short-chain fatty acid-producing bacteria and glucose and lipid metabolism of the gut microbiota in patients with schizophrenia. Schizophr Res 2023; 255:59-66. [PMID: 36965360 DOI: 10.1016/j.schres.2023.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/11/2023] [Accepted: 03/03/2023] [Indexed: 03/27/2023]
Abstract
Evidence has shown that the gut microbiota is closely related to the pathogenesis of schizophrenia, but temporal changes in the gut microbiota of patients with schizophrenia (SZ) during treatment remain unclear. Here, to evaluate temporal changes in the gut microbiota in schizophrenia, we performed whole-genome shotgun metagenomics on fecal samples from 36 healthy controls (HCs) and 19 baseline-period patients, and followed up with patients upon treatment. Compared to that in HCs, beta diversity in SZ was significantly distinct. The genera Bacteroides, Prevotella and Clostridium were the top 3 altered genera between SZ and HCs, and the Bacteroides-Prevotella ratio was significantly increased in SZ. Thirty-three percent of differentially abundant species were short-chain fatty acid (SCFA)-producing bacteria. Functional analysis showed that glucose and lipid metabolism of the gut microbiota was decreased in SZ compared with those in HCs. The abundances of two rate-limiting enzymes in glucose and lipid metabolism, phosphofructokinase (PFK) and acetyl-CoA carboxylase (ACC), were significantly decreased in SZ, and differentially abundant metabolism-related enzymes were significantly associated with SCFA-producing bacteria. Next, we found that the abundance of SCFA-producing bacteria also changed after treatment and that Clostridium was significantly negatively correlated with the total positive and negative syndrome scale (PANSS) score in patients. Functional analysis showed that glycoside hydrolase family 30 incrementally increased in abundance during treatment and were significantly associated with SCFA-producing bacteria. Our findings help to provide evidence for the role of gut microbiota in the occurrence and development of schizophrenia.
Collapse
Affiliation(s)
- Zhuyun Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Qing
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Gaoping Cui
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Minghui Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Tiantian Liu
- Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China; SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yanyan Zeng
- Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China; SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, China
| | - Chao Zhou
- Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China; SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Jiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Dandan Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Gao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Changqun Cai
- The Fourth People's Hospital of Wuhu, Wuhu, China
| | - Tao Wang
- Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China; SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, China.
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
38
|
Wu H, Liu Y, Wang J, Chen S, Xie L, Wu X. Schizophrenia and obesity: May the gut microbiota serve as a link for the pathogenesis? IMETA 2023; 2:e99. [PMID: 38868440 PMCID: PMC10989809 DOI: 10.1002/imt2.99] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 06/14/2024]
Abstract
Schizophrenia (SZ) places a tremendous burden on public health as one of the leading causes of disability and death. SZ patients are more prone to developing obesity than the general population from the clinical practice. The development of obesity frequently causes poor psychiatric outcomes in SZ patients. In turn, maternal obesity during pregnancy has been associated with an increased risk of SZ in offspring, suggesting that these two disorders may have shared neuropathological mechanisms. The gut microbiota is well known to serve as a major regulator of bidirectional interactions between the central nervous system and the gastrointestinal tract. It also plays a critical role in maintaining physical and mental health in humans. Recent studies have shown that the dysbiosis of gut microbiota is intimately associated with the onset of SZ and obesity through shared pathophysiological mechanisms, particularly the stimulation of immune inflammation. Therefore, gut microbiota may serve as a common biological basis for the etiology in both SZ and obesity, and the perturbed gut-brain axis may therefore account for the high prevalence of obesity in patients with SZ. On the basis of these findings, this review provides updated perspectives and intervention approaches on the etiology, prevention, and management of obesity in SZ patients by summarizing the recent findings on the role of gut microbiota in the pathogenesis of SZ and obesity, highlighting the role of gut-derived inflammation.
Collapse
Affiliation(s)
- Hui Wu
- Psychiatry DepartmentThird Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Yaxi Liu
- Psychiatry DepartmentThird Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Jie Wang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of MicrobiologyGuangdong Academy of SciencesGuangzhouChina
- Department of Life SciencesImperial College LondonLondonUnited Kingdom
| | - Shengyun Chen
- Psychiatry DepartmentThird Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of MicrobiologyGuangdong Academy of SciencesGuangzhouChina
| | - Xiaoli Wu
- Psychiatry DepartmentThird Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
39
|
Liu G, Gu K, Liu X, Jia G, Zhao H, Chen X, Wang J. Dietary glutamate enhances intestinal immunity by modulating microbiota and Th17/Treg balance-related immune signaling in piglets after lipopolysaccharide challenge. Food Res Int 2023; 166:112597. [PMID: 36914323 DOI: 10.1016/j.foodres.2023.112597] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/03/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
The purpose of this study was to explore the effects of glutamate on piglet growth performance and intestinal immunity function, and to further elucidate its mechanism. In a 2 × 2 factorial design involving immunological challenge (lipopolysaccharide (LPS) or saline) and diet (with or without glutamate), twenty-four piglets were randomly assigned to four groups, each with 6 replicates. Piglets were fed with a basal or glutamate diet for 21 d before being injected intraperitoneally with LPS or saline. Piglet's intestinal samples were collected 4 h after injection. Results showed that glutamate increased daily feed intake, average daily gain, villus length, villus area, and villus length to crypt depth ratio (V/C), and decreased the crypt depth (P < 0.05). Furthermore, glutamate increased the mRNA expression of forkhead box P3 (FOXP3), a signal transducer and activator of transcription 5 (STAT5) and transforming growth factor beta, while decreasing the mRNA expression of RAR-related orphan receptor c and STAT3. Glutamate increased interleukin-10 (IL-10) mRNA expression while decreasing the mRNA expression of IL-1β, IL-6, IL-8, IL-17, IL-21, and tumor necrosis factor-α. At the phylum level, glutamate increased the Actinobacteriota abundance and Firmicutes-to-Bacteroidetes ratio while decreasing Firmicutes abundance. At the genus level, glutamate improved the abundance of beneficial bacteria (e.g., Lactobacillus, Prevotellaceae-NK3B31-group, and UCG-005). Furthermore, glutamate increased the concentrations of short-chain fatty acids (SCFAs). Correlation analysis revealed that the intestinal microbiota is closely related to Th17/Treg balance-related index and SCFAs. Collectively, glutamate can improve piglet growth performance and intestinal immunity by modulating gut microbiota and Th17/Treg balance-related signaling pathways.
Collapse
Affiliation(s)
- Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China.
| | - Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China
| | - Xinlian Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu 611130, Sichuan, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| |
Collapse
|
40
|
Iseli GC, Ulrich S, Schmidt A. Elucidating gut microbiota-hippocampus interactions in emerging psychosis: A new perspective for the development of early interventions for memory impairments. Front Psychiatry 2023; 14:1098019. [PMID: 37032923 PMCID: PMC10076719 DOI: 10.3389/fpsyt.2023.1098019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Hippocampal dysregulation might be a key pathophysiological factor for memory impairments in psychosis. Contemporary models particularly postulate that an imbalance of hippocampal glutamate and GABA leads to impaired memory and may thus serve as a therapeutic target to improve memory deficits. However, currently available interventions in early stages of psychosis do not explicitly target hippocampal pathology. A novel approach for manipulating hippocampus-dependent memory processes is provided via the gut microbiota. In this perspective article, we first recapitulate compelling evidence for emerging hippocampus pathology during the development of psychosis. The following sections emphasize the critical role of the gut microbiota in hippocampus plasticity and memory, and summarize existing evidence of gut microbiota alterations in different stages of psychosis. Finally, we propose a novel conceptual roadmap for future studies deciphering gut microbiota-hippocampus synergisms in emerging psychosis and argue that specific microbial supplementation might be promising for improving hippocampus-dependent memory deficits in early stages of psychosis.
Collapse
Affiliation(s)
| | | | - André Schmidt
- Department of Clinical Research (DKF), University Psychiatric Clinics (UPK), Translational Neurosciences, University of Basel, Basel, Switzerland
| |
Collapse
|
41
|
Zhang C, Liu T, Wang Y, Chen W, Liu J, Tao J, Zhang Z, Zhu X, Zhang Z, Ming M, Wang M, Lu G, Yan G. Metagenomic next-generation sequencing of bronchoalveolar lavage fluid from children with severe pneumonia in pediatric intensive care unit. Front Cell Infect Microbiol 2023; 13:1082925. [PMID: 37009495 PMCID: PMC10064343 DOI: 10.3389/fcimb.2023.1082925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/11/2023] [Indexed: 03/19/2023] Open
Abstract
BackgroundSevere pneumonia due to lower respiratory tract infections (LRTIs) is a significant cause of morbidity and mortality in children. Noninfectious respiratory syndromes resembling LRTIs can complicate the diagnosis and may also make targeted therapy difficult because of the difficulty of identifying LRTI pathogens. In the present study, a highly sensitive metagenomic next-generation sequencing (mNGS) approach was used to characterize the microbiome of bronchoalveolar lavage fluid (BALF) in children with severe lower pneumonia and identify pathogenic microorganisms that may cause severe pneumonia. The purpose of this study was to use mNGS to explore the potential microbiomes of children with severe pneumonia in a PICU.MethodsWe enrolled patients meeting diagnostic criteria for severe pneumonia admitted at PICU of the Children’s Hospital of Fudan University, China, from February 2018 to February 2020. In total, 126 BALF samples were collected, and mNGS was performed at the DNA and/or RNA level. The pathogenic microorganisms in BALF were identified and correlated with serological inflammatory indicators, lymphocyte subtypes, and clinical symptoms.ResultsmNGS of BALF identified potentially pathogenic bacteria in children with severe pneumonia in the PICU. An increased BALF bacterial diversity index was positively correlated with serum inflammatory indicators and lymphocyte subtypes. Children with severe pneumonia in the PICU had the potential for coinfection with viruses including Epstein–Barr virus, Cytomegalovirus, and Human betaherpesvirus 6B, the abundance of which was positively correlated with immunodeficiency and pneumonia severity, suggesting that the virus may be reactivated in children in the PICU. There was also the potential for coinfection with fungal pathogens including Pneumocystis jirovecii and Aspergillus fumigatus in children with severe pneumonia in the PICU, and an increase in potentially pathogenic eukaryotic diversity in BALF was positively associated with the occurrence of death and sepsis.ConclusionsmNGS can be used for clinical microbiological testing of BALF samples from children in the PICU. Bacterial combined with viral or fungal infections may be present in the BALF of patients with severe pneumonia in the PICU. Viral or fungal infections are associated with greater disease severity and death.
Collapse
Affiliation(s)
- Caiyan Zhang
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Tingyan Liu
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Yixue Wang
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Weiming Chen
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Jing Liu
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Jinhao Tao
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Zhengzheng Zhang
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Xuemei Zhu
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Zhenyu Zhang
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Meixiu Ming
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
| | - Mingbang Wang
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children’s Hospital of Fudan University, National Center for Children’s Health, Shanghai, China
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
- *Correspondence: Gangfeng Yan, ; Guoping Lu, ; Mingbang Wang,
| | - Guoping Lu
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
- *Correspondence: Gangfeng Yan, ; Guoping Lu, ; Mingbang Wang,
| | - Gangfeng Yan
- Paediatric Intensive Care Unit, Children’s Hospital of Fudan University, Shanghai, China
- *Correspondence: Gangfeng Yan, ; Guoping Lu, ; Mingbang Wang,
| |
Collapse
|
42
|
Nita IB, Ilie OD, Ciobica A, Hritcu LD, Dobrin I, Doroftei B, Dobrin R. Reviewing the Potential Therapeutic Approaches Targeting the Modulation of Gastrointestinal Microflora in Schizophrenia. Int J Mol Sci 2022; 23:ijms232416129. [PMID: 36555774 PMCID: PMC9784651 DOI: 10.3390/ijms232416129] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Schizophrenia (SCZ) is a severe brain disorder characterized by an intriguing clinical panel that has begun to gain interest due to its particular phenotype. Having considered the role of gut microflora in psychiatry, the latest discoveries might offer further insight into the underlying mechanisms. Thus, we aimed to offer an updated overview of the therapeutic potential of microorganism-derived supplements alongside dedicated protocols that target the re-establishment of the host's eubiosis. Based on combinations of specific keywords, we performed searches in four databases (PubMed/Medline, ISI Web of Knowledge, Scopus, and ScienceDirect) for the established interval (2018-2022) and identified twenty two eligible cases, restricted only to human patients' experiences. Up until the writing of this manuscript, it has been revealed that the administration of specific lactic acid bacteria strains (Lactobacillus and Bifidobacterium), or those combined with vitamin D and selenium, maintain the integrity of the gut flora, preventing antagonistic effects including inflammation, antipsychotic-related body weight gain (olanzapine) and other metabolic dysfunctionalities. However, there are multiple antipsychotics that exert a potent effect upon gut flora, influencing a plethora of pathways and creating a dysbalance ratio between beneficial and opportunistic pathogens. Risperidone, amisulpride, and clozapine are just a few examples, but the current literature is unfortunately inconsistent and reported data is contradictory, which is why we support additional studies in this context. Moreover, we further argue the utility of studying how distinct controlled substances influence microbial communities, considering that ketamine is proved to alleviate depressive-like behavior as opposed to amphetamine and phencyclidine, which are known substances to trigger SCZ-like symptoms in experimental models. Probiotics may be regarded as the most consequential vehicle through which the gut flora can be successfully influenced, in adequate doses exerting a beneficial role as an alternative approach to alleviate SCZ symptoms.
Collapse
Affiliation(s)
- Ilinca-Bianca Nita
- Department of Medicine III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| | - Ovidiu-Dumitru Ilie
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Luminita-Diana Hritcu
- Internal Medicine Clinic, Faculty of Veterinary Medicine, University of Life Sciences “Ion Ionescu de la Brad”, Mihail Sadoveanu Street, no 3, 700490 Iasi, Romania
- Correspondence: (L.-D.H.); (B.D.)
| | - Irina Dobrin
- Department of Medicine III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Institute of Psychiatry “Socola”, Bucium Street, no 36, 700282 Iasi, Romania
| | - Bogdan Doroftei
- Department of Medicine VIII, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Correspondence: (L.-D.H.); (B.D.)
| | - Romeo Dobrin
- Department of Medicine III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Institute of Psychiatry “Socola”, Bucium Street, no 36, 700282 Iasi, Romania
| |
Collapse
|
43
|
Gut microbiota alterations in stable outpatients with schizophrenia: findings from a case-control study. Acta Neuropsychiatr 2022; 35:147-155. [PMID: 36503629 DOI: 10.1017/neu.2022.38] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The pathogenesis of schizophrenia is multidimensional and intensively studied. The gut-brain axis disturbances might play a significant role in the development of schizophrenia. METHODS We compared the gut microbiota of 53 individuals with schizophrenia and 58 healthy controls, using the 16S rRNA sequencing method. Individuals with schizophrenia were assessed using the following scales: the Positive and Negative Syndrome Scale, the Calgary Depression Scale for Schizophrenia, the Social and Occupational Functioning Assessment Scale and the Repeatable Battery for the Assessment of Neuropsychological Status. RESULTS No significant between-group differences in α-diversity measures were observed. Increased abundance of Lactobacillales (order level), Bacilli (class level) and Actinobacteriota (phylum level) were found in individuals with schizophrenia regardless of potential confounding factors, and using two independent analytical approaches (the distance-based redundancy analysis and the generalised linear model analysis). Additionally, significant correlations between various bacterial taxa (the Bacteroidia class, the Actinobacteriota phylum, the Bacteroidota phylum, the Coriobacteriales order and the Coriobacteria class) and clinical manifestation (the severity of negative symptoms, performance of language abilities, social and occupational functioning) were observed. CONCLUSIONS The present study indicates that gut microbiota alterations are present in European patients with schizophrenia. The abundance of certain bacterial taxa might be associated with the severity of negative symptoms, cognitive performance and general functioning. Nonetheless, additional studies are needed before the translation of our results into clinical practice.
Collapse
|
44
|
Liu A, Cai C, Wang Z, Wang B, He J, Xie Y, Deng H, Liu S, Zeng S, Yin Z, Wang M. Inductively coupled plasma mass spectrometry based urine metallome to construct clinical decision models for autism spectrum disorder. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6849992. [PMID: 36442146 DOI: 10.1093/mtomcs/mfac091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The global prevalence of autism spectrum disorder (ASD) is on the rise, and high levels of exposure to toxic heavy metals may be associated with this increase. Urine analysis is a noninvasive method for investigating the accumulation and excretion of heavy metals. The aim of this study was to identify ASD-associated urinary metal markers. METHODS Overall, 70 children with ASD and 71 children with typical development (TD) were enrolled in this retrospective case-control study. In this metallomics investigation, inductively coupled plasma mass spectrometry was performed to obtain the urine profile of 27 metals. RESULTS Children with ASD could be distinguished from children with TD based on the urine metal profile, with ASD children showing an increased urine metal Shannon diversity. A metallome-wide association analysis was used to identify seven ASD-related metals in urine, with cobalt, aluminum, selenium, and lithium significantly higher, and manganese, mercury, and titanium significantly lower in the urine of children with ASD than in children with TD. The least absolute shrinkage and selection operator (LASSO) machine learning method was used to rank the seven urine metals in terms of their effect on ASD. On the basis of these seven urine metals, we constructed a LASSO regression model for ASD classification and found an area under the receiver operating characteristic curve of 0.913. We also constructed a clinical prediction model for ASD based on the seven metals that were different in the urine of children with ASD and found that the model would be useful for the clinical prediction of ASD risk. CONCLUSIONS The study findings suggest that altered urine metal concentrations may be an important risk factor for ASD, and we recommend further exploration of the mechanisms and clinical treatment measures for such alterations.
Collapse
Affiliation(s)
- Aiping Liu
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Chunquan Cai
- Tianjin Pediatric Research Institute, Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People's Hospital, Guangdong 518109, China
| | - Bin Wang
- The department of Dermatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Juntao He
- Shenzhen Prevention and Treatment Center for Occupational Diseases (Physical Testing & Chemical Analysis Department), Shenzhen 518020, China
| | - Yanhong Xie
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Honglian Deng
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Shaozhi Liu
- T he department of Laboratory, Baoan Public Health Service Center of Shenzhen, Baoan District, Shenzhen, 518108, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Guangdong 518116, China
| | - Zhaoqing Yin
- Division of Pediatrics, The People's Hospital of Dehong Autonomous Prefecture, Dehong Hospital of Kunming Medical University, Mangshi, Yunnan 678400, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China.,Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai 201102, China
| |
Collapse
|
45
|
Fan X, Deng H, Qiu J, Ji H, Shen X. Antibiotics-induced depression in mice via the microbiota-gut-brain axis. J Affect Disord 2022; 318:152-158. [PMID: 36075400 DOI: 10.1016/j.jad.2022.08.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 05/04/2022] [Accepted: 08/21/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Intestinal dysbacteriosis is associated with depression. This study aimed to establish an antibiotics-induced depression mouse model and explore the mechanism of antibiotic-induced depression. METHODS C57BL/6 J mice were treated with antibiotics to prepare the antibiotic-induced depression mouse model. Behavioral tests and depression-related bio-markers were examined. To understand the abundance of different bacteria in intestinal flora and screen out the predominant bacterial species, metagenomic analysis of feces was carried out. Finally, we detected the expression of NF-κB-p65 and p-NF-κB-p65 in PFC and the hippocampus using Western blot. RESULTS Mixtures A and B caused depression-like behavior in mice. Norepinephrine, 5-hydroxytryptamine, and brain-derived neurotrophic factor in hippocampus and PFC of antibiotic-induced depression mice significantly decreased. The serum adrenocorticotropic hormone and corticosterone concentrations increased. The abundance values of Bacteroides thetaiotaomicron, Klebsiella oxytoca, and Klebsiella aerogenes in antibiotic-induced depression mice significantly increased, and the characteristic KO genes and metabolic pathways in antibiotic-induced depression mice were significantly different with in CUMS depression mice (the positive control) and normal mice. The relative levels of p-NF-κB-p65 in antibiotics-induced depression mice were significantly higher than in normal mice. LIMITATIONS How dysbacteriosis induces inflammation in the central nervous system is unclear. CONCLUSIONS Specific antibiotic mixture can cause depression-like behavior and changes of depression-related bio-markers in mice. The antibiotic-induced depression mice display changes in the species and metabolism of intestinal bacterial flora. The activation of NF-κB inflammatory signaling pathway in the central nervous system may act as one of the mechanisms in the development of antibiotic-induced depression.
Collapse
Affiliation(s)
- Xingli Fan
- Departments of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China.
| | - Handan Deng
- Departments of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Jianing Qiu
- Departments of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Hua Ji
- Departments of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China.
| | - Xiangdi Shen
- Departments of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
46
|
Yan F, Xia L, Xu L, Deng L, Jin G. A comparative study to determine the association of gut microbiome with schizophrenia in Zhejiang, China. BMC Psychiatry 2022; 22:731. [PMID: 36424595 PMCID: PMC9694861 DOI: 10.1186/s12888-022-04328-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/14/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND With the rapid progress of high-throughput sequencing technology, characterization of schizophrenia (SZ) with underlying probing of the gut microbiome can explore pathogenic mechanisms, estimate disease risk, and allow customization of therapeutic and prophylactic modalities. In this study, we compared the differences in gut microbial diversity and composition between 50 SZ subjects and 50 healthy matched subjects in Zhejiang, China via targeted next-generation sequencing (16S rRNA amplicon). RESULTS Accordingly, the alpha diversity indices (observed species index, Shannon index, and Simpson index) of the gut microbiome in the healthy control group were higher than those in the SZ group. Additionally, principal coordinate analysis and non-metric multidimensional scaling of beta diversity revealed that patients with SZ clustered more tightly than healthy controls. At the phylum level, we found that the abundance of Bacteroidetes and Proteobacteria in the SZ group was significantly increased. At the genus level, the relative abundances of Prevotella, Parabacteroides, and Sutterella were significantly higher, whereas the abundances of Faecalibacterium, Blautia, Lachnospira, Clostridium, Ruminococcus, and Coprococcus were lower than those in the healthy control group. Further analyses revealed that Succinivibrio, Megasphaera, and Nesterenkonia may serve as potential biomarkers for distinguishing patients with SZ from those in the control cohort. CONCLUSIONS This study profiled differences in gut microbiome diversity, taxonomic composition, and function between SZ and healthy cohorts, and the insights from this research could be used to develop targeted next-generation sequencing-based diagnoses for SZ.
Collapse
Affiliation(s)
- Fuyang Yan
- The Second People’s Hospital of Lishui, Lishui, 323000 China
| | - Lehong Xia
- The Second People’s Hospital of Lishui, Lishui, 323000 China
| | - Li Xu
- The Second People’s Hospital of Lishui, Lishui, 323000 China
| | - Liyun Deng
- The Second People’s Hospital of Lishui, Lishui, 323000 China
| | - Guolin Jin
- The Second People's Hospital of Lishui, Lishui, 323000, China.
| |
Collapse
|
47
|
D'Alessandro G, Marrocco F, Limatola C. Microglial cells: Sensors for neuronal activity and microbiota-derived molecules. Front Immunol 2022; 13:1011129. [PMID: 36426369 PMCID: PMC9679421 DOI: 10.3389/fimmu.2022.1011129] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/24/2022] [Indexed: 11/26/2023] Open
Abstract
Microglial cells play pleiotropic homeostatic activities in the brain, during development and in adulthood. Microglia regulate synaptic activity and maturation, and continuously patrol brain parenchyma monitoring for and reacting to eventual alterations or damages. In the last two decades microglia were given a central role as an indicator to monitor the inflammatory state of brain parenchyma. However, the recent introduction of single cell scRNA analyses in several studies on the functional role of microglia, revealed a not-negligible spatio-temporal heterogeneity of microglial cell populations in the brain, both during healthy and in pathological conditions. Furthermore, the recent advances in the knowledge of the mechanisms involved in the modulation of cerebral activity induced by gut microbe-derived molecules open new perspectives for deciphering the role of microglial cells as possible mediators of these interactions. The aim of this review is to summarize the most recent studies correlating gut-derived molecules and vagal stimulation, as well as dysbiotic events, to alteration of brain functioning, and the contribution of microglial cells.
Collapse
Affiliation(s)
- Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Laboratory affiliated to Pasteur Italy, University of Rome La Sapienza, Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Francesco Marrocco
- Department of Physiology and Pharmacology, Laboratory affiliated to Pasteur Italy, University of Rome La Sapienza, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Laboratory affiliated to Pasteur Italy, University of Rome La Sapienza, Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| |
Collapse
|
48
|
Kanlioz M, Ekici U, Ferhatoğlu MF. Total Gastrointestinal Flora Transplantation in the Treatment of Leaky Gut Syndrome and Flora Loss. Cureus 2022; 14:e31071. [DOI: 10.7759/cureus.31071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 11/05/2022] Open
|
49
|
O’Donnell M, Teasdale SB, Chua XY, Hardman J, Wu N, Curtis J, Samaras K, Bolton P, Morris MJ, Shannon Weickert C, Purves-Tyson T, El-Assaad F, Jiang XT, Hold GL, El-Omar E. The Role of the Microbiome in the Metabolic Health of People with Schizophrenia and Related Psychoses: Cross-Sectional and Pre-Post Lifestyle Intervention Analyses. Pathogens 2022; 11:1279. [PMID: 36365032 PMCID: PMC9695516 DOI: 10.3390/pathogens11111279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 03/20/2024] Open
Abstract
The microbiome has been implicated in the development of metabolic conditions which occur at high rates in people with schizophrenia and related psychoses. This exploratory proof-of-concept study aimed to: (i) characterize the gut microbiota in antipsychotic naïve or quasi-naïve people with first-episode psychosis, and people with established schizophrenia receiving clozapine therapy; (ii) test for microbiome changes following a lifestyle intervention which included diet and exercise education and physical activity. Participants were recruited from the Eastern Suburbs Mental Health Service, Sydney, Australia. Anthropometric, lifestyle and gut microbiota data were collected at baseline and following a 12-week lifestyle intervention. Stool samples underwent 16S rRNA sequencing to analyse microbiota diversity and composition. Seventeen people with established schizophrenia and five people with first-episode psychosis were recruited and matched with 22 age-sex, BMI and ethnicity matched controls from a concurrent study for baseline comparisons. There was no difference in α-diversity between groups at baseline, but microbial composition differed by 21 taxa between the established schizophrenia group and controls. In people with established illness pre-post comparison of α-diversity showed significant increases after the 12-week lifestyle intervention. This pilot study adds to the current literature that detail compositional differences in the gut microbiota of people with schizophrenia compared to those without mental illness and suggests that lifestyle interventions may increase gut microbial diversity in patients with established illness. These results show that microbiome studies are feasible in patients with established schizophrenia and larger studies are warranted to validate microbial signatures and understand the relevance of lifestyle change in the development of metabolic conditions in this population.
Collapse
Affiliation(s)
- Maryanne O’Donnell
- Discipline of Psychiatry and Mental Health, School of Medicine and Health, University of New South Wales, Kensington 2033, Australia
- Eastern Suburbs Mental Health Service, South Eastern Sydney Local Health District, Randwick 2031, Australia
| | - Scott B. Teasdale
- Discipline of Psychiatry and Mental Health, School of Medicine and Health, University of New South Wales, Kensington 2033, Australia
- Mindgardens Neuroscience Network, Sydney 2033, Australia
| | - Xin-Yi Chua
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Kogarah 2217, Australia
| | - Jamie Hardman
- Eastern Suburbs Mental Health Service, South Eastern Sydney Local Health District, Randwick 2031, Australia
| | - Nan Wu
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Kogarah 2217, Australia
- Department of Gastroenterology, The Sutherland Hospital, Caringbah 2229, Australia
| | - Jackie Curtis
- Discipline of Psychiatry and Mental Health, School of Medicine and Health, University of New South Wales, Kensington 2033, Australia
- Eastern Suburbs Mental Health Service, South Eastern Sydney Local Health District, Randwick 2031, Australia
- Mindgardens Neuroscience Network, Sydney 2033, Australia
| | - Katherine Samaras
- Department of Endocrinology, St Vincent’s Hospital Sydney, Victoria St, Darlinghurst 2010, Australia
- Clinical Obesity, Nutrition and Adipose Biology Lab, 384 Garvan Institute of Medical Research, Victoria St, Darlinghurst 2010, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical Campus, University of New South Wales, Darlinghurst 2010, Australia
| | - Patrick Bolton
- Eastern Suburbs Mental Health Service, South Eastern Sydney Local Health District, Randwick 2031, Australia
- School of Public Health, University of New South Wales, Kensington 2033, Australia
| | - Margaret J. Morris
- School of Medical Sciences, University of New South Wales, Kensington 2033, Australia
| | - Cyndi Shannon Weickert
- Discipline of Psychiatry and Mental Health, School of Medicine and Health, University of New South Wales, Kensington 2033, Australia
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney 2033, Australia
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Tertia Purves-Tyson
- Discipline of Psychiatry and Mental Health, School of Medicine and Health, University of New South Wales, Kensington 2033, Australia
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney 2033, Australia
| | - Fatima El-Assaad
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Kogarah 2217, Australia
| | - Xiao-Tao Jiang
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Kogarah 2217, Australia
| | - Georgina L. Hold
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Kogarah 2217, Australia
| | - Emad El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Kogarah 2217, Australia
| |
Collapse
|
50
|
Zhang D, Liu J, Cheng H, Wang H, Tan Y, Feng W, Peng C. Interactions between polysaccharides and gut microbiota: A metabolomic and microbial review. Food Res Int 2022; 160:111653. [DOI: 10.1016/j.foodres.2022.111653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/17/2022]
|