1
|
Cotrin JC, Dos Santos Junior GC, Cadaxo AS, Pereira JS, Spitz M, de Rosso ALZ, Veras RP, Valente AP, Pimentel MMG, Santos-Rebouças CB. Plasma and urinary metabolomic signatures differentiate genetic and idiopathic Parkinson's disease. Brain Res 2025; 1858:149625. [PMID: 40204143 DOI: 10.1016/j.brainres.2025.149625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Parkinson's disease (PD) is marked by alpha-synuclein accumulation and progressive dopaminergic neuron loss. Using Nuclear Magnetic Resonance (NMR)-based metabolomics, we uncovered metabolic disturbances in idiopathic PD (iPD) and PD linked to LRRK2, GBA1, and PRKN variants in a Brazilian ethnically diverse cohort, free of comorbidities, in comparison to healthy, age-matched controls. In plasma, significant PD-associated metabolites included histidine, acetate, acetoacetate, glutamine, glucose, lipids and lipoproteins, N-acetyl-glycoproteins, and sarcosine. Urine samples revealed alterations in creatine, creatinine, L-asparagine, trimethylamine, 3-beta-hydroxybutyrate, isovaleric acid, glutamine, urea, glycine, choline, arginine, and cysteine in association with PD. Notably, creatine, creatinine, acetate, glucose, and histidine showed pathway influences from LRRK2, GBA1, and PRKN variants. Enrichment analyses highlighted disruptions in glyoxylate and dicarboxylate metabolism (plasma) as well as serine, threonine, and glycine metabolism (urine). Additionally, a metabolite-gene-disease interaction network identified 15 genes associated with PD that interact with key metabolites, highlighting MAPT, SNCA, RERE, and KCNN3 as key players in both plasmaandurine. NMR in saliva samples did not show significant differences between PD groups and controls. Our findings underscore PD-associated metabolites, particularly related to arginine metabolism, the urea cycle, glutamate metabolism, glucose metabolism, and gut microbiota. These pathways and gene interactions may serve as potential biomarkers for PD diagnosis and precision medicine strategies.
Collapse
Affiliation(s)
- Juliana Cordovil Cotrin
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Gilson Costa Dos Santos Junior
- Laboratory of Metabolomics, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - André Simões Cadaxo
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Joao Santos Pereira
- Movement Disorders Clinic, Neurology Service, Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Mariana Spitz
- Movement Disorders Clinic, Neurology Service, Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ana Lúcia Zuma de Rosso
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato Peixoto Veras
- Institute of Human Aging, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ana Paula Valente
- National Center of Nuclear Magnetic Resonance, Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Márcia Mattos Gonçalves Pimentel
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cíntia Barros Santos-Rebouças
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Zhang X, Chen W, Zhang H, Li Y, Han Y, Liu W, Liu Y, Wang X, Zhang X, Tian D, Wang X. Effects of Vibrio alginolyticus on intestinal health and intestinal flora of sea urchin (Strongylocentrotus intermedius). Comp Biochem Physiol B Biochem Mol Biol 2025; 278:111099. [PMID: 40250796 DOI: 10.1016/j.cbpb.2025.111099] [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: 12/17/2024] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The aim of this study was to understand the effect of Vibrio alginolyticus on the intestinal tract of Strongylocentrotus intermedius. The effects of injecting V. alginolyticus into the body cavity via the perioral membrane at concentrations of 0 CFU/mL (C), 1.5 × 107 CFU/mL (VA1), and 1.5 × 108 CFU/mL (VA10) on the intestinal tract of S. intermedius were analyzed using histological examination, immunoenzyme activity, and 16S rRNA sequencing. The results showed that V. alginolyticus caused intestinal tissue damage and oxidative stress (e.g. altered levels of superoxide dismutase). In addition, the intestinal flora was altered. At the phylum level, the abundance of Bacteroidota was significantly decreased in the VA10 group, at the genus level, Vibrio spp. exhibited a significant increase following V. alginolyticus injection. Prediction of Kyoto Encyclopedia of Genes and Genomes function in the intestinal flora revealed that high concentrations of V. alginolyticus may have induced pathways such as energy metabolism. These results indicated that V. alginolyticus caused lesions in the intestinal morphology of S. intermedius and disrupted the balance of intestinal flora.
Collapse
Affiliation(s)
- Xiaochen Zhang
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Wei Chen
- Yantai Marine Economic Research Institute, Yantai, Shandong 264003, China
| | - Haoyu Zhang
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Yan Li
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Yijing Han
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China.
| | - Wan Liu
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Yaqiong Liu
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Xiaona Wang
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Xuekai Zhang
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Deyang Tian
- Laizhou LiYang Aquatic Development Co., Ltd., Yantai 261441, China
| | - Xiaotong Wang
- School of Fisheries, Ludong University, Yantai, Shandong 264025, China.
| |
Collapse
|
3
|
López-Santamarina A, Cardelle-Cobas A, Mondragón Portocarrero ADC, Cepeda Sáez A, Miranda JM. Modulatory effects of red seaweeds (Palmaria palmata, Porphyra umbilicalis and Chondrus crispus) on the human gut microbiota via an in vitro model. Food Chem 2025; 476:143437. [PMID: 39987804 DOI: 10.1016/j.foodchem.2025.143437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
This work investigated the nutritional and mineral composition of three red seaweed species, Palmaria palmata, Porphyra umbilicalis and Chondrus crispus, and an in vitro assay was performed to determinate the effects of the intake of whole red seaweed on gut microbiota, short chain fatty acids production and metabolic pathways. The results obtained showed that 100 g of seaweeds contained essential minerals such as 28-107 % daily needs of Ca, 183-600 % daily needs of Fe and 18-54 % daily needs of Zn, whereas low content were found for Cu and I. Seaweed digestion fermentation showed beneficial effects of gut microbiota, as increases in beneficial species such as Akkermansia muciniphila, and in some seaweed, Bifidobacterium adoslescentis, Bacteroides ovatus or Lactobacillus ruminis. Metabolic pathways did only found little significant differences with respect to inulin fermentation. In view of the results, red seaweed showed prebiotic effects on human gut microbiota.
Collapse
Affiliation(s)
- Aroa López-Santamarina
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Universidade de Santiago de Compostela, Campus Terra, Lugo 27002, Spain
| | - Alejandra Cardelle-Cobas
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Universidade de Santiago de Compostela, Campus Terra, Lugo 27002, Spain
| | - Alicia Del Carmen Mondragón Portocarrero
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Universidade de Santiago de Compostela, Campus Terra, Lugo 27002, Spain
| | - Alberto Cepeda Sáez
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Universidade de Santiago de Compostela, Campus Terra, Lugo 27002, Spain
| | - Jose Manuel Miranda
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Universidade de Santiago de Compostela, Campus Terra, Lugo 27002, Spain.
| |
Collapse
|
4
|
Gao Y, Liu Y, Ma T, Liang Q, Sun J, Wu X, Song Y, Nie H, Huang J, Mu G. Fermented Dairy Products as Precision Modulators of Gut Microbiota and Host Health: Mechanistic Insights, Clinical Evidence, and Future Directions. Foods 2025; 14:1946. [PMID: 40509473 PMCID: PMC12154003 DOI: 10.3390/foods14111946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2025] [Revised: 05/24/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Dairy products-encompassing yogurt, kefir, cheese, and cultured milk beverages-are emerging as versatile, food-based modulators of gut microbiota and host physiology. This review synthesizes mechanistic insights demonstrating how live starter cultures and their fermentation-derived metabolites (short-chain fatty acids, bioactive peptides, and exopolysaccharides) act synergistically to enhance microbial diversity, reinforce epithelial barrier integrity via upregulation of tight-junction proteins, and modulate immune signaling. Clinical evidence supports significant improvements in metabolic parameters (fasting glucose, lipid profiles, blood pressure) and reductions in systemic inflammation across metabolic syndrome, hypertension, and IBS cohorts. We highlight critical modulatory factors-including strain specificity, host enterotypes and FUT2 genotype, fermentation parameters, and matrix composition-that govern probiotic engraftment, postbiotic yield, and therapeutic efficacy. Despite promising short-term outcomes, current studies are limited by heterogeneous designs and brief intervention periods, underscoring the need for long-term, adaptive trials and integrative multi-omics to establish durability and causality. Looking forward, precision nutrition frameworks that harness baseline microbiota profiling, host genetics, and data-driven fermentation design will enable bespoke fermented dairy formulations, transforming these traditional foods into next-generation functional matrices for targeted prevention and management of metabolic, inflammatory, and neuroimmune disorders.
Collapse
Affiliation(s)
- Yuan Gao
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Yanyan Liu
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Tingting Ma
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Qimeng Liang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Junqi Sun
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Xiaomeng Wu
- Dalian Probiotics Function Research Key Laboratory, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Yinglong Song
- Dalian Probiotics Function Research Key Laboratory, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Hui Nie
- Guangxi Key Laboratory of Health Care Food Science and Technology, Hezhou University, Hezhou 542899, China
| | - Jun Huang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Guangqing Mu
- Dalian Probiotics Function Research Key Laboratory, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
5
|
Belelli D, Lambert JJ, Wan MLY, Monteiro AR, Nutt DJ, Swinny JD. From bugs to brain: unravelling the GABA signalling networks in the brain-gut-microbiome axis. Brain 2025; 148:1479-1506. [PMID: 39716883 PMCID: PMC12074267 DOI: 10.1093/brain/awae413] [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] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 12/25/2024] Open
Abstract
Convergent data across species paint a compelling picture of the critical role of the gut and its resident microbiota in several brain functions and disorders. The chemicals mediating communication along these sophisticated highways of the brain-gut-microbiome (BGM) axis include both microbiota metabolites and classical neurotransmitters. Amongst the latter, GABA is fundamental to brain function, mediating most neuronal inhibition. Until recently, GABA's role and specific molecular targets in the periphery within the BGM axis had received limited attention. Yet, GABA is produced by neuronal and non-neuronal elements of the BGM, and recently, GABA-modulating bacteria have been identified as key players in GABAergic gut systems, indicating that GABA-mediated signalling is likely to transcend physiological boundaries and species. We review the available evidence to better understand how GABA facilitates the integration of molecularly and functionally disparate systems to bring about overall homeostasis and how GABA perturbations within the BGM axis can give rise to multi-system medical disorders, thereby magnifying the disease burden and the challenges for patient care. Analysis of transcriptomic databases revealed significant overlaps between GABAAR subunits expressed in the human brain and gut. However, in the gut, there are notable expression profiles for a select number of subunits that have received limited attention to date but could be functionally relevant for BGM axis homeostasis. GABAergic signalling, via different receptor subtypes, directly regulates BGM homeostasis by modulating the excitability of neurons within brain centres responsible for gastrointestinal (GI) function in a sex-dependent manner, potentially revealing mechanisms underlying the greater prevalence of GI disturbances in females. Apart from such top-down regulation of the BGM axis, a diverse group of cell types, including enteric neurons, glia, enteroendocrine cells, immune cells and bacteria, integrate peripheral GABA signals to influence brain functions and potentially contribute to brain disorders. We propose several priorities for this field, including the exploitation of available technologies to functionally dissect components of these GABA pathways within the BGM, with a focus on GI and brain-behaviour-disease. Furthermore, in silico ligand-receptor docking analyses using relevant bacterial metabolomic datasets, coupled with advances in knowledge of GABAAR 3D structures, could uncover new ligands with novel therapeutic potential. Finally, targeted design of dietary interventions is imperative to advancing their therapeutic potential to support GABA homeostasis across the BGM axis.
Collapse
Affiliation(s)
- Delia Belelli
- GABA Labs (Research) Ltd., Hemel Hempstead HP2 5HD, UK
- Division of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dundee DD1 5HL, UK
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Jeremy J Lambert
- Division of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dundee DD1 5HL, UK
| | - Murphy Lam Yim Wan
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Ana Rita Monteiro
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - David J Nutt
- GABA Labs (Research) Ltd., Hemel Hempstead HP2 5HD, UK
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Jerome D Swinny
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| |
Collapse
|
6
|
Moghaddam ZS, Dehghan A, Halimi S, Najafi F, Nokhostin A, Naeini AE, Akbarzadeh I, Ren Q. Bacterial Extracellular Vesicles: Bridging Pathogen Biology and Therapeutic Innovation. Acta Biomater 2025:S1742-7061(25)00352-6. [PMID: 40349898 DOI: 10.1016/j.actbio.2025.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The main role of bacterial extracellular vesicles (BEVs) has been associated with various processes such as intercellular communication and host-pathogen interactions. This comprehensive review explores the multifaceted functions of BEVs across different biological domains, emphasizing their dual nature as contributors to disease and potential vehicles for therapeutic intervention. We examine the intricate interactions of BEVs within bacterial communities and between bacteria and hosts, their involvement in disease development through cargo delivery mechanisms, and their beneficial impact on microbial ecology. The review also highlights BEVs' applications in biomedical field, where they are revolutionizing vaccine development, targeted drug delivery, and cancer therapy. By utilizing the inherent properties of BEVs for controlled drug release, targeted antigen delivery, and immune modulation, they offer a promising frontier in precision medicine. In addition, the diagnostic potential of BEVs is explored through their utility as biomarkers, providing valuable insights into disease states and treatment efficacy. Looking forward, this review underscores the challenges and opportunities in translating BEV research to clinical practice, promoting the need of standardized methods in BEV characterization and scaling up production. The diverse abilities of BEVs, ranging from contributing to pathogen virulence to driving therapeutic innovation, highlight their potential as a cornerstone in the future of biomedical advancements. STATEMENT OF SIGNIFICANCE: Bacterial extracellular vesicles (BEVs) are emerging as pivotal players in both pathogenesis and therapeutic innovation. This review explores their dual nature as agents of disease and as promising biomaterials for biomedical applications, and provides a comprehensive survey on their involvement in disease mechanisms and microbial ecology, and their potential in biomedical applications such as vaccine development, targeted drug delivery, cancer therapy, and diagnosis. It highlights the complex interactions of BEVs within bacterial communities and between bacteria and hosts. This review also addresses current advancements, challenges, and opportunities in translating BEV research into clinical practice. The insights presented here position BEVs as a cornerstone in the future of biomedical advancements, advocating for standardized methods in BEV characterization and scalable production techniques.
Collapse
Affiliation(s)
| | - Ashkan Dehghan
- W Booth School of Engineering Practice and Technology Faculty of Engineering, McMaster University Hamilton, ON, Canada, L8S 0A3
| | - Saba Halimi
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Fatemeh Najafi
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802-1503, United States
| | - Ali Nokhostin
- Medical Sciences & Technologies Faculty, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | | | - Iman Akbarzadeh
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| | - Qun Ren
- Laboratory for Biointerfaces, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland.
| |
Collapse
|
7
|
Yi W, Shi J, Zhou W, Wei J, Sun Y, Zeng X, Wang W. In vitro fermentation of a purified fraction of polysaccharides from the root of Brassica rapa L. by human gut microbiota and its interaction with Bacteroides ovatus. Food Chem 2025; 473:143109. [PMID: 39892338 DOI: 10.1016/j.foodchem.2025.143109] [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: 10/08/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
In this study, the physicochemical properties, in vitro fermentation, and interaction with Bacteroides ovatus of a purified fraction of polysaccharides from the root of Brassica rapa (BRP2-2) were investigated. It was demonstrated with an in vitro anaerobic fermentation model that BRP2-2 significantly increased (p < 0.05) the relative abundance of Bacteroides spp. After 24 h incubation of BRP2-2 with Bacteroides ovatus alone, 10.24 ± 0.69, 8.76 ± 0.48 and 3.37 ± 0.26 mM of acetate, propionate and isovalerate were produced, respectively. Moreover, transcriptome analysis of B. ovatus showed that 143 genes were up-regulated by BRP2-2, including five discrete polysaccharide utilization loci and two carbohydrate-active enzyme clusters. Based on the annotation of carbohydrate enzyme function, we speculate that BRP2-2 is a pectic polysaccharide containing homogalacturonan, rhamnogalacturonan I and rhamnogalacturonan II domains. These results suggested that BRP2-2 was degraded by B. ovatus, which produced metabolites with beneficial effects on host health.
Collapse
Affiliation(s)
- Wei Yi
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiameng Shi
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangting Zhou
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jinghong Wei
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Wei Wang
- College of Food Science and Pharmacy, Xinjiang Agricultural University, Urumqi 830052, China
| |
Collapse
|
8
|
Zheng R, Huang S, Feng P, Liu S, Jiang M, Li H, Zheng P, Mi Y, Li E. Comprehensive analysis of gut microbiota and fecal metabolites in patients with autism spectrum disorder. Front Microbiol 2025; 16:1557174. [PMID: 40351314 PMCID: PMC12062028 DOI: 10.3389/fmicb.2025.1557174] [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: 01/09/2025] [Accepted: 02/17/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and the presence of restricted, repetitive behaviors or interests. Studies have revealed that gut microbiota and their metabolism play important roles in ASD, and become the underlying mechanisms of ASD. Methods In this study, we performed long-read 16S rRNA sequencing and untargeted metabolomics to comprehensively characterize the profiles of gut microbiota and fecal metabolites in 34 ASD patients and 18 healthy controls. The associations between gut microbiota, fecal metabolites and clinical symptoms were analyzed to screen related biomarkers for ASD. Results The results showed the similarity of the overall microbial richness and diversity between ASD patients and controls, however, some specific bacterial taxa exhibited significant differences, including Klebsiella and Escherichia-Shigella at genera level, and Clostridium-sporogenes, Escherichia-coli-O157H7 and Bacteroides-ovatus at species level. The fecal metabolomics validated that a lot of metabolites had significantly differential levels, including a series of organic acids, amino acids and dopamine. Discussion The associations of gut microbiota and fecal metabolites might shed new light on the pathogenesis of ASD and help us to understand the importance of gut microbiota as potential biomarkers and therapeutic targets in the development of ASD.
Collapse
Affiliation(s)
- Ruijuan Zheng
- Department of Child Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Silu Huang
- Department of Child Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengya Feng
- Department of Child Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Simeng Liu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Miaomiao Jiang
- Department of Child Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huijuan Li
- Department of Child Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Enyao Li
- Department of Child Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Dadi P, Pauling CW, Shrivastava A, Shah DD. Synthesis of versatile neuromodulatory molecules by a gut microbial glutamate decarboxylase. iScience 2025; 28:112289. [PMID: 40264799 PMCID: PMC12013497 DOI: 10.1016/j.isci.2025.112289] [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: 10/23/2024] [Revised: 01/14/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Dysbiosis of the microbiome correlates with many neurological disorders, yet very little is known about the chemistry that controls the production of neuromodulatory molecules by gut microbes. Here, we found that an enzyme glutamate decarboxylase (BfGAD) of a gut microbe Bacteroides fragilis forms multiple neuromodulatory molecules such as γ-aminobutyric acid (GABA), hypotaurine, taurine, homotaurine, and β-alanine. We evolved BfGAD and doubled its taurine productivity. Additionally, we increased its specificity toward the substrate L-glutamate. Here, we provide a chemical strategy via which the BfGAD activity could be fine-tuned. In future, this strategy could be used to modulate the production of neuromodulatory molecules by gut microbes.
Collapse
Affiliation(s)
- Pavani Dadi
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Clint W. Pauling
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281, USA
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA
| | - Abhishek Shrivastava
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Dhara D. Shah
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281, USA
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA
| |
Collapse
|
10
|
Urbani G, Rondini E, Distrutti E, Marchianò S, Biagioli M, Fiorucci S. Phenotyping the Chemical Communications of the Intestinal Microbiota and the Host: Secondary Bile Acids as Postbiotics. Cells 2025; 14:595. [PMID: 40277921 PMCID: PMC12025480 DOI: 10.3390/cells14080595] [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: 03/04/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
The current definition of a postbiotic is a "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Postbiotics can be mainly classified as metabolites, derived from intestinal bacterial fermentation, or structural components, as intrinsic constituents of the microbial cell. Secondary bile acids deoxycholic acid (DCA) and lithocholic acid (LCA) are bacterial metabolites generated by the enzymatic modifications of primary bile acids by microbial enzymes. Secondary bile acids function as receptor ligands modulating the activity of a family of bile-acid-regulated receptors (BARRs), including GPBAR1, Vitamin D (VDR) receptor and RORγT expressed by various cell types within the entire human body. Secondary bile acids integrate the definition of postbiotics, exerting potential beneficial effects on human health given their ability to regulate multiple biological processes such as glucose metabolism, energy expenditure and inflammation/immunity. Although there is evidence that bile acids might be harmful to the intestine, most of this evidence does not account for intestinal dysbiosis. This review examines this novel conceptual framework of secondary bile acids as postbiotics and how these mediators participate in maintaining host health.
Collapse
Affiliation(s)
- Ginevra Urbani
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Elena Rondini
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy; (E.R.); (E.D.)
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy; (E.R.); (E.D.)
| | - Silvia Marchianò
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| |
Collapse
|
11
|
Cheng Y, Zhu Z, Yang Z, Liu X, Qian X, Zhu J, Hu X, Jiang P, Cui T, Wang Y, Ding W, Lei W, Gao J, Zhang J, Li Y, Shao L, Ling Z, Hu W. Alterations in fecal microbiota composition and cytokine expression profiles in adolescents with depression: a case-control study. Sci Rep 2025; 15:12177. [PMID: 40204825 PMCID: PMC11982373 DOI: 10.1038/s41598-025-97369-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025] Open
Abstract
Emerging evidence has highlighted that altered gut microbiota are associated with the onset and progression of depression via regulating the gut-brain axis. However, existing research has predominantly focused on children and adults, frequently neglecting adolescent depression. Given the rising prevalence and substantial impact of adolescent depression on functional impairment and suicidality, it is essential to focus more on this age group. In this study, we examined the fecal microbiota and inflammatory profiles of 99 depressed adolescents and 106 age-matched healthy controls using Illumina NovaSeq sequencing and multiplex immunoassays, respectively. Our findings revealed lower bacterial α-diversity and richness, alongside altered β-diversity in adolescents with depression. Gut dysbiosis associated with adolescent depression was characterized by increased pro-inflammatory genera such as Streptococcus and decreased anti-inflammatory genera like Faecalibacterium. These differential genera may serve as potential non-invasive biomarkers for adolescent depression, either individually or in combination. We also observed disruptions in the inferred microbiota functions in adolescent depression-associated microbiota, particularly in glycolysis and gluconeogenesis. Additionally, depressed adolescents exhibited systemic immune dysfunction, with elevated levels of pro-inflammatory cytokines and chemokines, which showed significant correlations with the differential genera. Our study bridges the gap between children and adults by providing new insights into the fecal microbiota characteristics and their links to immune system disruptions in depressed adolescents, which offer new targets for the diagnosis and treatment of depression in this age group.
Collapse
Affiliation(s)
- 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, 310003, Zhejiang, China
| | - Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhi Yang
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Xiulian Qian
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China
| | - Juntao Zhu
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China
| | - Xinzhu Hu
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China
| | - Peijie Jiang
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China
| | - Tingting Cui
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China
| | - Yuwei Wang
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China
| | - Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, Shandong, 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, 310003, Zhejiang, China
| | - Jingchen Zhang
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Yating Li
- 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, 310003, Zhejiang, China
| | - Li Shao
- School of Clinical Medicine, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310015, Zhejiang, China
| | - 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, 310003, Zhejiang, China.
| | - Weiming Hu
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, 324003, Zhejiang, China.
| |
Collapse
|
12
|
Malinauskiene V, Cijauskaite E, Sadauskaite G, Stundiene I. Role of Gut Microbiota and Metabolomics in Predicting Response to Vedolizumab in Inflammatory Bowel Disease: A Systematic Review. Pharmaceutics 2025; 17:476. [PMID: 40284471 PMCID: PMC12029995 DOI: 10.3390/pharmaceutics17040476] [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: 03/02/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Background: This review explores the impact of gut microbiota profiles in predicting the response to anti-integrin biologic therapy, particularly vedolizumab, in inflammatory bowel disease (IBD) patients. IBD, encompassing Crohn's disease and ulcerative colitis, is a chronic inflammatory condition with a growing prevalence linked to industrialization and lifestyle changes. Disruption in the gut microbiota balance, characterized by reduced diversity and altered short-chain fatty acid (SCFA) production, is associated with IBD and its symptoms. Current pharmacological treatments target healing and remission, with vedolizumab offering a gut-selective treatment approach. Methods: A search of the literature was performed on the relationship between anti-integrin treatment and the microbiome profile in IBD. Articles were examined from the PubMed, Medline, Cochrane, and Web of Science databases. Results: This review identified five human studies investigating the relationship between gut microbiome composition, SCFAs, and response to vedolizumab, revealing an increased abundance of beneficial bacteria and levels of SCFAs like butyrate in remission cases. Despite promising findings, the small sample sizes and limited scope of the existing studies highlight the need for larger, comprehensive research. Conclusions: This review underscores the potential of gut microbiome and metabolite profiling as non-invasive biomarkers for IBD severity and treatment outcomes, advocating for personalized therapeutic strategies to enhance efficacy. The insights gained could lead to novel diagnostic and treatment modalities, although further validation is necessary to fully understand the intricate connections between gut microbiota and IBD prognosis.
Collapse
Affiliation(s)
- Vaidota Malinauskiene
- Clinic of Gastroenterology, Nephrourology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (E.C.); (G.S.); (I.S.)
| | | | | | | |
Collapse
|
13
|
Wei X, Tang D. Effect of Bacteroides on Crohn's disease. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:393-402. [PMID: 39586813 DOI: 10.1055/a-2435-2659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Crohn's disease (CD), also known as cicatrizing enteritis, is an inflammatory bowel disease that occurs in the distal ileum and right colon of unknown cause and is also called inflammatory bowel disease (IBD) with ulcerative colitis (UC). In recent years, intestinal biota have been confirmed to play a significant role in various gastrointestinal diseases. Studies have found that intestinal microbiota disorders are closely associated with the onset and progression of Crohn's disease. Bacteroidetes, the second largest microbiota in the intestine, are crucial for equilibrium in the microbiota and intestinal environment. Certain Bacteroides can induce the development of Crohn's disease and aggravate intestinal inflammation directly or through their metabolites. Conversely, certain Bacteroides can reduce intestinal inflammation and symptoms of Crohn's disease. This article reviews the effect of several intestinal Bacteroides in the onset and progression of Crohn's disease and their impact on its treatment.
Collapse
Affiliation(s)
- Xuanyu Wei
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital, Nanjing University, Yangzhou, China
| |
Collapse
|
14
|
Liu W, Wang J, Yang H, Li C, Lan W, Chen T, Tang Y. The Metabolite Indole-3-Acetic Acid of Bacteroides Ovatus Improves Atherosclerosis by Restoring the Polarisation Balance of M1/M2 Macrophages and Inhibiting Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413010. [PMID: 39840614 PMCID: PMC11924036 DOI: 10.1002/advs.202413010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/13/2024] [Indexed: 01/23/2025]
Abstract
Emerging research has highlighted the significant role of the gut microbiota in atherosclerosis (AS), with microbiota-targeted interventions offering promising therapeutic potential. A central component of this process is gut-derived metabolites, which play a crucial role in mediating the distal functioning of the microbiota. In this study, a comprehensive microbiome-metabolite analysis using fecal and serum samples from patients with atherosclerotic cardiovascular disease and volunteers with risk factors for coronary heart disease and culture histology is performed, and identified the core strain Bacteroides ovatus (B. ovatus). Fecal microbiota transplantation experiments further demonstrated that the gut microbiota significantly influences AS progression, with B. ovatus alone exerting effects comparable to volunteer feces from volunteers. Notably, B. ovatus alleviated AS primarily by restoring the intestinal barrier and enhancing bile acid metabolism, particularly through the production of indole-3-acetic acid (IAA), a tryptophan-derived metabolite. IAA inhibited the TLR4/MyD88/NF-κB pathway in M1 macrophages, promoted M2 macrophage polarisation, and restored the M1/M2 polarisation balance, ultimately reducing aortic inflammation. These findings clarify the mechanistic interplay between the gut microbiota and AS, providing the first evidence that B. ovatus, a second-generation probiotic, can improve bile acid metabolism and reduce inflammation, offering a theoretical foundation for future AS therapeutic applications involving this strain.
Collapse
Affiliation(s)
- Wu Liu
- Department of Cardiovascular SurgeryThe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330008China
- The Second Clinical Medical College of Nanchang UniversityNanchang330008China
| | - Jingyu Wang
- The Second Clinical Medical College of Nanchang UniversityNanchang330008China
- Department of HematologyThe Second Affiliated Hospital of Nanchang UniversityNanchang330008China
| | - Heng Yang
- Department of Cardiovascular SurgeryThe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330008China
- The Second Clinical Medical College of Nanchang UniversityNanchang330008China
| | - Congcong Li
- Department of Cardiovascular SurgeryThe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330008China
- The Second Clinical Medical College of Nanchang UniversityNanchang330008China
| | - Wanqi Lan
- Department of Cardiovascular SurgeryThe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330008China
- The Second Clinical Medical College of Nanchang UniversityNanchang330008China
| | - Tingtao Chen
- The Institute of Translational MedicineJiangxi Medical CollegeNanchang UniversityNanchang330036China
- Jiangxi Province Key Laboratory of Bioengineering DrugsSchool of PharmacyJiangxi Medical CollegeNanchang UniversityNanchang330036China
| | - Yanhua Tang
- Department of Cardiovascular SurgeryThe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330008China
| |
Collapse
|
15
|
Liang M, Dong Q, Wu W, Fan J. Short-Chain Fatty Acids: Promising Therapeutic Targets for Respiratory Syncytial Virus Infection. Clin Rev Allergy Immunol 2025; 68:8. [PMID: 39873814 DOI: 10.1007/s12016-024-09018-x] [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] [Accepted: 11/02/2024] [Indexed: 01/30/2025]
Abstract
The intestinal microbiota is a complex community of organisms present in the human gastrointestinal tract, some of which can produce short-chain fatty acids (SCFAs) through the fermentation of dietary fiber. SCFAs play a major role in mediating the intestinal microbiota's regulation of host immunity and intestinal homeostasis. Respiratory syncytial virus (RSV) can cause an imbalance between anti-inflammatory and proinflammatory responses in the host. In addition, changes in SCFA levels and the structure of the intestinal microbiota have been observed after RSV infection. Therefore, there may be a link between SCFAs and RSV infection, and SCFAs are expected to be therapeutic targets for RSV infection.
Collapse
Affiliation(s)
- Mingxin Liang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Qinqin Dong
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Weiyi Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Juan Fan
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
16
|
Cao PP, Hu CL, Li MJ, An YH, Feng X, Ma XH, Wang DZ, Song ZH, Ji GS, Yang D, Ma Q, Yang WF, Dong JN, Zhang HR, Ma Y, Ma YF. 16S rRNA and metabolomics reveal the key microbes and key metabolites that regulate diarrhea in Holstein male calves. Front Microbiol 2025; 15:1521719. [PMID: 39881985 PMCID: PMC11778179 DOI: 10.3389/fmicb.2024.1521719] [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: 11/04/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Diarrhea is a prevalent disease among calves, which significantly hinders their growth and development, thereby impacting farm productivity and revenue. This study aimed to investigate the impact of diarrhea on calf growth. Methods Holstein male calves with similar birth weight (39.5 ± 4.2 kg) were included in this study, and key parameters such as fecal score, diarrhea incidence, and growth performance from birth to weaning were measured. Rectal fecal samples from both diarrheic (n = 24) and healthy calves (n = 24) aged 1-4 weeks were analyzed using 16S rRNA gene sequencing and untargeted metabolomics. Results Our findings indicated a high prevalence of diarrhea among calves between 1-4 weeks of age on pasture, which led to a marked decrease in growth performance, including average daily gain. At the genus level, the relative abundance of GCA-900066575 in one-week-old diarrheic calves was significantly higher; Escherichia-Shigella and Pseudoflavonifractor were more abundant in two-week-old calves; while Tyzzerella and Lachnospiraceae_UCG-004 increased significantly in four-week-old calves, and correlated negatively with average daily gain, suggesting that these bacteria may promote the occurrence of diarrhea. Correlation analysis revealed that fecal metabolites such as arachidonic acid, cis-vaccenic acid, oleic acid, choline, creatinine, and others were significantly negatively correlated with calf growth performance and were significantly increased in diarrheic calves. WGNCA identified that dark magenta module metabolites were significantly associated with diarrhea traits from 1-4 weeks. Thirteen metabolites, including glycerophospholipids (such as 1-stearoyl-2-hydroxy-sn-glycero-3-phosphoethanolamine), fatty acids (such as dodecanoic acid), and arachidonic acid, were positively correlated with GCA-900066575, Escherichia-shigella, Tyzzerella, and Clostridium_butyricum, but negatively correlated with UBA1819, Lachnoclostridium_sp_YL32, and Clostridium_scindens. Discussion Therefore, GCA-900066575, Escherichia-shigella, Lachnospiraceae_UCG-004, and Tyzzerella are likely key bacterial genera causing diarrhea in calves, while arachidonic acid, glycerol phospholipids, and fatty acids are critical metabolites associated with this condition. These alterations in the fecal microbiota and metabolite composition were found to be the principal contributors to growth retardation in diarrheic calves.
Collapse
Affiliation(s)
- P. P. Cao
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - C. L. Hu
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - M. J. Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Y. H. An
- Ningxia Borui Technology Co., Ltd, Yinchuan, China
| | - X. Feng
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - X. H. Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - D. Z. Wang
- Ningxia Borui Technology Co., Ltd, Yinchuan, China
| | - Z. H. Song
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - G. S. Ji
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - D. Yang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Q. Ma
- Ningxia Xin'ao Agriculture and Animal Husbandry Co., Ltd, Yinchuan, China
| | - W. F. Yang
- Ningxia Xin'ao Agriculture and Animal Husbandry Co., Ltd, Yinchuan, China
| | - J. N. Dong
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - H. R. Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Y. Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Y. F. Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
17
|
Schwartz L, Norman JO, Hasan S, Adamek OE, Dzuong E, Lowenstein JC, Yost OG, Sankaran B, McLaughlin KJ. Carbohydrate Deacetylase Unique to Gut Microbe Bacteroides Reveals Atypical Structure. Biochemistry 2025; 64:180-191. [PMID: 39663570 PMCID: PMC11713874 DOI: 10.1021/acs.biochem.4c00519] [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: 08/29/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
Bacteroides are often the most abundant, commensal species in the gut microbiome of industrialized human populations. One of the most commonly detected species is Bacteroides ovatus. It has been linked to benefits like the suppression of intestinal inflammation but is also correlated with some autoimmune disorders, for example irritable bowel disorder (IBD). Bacterial cell surface carbohydrates, like capsular polysaccharides (CPS), may play a role in modulating these varied host interactions. Recent studies have begun to explore the diversity of CPS loci in Bacteroides; however, there is still much unknown. Here, we present structural and functional characterization of a putative polysaccharide deacetylase from Bacteroides ovatus (BoPDA) encoded in a CPS biosynthetic locus. We solved four high resolution crystal structures (1.36-1.56 Å) of the enzyme bound to divalent cations Co2+, Ni2+, Cu2+, or Zn2+ and performed carbohydrate binding and deacetylase activity assays. Structural analysis of BoPDA revealed an atypical domain architecture that is unique to this enzyme, with a carbohydrate esterase 4 (CE4) superfamily catalytic domain inserted into a carbohydrate binding module (CBM). Additionally, BoPDA lacks the canonical CE4 His-His-Asp metal binding motif and our structures show it utilizes a noncanonical His-Asp dyad to bind metal ions. BoPDA is the first protein involved in CPS biosynthesis from B. ovatus to be characterized, furthering our understanding of significant biosynthetic processes in this medically relevant gut microbe.
Collapse
Affiliation(s)
- Lilith
A. Schwartz
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Jordan O. Norman
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Sharika Hasan
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Olive E. Adamek
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Elisa Dzuong
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Jasmine C. Lowenstein
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Olivia G. Yost
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Banumathi Sankaran
- Molecular
Biophysics and Integrated Bioimaging, Berkeley Center for Structural
Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Krystle J. McLaughlin
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| |
Collapse
|
18
|
Taherkhani S, Ahmadi P, Nasiraie LR, Janzadeh A, Honardoost M, Sedghi Esfahani S. Flavonoids and the gut microbiome: a powerful duo for brain health. Crit Rev Food Sci Nutr 2024:1-16. [PMID: 39632543 DOI: 10.1080/10408398.2024.2435593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Flavonoids, a class of polyphenolic compounds, are widely distributed in plant-based foods and have been recognized for their potential to promote overall health and well-being. Flavonoids in fruits and vegetables offer various beneficial effects such as anti-aging, anticancer, and anti-inflammatory properties. Flavonoids have been extensively studied for their neuroprotective properties, which are attributed to their ability to cross the blood-brain barrier and interact with neural cells. Factors like gut microbiota composition, age, genetics, and diet can impact how well flavonoids are absorbed in the gut. The gut microbiota can enhance the absorption of flavonoids through enzymatic processes, making microbiota composition a key factor influenced by age, genetics, and diet. Flavonoids can modulate the gut microbiota through prebiotic and antimicrobial effects, affecting the production of beneficial microbial metabolites like short-chain fatty acids (SCFAs) such as butyrate, which play a role in brain function and health. The gut microbiome also modulates the immune system, which is critical for preventing neuroinflammation. Additionally, flavonoids can benefit mental and psychological health by influencing anti-inflammatory signaling pathways in brain cells and increasing the absorption of tyrosine and tryptophan, precursors to neurotransmitters like serotonin, dopamine, norepinephrine, adrenaline, and gamma-aminobutyric acid (GABA). The flavonoid-gut microbiome axis is a complex and multifaceted relationship that has significant implications for neurological health. This review will explore how genetic and environmental factors can impact flavonoid absorption and the positive effects of flavonoids on brain health and the gut microbiota network.
Collapse
Affiliation(s)
- Soroush Taherkhani
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Ahmadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuromusculoskeletal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Roozbeh Nasiraie
- Department of Food Science and Technology, Islamic Azad University, Nour Branch, Nour, Iran
| | - Atousa Janzadeh
- Neuromusculoskeletal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Honardoost
- Cardio-Oncology Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sanaz Sedghi Esfahani
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
19
|
WANG Y, LIU J, XIONG Y, ZHANG Y, WEN Y, XUE M, GUO H, QIU J. Analysis of composition of gut microbial community in a rat model of functional dyspepsia treated with Simo Tang. J TRADIT CHIN MED 2024; 44:1168-1176. [PMID: 39617702 PMCID: PMC11589550 DOI: 10.19852/j.cnki.jtcm.20240927.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To investigate composition of gut microbial community in a rat model of functional dyspepsia (FD) and to explore the interventional effects of Simo Tang (, SMT). METHODS A rat model of FD was established through the tail-clamping stimulation method. The rat model of FD was assessed by the state of rats, their weight, sucrose preference rate, and intestinal propulsion rate. The DNA was extracted from stool samples after treatment with SMT. Amplified polymerase chain reaction (PCR) products of the 16S rDNA were sequenced using NovaseQ6000 after construction of libraries. Composition of gut microbial community in the stool samples was determined and analyzed by cluster analysis, bioinformatic analysis, and analysis of α-diversity and β-diversity. RESULTS The rat model of FD was successfully established using the tail-clamping stimulation method. The statistical results of cluster analysis of operational taxonomic units (OTUs) showed that the relative abundance of OTUs in the FD group was the lowest, while it was the highest in the normal (N) group. The composition of microbiome in the four groups was similar at phyla level. Compared with the FD group, the abundance of Firmicutes was downregulated, and the abundance of Proteobacteria and Bacteroidetes was upregulated in the Simo Tang (SMT) and high-dose Simo Tang (SMT.G) groups. The ratio of Bacteroidetes/ Firmicutes was also elevated. According to the analysis of α-diversity and β-diversity, the abundance of flora in FD rats was significantly reduced. The treatment using SMT appeared beneficial to improve the diversity of flora. SMT could improve the intestinal flora in FD rats. The results showed that FD rats had intestinal flora imbalance, and species diversity increased. The results suggested that SMT could regulate the disorders of intestinal flora caused by FD. CONCLUDIONS SMT could restore gut homeostasis and maintain gut flora diversity by modulating the gut microbiota and its associated metabolites in rats, thereby treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Yiying WANG
- 1 Centralab, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jianjun LIU
- 2 Clinical Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yongjian XIONG
- 1 Centralab, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yongli ZHANG
- 3 Department of Critical Care Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yuqi WEN
- 1 Centralab, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Mengli XUE
- 1 Centralab, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Huishu GUO
- 1 Centralab, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Juanjuan QIU
- 1 Centralab, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
20
|
Yang X, Hong C, Guan T, Zhang C, Xiao P, Yang Y, Xiao H, He Z. Investigation of the effects of Periplaneta americana (L.) extract on ischemic stroke based on combined multi-omics of gut microbiota. Front Pharmacol 2024; 15:1429960. [PMID: 39679371 PMCID: PMC11638836 DOI: 10.3389/fphar.2024.1429960] [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: 05/09/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Ischemic stroke (IS) is a highly lethal type of cardiovascular and cerebrovascular disease. Improving survival rates and promoting recovery in patients with IS pose significant challenges, however, recent research has identified the gut-brain axis as a therapeutic target. In this study, we evaluated the regulatory effect of Periplaneta americana (L.) extract (PAS840), which has established anti-inflammatory, antioxidant, and neuroprotective effects, on the gut microbiota using a rat model of temporary middle cerebral artery occlusion (tMCAO). We evaluated the protective effects of PAS840 on brain damage in IS rats through TTC (triphenyltetrazolium chloride), Nissl staining, and pathological section analysis. Additionally, we investigated the impact of PAS840 on the gut microbiota and metabolites using 16S rRNA sequencing, untargeted metabolomics of gut contents, and transcriptomics analyses of brain tissues to explore its mechanism of action. PAS840 intervention resulted in significant changes in the gut microbiota, including an increase in the abundance of probiotic flora, decrease in the abundance of harmful flora, and significant changes in metabolite profiles. It also attenuated brain damage, decreased platelet activity, inhibited oxidative stress and genes related to inflammation, and improved neurological function in rats. These findings suggest that PAS840 has preventive and therapeutic effects against IS via the gut-brain axis by regulating the gut microbiota and related metabolites. Accordingly, PAS840 is a candidate therapeutic drug for further research.
Collapse
Affiliation(s)
- Xin Yang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Canhui Hong
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Tangfei Guan
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Sichuan, Chengdu, China
| | - ChengGui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Peiyun Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Yongshou Yang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Huai Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Zhengchun He
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| |
Collapse
|
21
|
Šardzíková S, Gajewska M, Gałka N, Štefánek M, Baláž A, Garaiová M, Holič R, Świderek W, Šoltys K. Can longer lifespan be associated with gut microbiota involvement in lipid metabolism? FEMS Microbiol Ecol 2024; 100:fiae135. [PMID: 39354675 PMCID: PMC11503954 DOI: 10.1093/femsec/fiae135] [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: 12/01/2023] [Revised: 08/25/2024] [Accepted: 09/30/2024] [Indexed: 10/03/2024] Open
Abstract
Biological aging is linked to altered body composition and reduced neuroactive steroid hormones like dehydroepiandrosterone sulfate (DHEAS), which can stimulate the GABA signaling pathway via gut microbiota. Our study examined the association of gut microbiota with lifespan in mice through comprehensive analysis of its composition and functional involvement in cholesterol sulfate, a precursor of DHEAS, metabolism. We used 16S rRNA and metagenomic sequencing, followed by metabolic pathway prediction and thin layer chromatography and MALDI-TOF cholesterol sulfate identification. Significant increases in bacteria such as Bacteroides, typical for long-lived and Odoribacter and Colidextribacter, specific for short-lived mice were detected. Furthermore, for males (Rikenella and Alloprevotella) and females (Lactobacillus and Bacteroides), specific bacterial groups emerged as predictors (AUC = 1), highlighting sex-specific patterns. Long-lived mice showed a strong correlation of Bacteroides (0.918) with lipid and steroid hormone metabolism, while a negative correlation of GABAergic synapse with body weight (-0.589). We found that several Bacteroides species harboring the sulfotransferase gene and gene cluster for sulfonate donor synthesis are involved in converting cholesterol to cholesterol sulfate, significantly higher in the feces of long-lived individuals. Overall, we suggest that increased involvement of gut bacteria, mainly Bacteroides spp., in cholesterol sulfate synthesis could ameliorate aging through lipid metabolism.
Collapse
Affiliation(s)
- Sára Šardzíková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Marta Gajewska
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Norbert Gałka
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Matúš Štefánek
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Andrej Baláž
- Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia
| | - Martina Garaiová
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Roman Holič
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Wiesław Świderek
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Katarína Šoltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| |
Collapse
|
22
|
Qin B, Li Z, Zhu Q, Chen T, Lan W, Cui Y, Azad MAK, Kong X. Dietary Fermented Blueberry Pomace Supplementation Improves Small Intestinal Barrier Function and Modulates Cecal Microbiota in Aged Laying Hens. Animals (Basel) 2024; 14:2786. [PMID: 39409735 PMCID: PMC11475786 DOI: 10.3390/ani14192786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
This study aimed to investigate the effects of fermented blueberry pomace (FBP) on the intestinal barrier function and cecal microbiome of aged laying hens. A total of 320 Yukou Jingfen No. 8 laying hens (345-day-old) were randomly divided into a control group, 0.25% FBP group, 0.5% FBP group, or 1.0% FBP group. The results showed that the villus height (VH) in the jejunum of the 0.25-0.5% FBP groups and villus surface area in the jejunum of the 0.25% FBP group were higher (p < 0.05), while 0.25% FBP supplementation displayed a higher (p = 0.070) VH in the ileum compared to the control group. Mucin-2 expression was upregulated (p < 0.05) in the jejunum of the 0.5% FBP group and the ileum of the 0.25-0.5% FBP groups. Compared to the control group, interleukin (IL)-4 and IL-13 expressions were upregulated (p < 0.05) in the 1.0% FBP group. Microbiota analysis revealed that Prevotella abundance in the cecum of the 0.5-1.0% FBP groups was higher (p < 0.05) than in the 0.25% FBP group. In addition, microbial function prediction analysis showed that cecal microbiota in the 0.25% FBP group were mainly enriched by alanine/aspartate/glutamate metabolism and methane metabolism. Moreover, Spearman's correlation analysis revealed the potential correlations between the abundance of the cecal microbiota and intestinal-barrier-function-related gene expressions, as well as the short-chain fatty acid content, of laying hens. In summary, dietary FBP supplementation enhanced intestinal barrier function by improving intestinal morphology, upregulating gene expressions related to barrier function, and altering the cecal microbiota of aged laying hens.
Collapse
Affiliation(s)
- Binghua Qin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhihua Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
| | - Qian Zhu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Chen
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Lan
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
| | - Yadong Cui
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
| | - Md. Abul Kalam Azad
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiangfeng Kong
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Li X, Zhang Y, Zhang Q, Cao A, Feng J. Eucalyptus essential oil exerted a sedative-hypnotic effect by influencing brain neurotransmitters and gut microbes via the gut microbiota-brain axis. Front Pharmacol 2024; 15:1464654. [PMID: 39386024 PMCID: PMC11461282 DOI: 10.3389/fphar.2024.1464654] [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: 07/14/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Sleep disorders are becoming more and more common, leading to many health problems. However, most of current available medications to treat sleep disorders are addictive and even impair cognitive abilities. Therefore, it is important to find a natural and safe alternative to treat sleep disorders. In this study, twenty-four 8-week-old male ICR mice (25 ± 2 g) were equally divided into three groups: the control group (gavage of 0.9% saline), the eucalyptus essential oil (EEO) group (10 mg/kg B.W.), and the diazepam group (1 mg/kg B.W.). Firstly, open field test and sleep induction test were used to determine the sedative-hypnotic effect of EEO. Secondly, the effect of EEO on neurotransmitters in the mice brain was determined. Finally, based on the gut microbiota-brain axis (GMBA), the effect of EEO on the intestinal flora of mice was explored. It was found that EEO significantly reduce the activity and prolong the sleep duration of mice, exhibiting a good sedative-hypnotic effect. In the brain, EEO could increase the levels of sleep-promoting neurotransmitters, such as glutamine, Gamma-aminobutyric acid (GABA), glycine, tryptophan, N-acetylserotonin, and 5-hydroxyindoleacetic acid (5-HIAA). In the intestine, EEO was found to increase the diversity of gut microbes, the abundance of short chain fatty acid (SCFA) producing flora, and the abundance of functional flora synthesizing GABA and glycine neurotransmitters. These studies suggested that EEO exerted a sedative-hypnotic effect by acting on gut microbes and neurotransmitters in the brain. EEO has the potential to become a natural and safe alternative to traditional hypnotic sedative drugs.
Collapse
Affiliation(s)
- Xuejiao Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yuanyi Zhang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qian Zhang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Aizhi Cao
- Biotechnology R&D Center of Shandong Longchang Animal Health Products Co., Ltd., Jinan, China
| | - Jie Feng
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
24
|
Park B, Kim JY, Riffey OF, Walsh TJ, Johnson J, Donohoe DR. Crosstalk between butyrate oxidation in colonocyte and butyrate-producing bacteria. iScience 2024; 27:110853. [PMID: 39310762 PMCID: PMC11416512 DOI: 10.1016/j.isci.2024.110853] [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: 04/14/2024] [Revised: 08/01/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
The composition of gut microbiota, including butyrate-producing bacteria (BPB), is influenced by diet and physiological conditions. As such, given the importance of butyrate as an energetic substrate in colonocytes, it is unclear whether utilization of this substrate by the host would enhance BPB levels, thus defining a host-microbiome mutualistic relationship based on cellular metabolism. Here, it is shown through using a mouse model that lacks short-chain acyl dehydrogenase (SCAD), which is the first enzyme in the beta-oxidation pathway for short-chain fatty acids (SCFAs), that there is a significant diminishment in BPB at the phylum, class, species, and genus level compared to mice that have SCAD. Furthermore, SCAD-deficient mice do not show a prebiotic response from dietary fiber. Thus, oxidation of SCFAs by the host, which includes butyrate, is important in promoting BPB. These data help define the functional importance of diet-microbiome-host interactions toward microbiome composition, as it relates to function.
Collapse
Affiliation(s)
- Bohye Park
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA
| | - Ji Yeon Kim
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA
| | - Olivia F. Riffey
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Triston J. Walsh
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Jeremiah Johnson
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Dallas R. Donohoe
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
25
|
Hayase E, Hayase T, Mukherjee A, Stinson SC, Jamal MA, Ortega MR, Sanchez CA, Ahmed SS, Karmouch JL, Chang CC, Flores II, McDaniel LK, Brown AN, El-Himri RK, Chapa VA, Tan L, Tran BQ, Xiao Y, Fan C, Pham D, Halsey TM, Jin Y, Tsai WB, Prasad R, Glover IK, Enkhbayar A, Mohammed A, Schmiester M, King KY, Britton RA, Reddy P, Wong MC, Ajami NJ, Wargo JA, Shelburne S, Okhuysen PC, Liu C, Fowler SW, Conner ME, Katsamakis Z, Smith N, Burgos da Silva M, Ponce DM, Peled JU, van den Brink MRM, Peterson CB, Rondon G, Molldrem JJ, Champlin RE, Shpall EJ, Lorenzi PL, Mehta RS, Martens EC, Alousi AM, Jenq RR. Bacteroides ovatus alleviates dysbiotic microbiota-induced graft-versus-host disease. Cell Host Microbe 2024; 32:1621-1636.e6. [PMID: 39214085 PMCID: PMC11441101 DOI: 10.1016/j.chom.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Acute lower gastrointestinal GVHD (aLGI-GVHD) is a serious complication of allogeneic hematopoietic stem cell transplantation. Although the intestinal microbiota is associated with the incidence of aLGI-GVHD, how the intestinal microbiota impacts treatment responses in aLGI-GVHD has not been thoroughly studied. In a cohort of patients with aLGI-GVHD (n = 37), we found that non-response to standard therapy with corticosteroids was associated with prior treatment with carbapenem antibiotics and a disrupted fecal microbiome characterized by reduced abundances of Bacteroides ovatus. In a murine GVHD model aggravated by carbapenem antibiotics, introducing B. ovatus reduced GVHD severity and improved survival. These beneficial effects of Bacteroides ovatus were linked to its ability to metabolize dietary polysaccharides into monosaccharides, which suppressed the mucus-degrading capabilities of colonic mucus degraders such as Bacteroides thetaiotaomicron and Akkermansia muciniphila, thus reducing GVHD-related mortality. Collectively, these findings reveal the importance of microbiota in aLGI-GVHD and therapeutic potential of B. ovatus.
Collapse
Affiliation(s)
- Eiko Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Tomo Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Akash Mukherjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stuart C Stinson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mohamed A Jamal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Miriam R Ortega
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Christopher A Sanchez
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Saira S Ahmed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jennifer L Karmouch
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Chia-Chi Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Ivonne I Flores
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Lauren K McDaniel
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Alexandria N Brown
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Rawan K El-Himri
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Valerie A Chapa
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Bao Q Tran
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Yao Xiao
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christopher Fan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Dung Pham
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Taylor M Halsey
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Yimei Jin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Wen-Bin Tsai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Rishika Prasad
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Israel K Glover
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Altai Enkhbayar
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Aqsa Mohammed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Maren Schmiester
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Katherine Y King
- Center for Cell and Gene Therapy and Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pavan Reddy
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew C Wong
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Nadim J Ajami
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Samuel Shelburne
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pablo C Okhuysen
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Stephanie W Fowler
- Department of Molecular Virology and Microbiology and Department of Education, Innovation, and Technology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Comparative Medicine and Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Margaret E Conner
- Department of Molecular Virology and Microbiology and Department of Education, Innovation, and Technology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zoe Katsamakis
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Natalie Smith
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marina Burgos da Silva
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Doris M Ponce
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10021, USA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10021, USA
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey J Molldrem
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Hematopoietic Biology & Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Rohtesh S Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eric C Martens
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Amin M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert R Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; CPRIT Scholar in Cancer Research, Houston, TX, USA.
| |
Collapse
|
26
|
Sun C, Xiong X, Liu M, Liang Q, Zhao Q, Wei G, Shi J, Li X. Bacteroides ovatus alleviates high-fat and high-cholesterol -induced nonalcoholic fatty liver disease via gut-liver axis. Biomed Pharmacother 2024; 178:117156. [PMID: 39032286 DOI: 10.1016/j.biopha.2024.117156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/08/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024] Open
Abstract
Gut microbiota acts as a critical regulator in the development of nonalcoholic fatty liver disease (NAFLD), making probiotics a promise therapeutic strategy. Studies are needed to identify beneficial Bacteroides strains against NAFLD. Bacteroides ovatus (B. ovatus) may also exhibit therapy effect on NAFLD. The aim of this work was to evaluate the effect of B. ovatus on NAFLD and examine the mechanism. C57BL/6 J male mice were randomly divided into three groups: a control group (NCD) that received control standard diet, a model group (M) with high-fat and high-cholesterol (HFHC) diet, and M_Bo group that was fed HFFC supplemented with B. ovatus. Treatment with B. ovatus could reduce body weight, prevent hepatic steatohepatitis and liver injury. Mechanistically, B. ovatus induced changes of gut microbial diversity and composition, characterized by a decreased Firmicutes/Bacteroidetes (F/B) ratio in M_Bo group mice, a lower abundance of Proteobacteria, Verrucomicrobiota at phylum level and Ruminococcus_torques_group, Ruminococcus_gauvreauii_group, Erysipelatoclostridium at genus level, simultaneously a remarkablely higher fecal abundance of Lachnospiraceae_NK4A136_group, norank_f__Oscillospiraceae, Colidextribacter. Compared with M group, mice treated with B. ovatus showed an markedly altered fecal short chain fatty acids (SCFAs), a decline in serum levels of lipopolysaccharide (LPS), CD163, IL-1β, TNF-α, reduced macrophages in livers. Additionally, B. ovatus treatment caused downregulation of genes involved in denovo lipogenesis (such as Srebfl, Acaca, Scd1, Fasn), which was accompanied by the upregulation of genes related with fatty acid oxidation (such as Ppara). In conclusion, this study provides evidence that B. ovatus could ameliorate NAFLD by modulating the gut-liver axis.
Collapse
Affiliation(s)
- Changrui Sun
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Xuan Xiong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Maoyu Liu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Liang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Zhao
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guocui Wei
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianyou Shi
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Xiuxia Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
27
|
Crawford J, Liu S, Tao R, Kramer P, Bender S, Tao F. The ketogenic diet mitigates opioid-induced hyperalgesia by restoring short-chain fatty acids-producing bacteria in the gut. Pain 2024; 165:e106-e114. [PMID: 38452211 PMCID: PMC11333194 DOI: 10.1097/j.pain.0000000000003212] [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: 07/28/2023] [Accepted: 02/01/2024] [Indexed: 03/09/2024]
Abstract
ABSTRACT Opioids are commonly prescribed to patients with chronic pain. Chronic opioid usage comes with a slew of serious side effects, including opioid-induced hyperalgesia (OIH). The patients with long-term opioid treatment experience paradoxical increases in nociceptive hypersensitivity, namely, OIH. Currently, treatment options for OIH are extremely lacking. In this study, we show that the ketogenic diet recovers the abnormal pain behavior caused by chronic morphine treatment in male mice, and we further show that the therapeutic effect of the ketogenic diet is mediated through gut microbiome. Our 16S rRNA sequencing demonstrates that chronic morphine treatment causes changes in mouse gut microbiota, specifically a decrease in short-chain fatty acids-producing bacteria, and the sequencing data also show that the ketogenic diet rescues those bacteria in the mouse gut. More importantly, we show that supplementation with short-chain fatty acids (butyrate, propionate, and acetate) can delay the onset of OIH, indicating that short-chain fatty acids play a direct role in the development of OIH. Our findings suggest that gut microbiome could be targeted to treat OIH, and the ketogenic diet can be used as a complementary approach for pain relief in patients with chronic opioid treatment. We only used male mice in this study, and thus, our findings cannot be generalized to both sexes.
Collapse
Affiliation(s)
- Joshua Crawford
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Texas, USA
| | - Sufang Liu
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Texas, USA
| | - Ran Tao
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Texas, USA
| | - Phillip Kramer
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Texas, USA
| | - Steven Bender
- Department of Oral and Maxillofacial Surgery, Texas A&M University School of Dentistry, Dallas, Texas, USA
| | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Texas, USA
| |
Collapse
|
28
|
Lutsiv T, Hussan H, Thompson HJ. Ecosystemic Approach to Understanding Gut Microbiome-Mediated Prevention of Colorectal Cancer. Cancer J 2024; 30:329-344. [PMID: 39312453 DOI: 10.1097/ppo.0000000000000743] [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: 09/25/2024]
Abstract
Humans and their associated microorganisms coexist in complex symbiotic relationships. Continuously advancing research is demonstrating the crucial role of host-associated microbiota in the pathophysiology and etiology of disease and in mediating the prevention thereof. As an exemplar, the gut microbiota, especially colonic bacteria, have been extensively studied in colorectal cancer (CRC), and the growing body of evidence establishes new oncomicrobes and their oncometabolites associated with the initiation and promotion of carcinogenesis. Herein, we discuss the importance of approaching the gut microbiome as an ecosystem rather than an assortment of individual factors, especially in the context of cancer prevention. Furthermore, we argue that a dietary pattern effectively drives multiple nodes of the gut microbial ecosystem toward disease- or health-promoting qualities. In the modern circumstances of excessive consumption of ultraprocessed and animal-based foods and concomitant escalation of chronic disease burden worldwide, we focus on whole food-derived dietary fiber as a key to establishing a health-promoting eubiosis in the gut.
Collapse
|
29
|
Chong CW, Liew MS, Ooi W, Jamil H, Lim A, Hooi SL, Tay CSC, Tan G. Effect of green banana and pineapple fibre powder consumption on host gut microbiome. Front Nutr 2024; 11:1437645. [PMID: 39246394 PMCID: PMC11378528 DOI: 10.3389/fnut.2024.1437645] [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: 05/24/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Purpose To determine whether green banana powder (GBP) and pineapple fibre powder (PFP) promote beneficial bacterial species, directly improve human gut health and modulate the gut microbiome and understand their utility as functional foods and dietary supplements. Methods Over 14 days, 60 adults followed protocol requirements, completed food diaries and study questionnaires, avoided consuming supplements with prebiotics, probiotics or postbiotics, and ingested food containing 5 g of total daily fibre [placebo (10.75 g), GBP (10.75 g) or PFP (7.41 g)]. Participants' medical and baseline wellness histories, as well as stool samples, were collected at baseline, day 7 and 14. Stool DNA was processed for sequencing. Results Dietary fibre and resistant starches (RS) in GBP and PFP promoted temporal increases in beneficial bacteria. GBP significantly elevated 7 species (F. prausnitzii, B. longum, B. bifidum, B. adolescentis, B. pseudocatenulatum, B. obeum, and R. inulinivorans), while PFP enriched 6 species (B. ovatus, B. cellulosilyticus, B. bifidum, B. intestinalis, R. inulinivorans, and E. siraeum). These bacteria, found to be deficient in younger adults, were promoted by both powders. PFP benefitted both genders aged 16-23, while GBP benefitted overweight/obese individuals, including females. GBP and PFP fiber and RS improved bowel regularity and health as well as metabolism by promoting histidine, branched-chain amino acids, short-chain fatty acids, and biotin production. The additional fiber caused "low" bloatedness and reduced "fairly bad" sleep disruptions, without affecting sleep durations. Conclusion GBP and PFP supplementation increased beneficial bacteria and metabolites, improved host gut health, and present a valuable nutritional strategy for enhancing human health. Clinical trial registration AMILI Institutional Review Board, Identifier 2023/0301.
Collapse
Affiliation(s)
- Chun Wie Chong
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Mei Shan Liew
- Dole Specialty Ingredients, Dole Asia Holdings Pte., Ltd., Singapore, Singapore
| | - Weitze Ooi
- Dole Specialty Ingredients, Dole Asia Holdings Pte., Ltd., Singapore, Singapore
| | - Hassan Jamil
- Dole Specialty Ingredients, Dole Asia Holdings Pte., Ltd., Singapore, Singapore
| | | | | | | | | |
Collapse
|
30
|
Liu Z, Bai P, Wang L, Zhu L, Zhu Z, Jiang L. Clostridium tyrobutyricum in Combination with Chito-oligosaccharides Modulate Inflammation and Gut Microbiota for Inflammatory Bowel Disease Treatment. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18497-18506. [PMID: 39099138 DOI: 10.1021/acs.jafc.4c03486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Synbiotics, the combination of probiotics and prebiotics, are thought to be a pragmatic approach for the treatment of various diseases, including inflammatory bowel disease (IBD). The synergistic therapeutic effects of probiotics and prebiotics remain underexplored. Clostridium tyrobutyricum, a short-chain fatty acid (SCFA) producer, has been recognized as a promising probiotic candidate that can offer health benefits. In this study, the treatment effects of synbiotics containing C. tyrobutyricum and chitooligosaccharides (COSs) on IBD were evaluated. The results indicated that the synbiotic supplement effectively relieved inflammation and restored intestinal barrier function. Additionally, the synbiotic supplement could contribute to the elimination of reactive oxygen species (ROS) and improve the production of SCFAs through the SCFAs-producer of C. tyrobutyricum. Furthermore, such the synbiotic could also regulate the composition of gut microbiota. These findings underscore the potential of C. tyrobutyricum and COSs as valuable living biotherapeutics for the treatment of intestinal-related diseases.
Collapse
Affiliation(s)
- Zhenlei Liu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Pengfei Bai
- Nanjing Foreign Language School, Nanjing 210008, China
| | - Lefei Wang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Liying Zhu
- College of Chemical and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Zhengming Zhu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Ling Jiang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
31
|
Hernandez-Leyva AJ, Berna AZ, Liu Y, Rosen AL, Lint MA, Whiteside SA, Jaeger N, McDonough RT, Joardar N, Santiago-Borges J, Tomera CP, Luo W, John ARO, Kau AL. The breath volatilome is shaped by the gut microbiota. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.02.24311413. [PMID: 39132488 PMCID: PMC11312666 DOI: 10.1101/2024.08.02.24311413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The gut microbiota is widely implicated in host health and disease, inspiring translational efforts to implement our growing body of knowledge in clinical settings. However, the need to characterize gut microbiota by its genomic content limits the feasibility of rapid, point-of-care diagnostics. The microbiota produces a diverse array of xenobiotic metabolites that disseminate into tissues, including volatile organic compounds (VOCs) that may be excreted in breath. We hypothesize that breath contains gut microbe-derived VOCs that inform the composition and metabolic state of the microbiota. To explore this idea, we compared the breath volatilome and fecal gut microbiomes of 27 healthy children and found that breath VOC composition is correlated with gut microbiomes. To experimentally interrogate this finding, we devised a method for capturing exhaled breath from gnotobiotic mice. Breath volatiles are then profiled by gas-chromatography mass-spectrometry (GC-MS). Using this novel methodology, we found that the murine breath profile is markedly shaped by the composition of the gut microbiota. We also find that VOCs produced by gut microbes in pure culture can be identified in vivo in the breath of mice monocolonized with the same bacteria. Altogether, our studies identify microbe-derived VOCs excreted in breath and support a mechanism by which gut bacterial metabolism directly contributes to the mammalian breath VOC profiles.
Collapse
Affiliation(s)
- Ariel J. Hernandez-Leyva
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Amalia Z. Berna
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yang Liu
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anne L. Rosen
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael A. Lint
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Samantha A. Whiteside
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Natalia Jaeger
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Current address: Department of Immunology, Augusta University, Augusta, GA 30912, USA
| | - Ryan T. McDonough
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nikhilesh Joardar
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jesús Santiago-Borges
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Christopher P. Tomera
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Wentai Luo
- Department of Chemistry, Portland State University, Portland, OR 97201, USA
| | - Audrey R. Odom John
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew L. Kau
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
32
|
Zheng L, Jiao Y, Zhong H, Tan Y, Yin Y, Liu Y, Liu D, Wu M, Wang G, Huang J, Wang P, Qin M, Wang M, Xiao Y, Lv T, Luo Y, Hu H, Hou ST, Kui L. Human-derived fecal microbiota transplantation alleviates social deficits of the BTBR mouse model of autism through a potential mechanism involving vitamin B 6 metabolism. mSystems 2024; 9:e0025724. [PMID: 38780265 PMCID: PMC11237617 DOI: 10.1128/msystems.00257-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition characterized by social communication deficiencies and stereotypic behaviors influenced by hereditary and/or environmental risk factors. There are currently no approved medications for treating the core symptoms of ASD. Human fecal microbiota transplantation (FMT) has emerged as a potential intervention to improve autistic symptoms, but the underlying mechanisms are not fully understood. In this study, we evaluated the effects of human-derived FMT on behavioral and multi-omics profiles of the BTBR mice, an established model for ASD. FMT effectively alleviated the social deficits in the BTBR mice and normalized their distinct plasma metabolic profile, notably reducing the elevated long-chain acylcarnitines. Integrative analysis linked these phenotypic changes to specific Bacteroides species and vitamin B6 metabolism. Indeed, vitamin B6 supplementation improved the social behaviors in BTBR mice. Collectively, these findings shed new light on the interplay between FMT and vitamin B6 metabolism and revealed a potential mechanism underlying the therapeutic role of FMT in ASD.IMPORTANCEAccumulating evidence supports the beneficial effects of human fecal microbiota transplantation (FMT) on symptoms associated with autism spectrum disorder (ASD). However, the precise mechanism by which FMT induces a shift in the microbiota and leads to symptom improvement remains incompletely understood. This study integrated data from colon-content metagenomics, colon-content metabolomics, and plasma metabolomics to investigate the effects of FMT treatment on the BTBR mouse model for ASD. The analysis linked the amelioration of social deficits following FMT treatment to the restoration of mitochondrial function and the modulation of vitamin B6 metabolism. Bacterial species and compounds with beneficial roles in vitamin B6 metabolism and mitochondrial function may further contribute to improving FMT products and designing novel therapies for ASD treatment.
Collapse
Affiliation(s)
- Lifeng Zheng
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
- Xbiome Co. Ltd., Shenzhen, China
| | - Yinming Jiao
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Haolin Zhong
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Yan Tan
- Xbiome Co. Ltd., Shenzhen, China
| | | | | | - Ding Liu
- Xbiome Co. Ltd., Shenzhen, China
| | - Manli Wu
- Xbiome Co. Ltd., Shenzhen, China
| | - Guoyun Wang
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | | | - Ping Wang
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Meirong Qin
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children’s Hospital of Fudan University, National Center for Children’s Health, Shanghai, China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Tiying Lv
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yangzi Luo
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Han Hu
- Xbiome Co. Ltd., Shenzhen, China
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Ling Kui
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
33
|
Sastre DE, Sultana N, V A S Navarro M, Huliciak M, Du J, Cifuente JO, Flowers M, Liu X, Lollar P, Trastoy B, Guerin ME, Sundberg EJ. Human gut microbes express functionally distinct endoglycosidases to metabolize the same N-glycan substrate. Nat Commun 2024; 15:5123. [PMID: 38879612 PMCID: PMC11180146 DOI: 10.1038/s41467-024-48802-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Bacteroidales (syn. Bacteroidetes) are prominent members of the human gastrointestinal ecosystem mainly due to their efficient glycan-degrading machinery, organized into gene clusters known as polysaccharide utilization loci (PULs). A single PUL was reported for catabolism of high-mannose (HM) N-glycan glyco-polypeptides in the gut symbiont Bacteroides thetaiotaomicron, encoding a surface endo-β-N-acetylglucosaminidase (ENGase), BT3987. Here, we discover an ENGase from the GH18 family in B. thetaiotaomicron, BT1285, encoded in a distinct PUL with its own repertoire of proteins for catabolism of the same HM N-glycan substrate as that of BT3987. We employ X-ray crystallography, electron microscopy, mass spectrometry-based activity measurements, alanine scanning mutagenesis and a broad range of biophysical methods to comprehensively define the molecular mechanism by which BT1285 recognizes and hydrolyzes HM N-glycans, revealing that the stabilities and activities of BT1285 and BT3987 were optimal in markedly different conditions. BT1285 exhibits significantly higher affinity and faster hydrolysis of poorly accessible HM N-glycans than does BT3987. We also find that two HM-processing endoglycosidases from the human gut-resident Alistipes finegoldii display condition-specific functional properties. Altogether, our data suggest that human gut microbes employ evolutionary strategies to express distinct ENGases in order to optimally metabolize the same N-glycan substrate in the gastroinstestinal tract.
Collapse
Affiliation(s)
- Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
| | - Nazneen Sultana
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Structural Biochemistry Unit, National Institute of Dental and Craniofacial Research (NIDCR/NIH), Bethesda, MD, USA
| | - Marcos V A S Navarro
- Institute of Physics (IFSC-USP), University of São Paulo, São Carlos, SP, Brazil
- Center for Innovative Proteomics, Cornell University, Ithaca, NY, USA
| | - Maros Huliciak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Javier O Cifuente
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain
| | - Maria Flowers
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Xu Liu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Pete Lollar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Beatriz Trastoy
- Structural Glycoimmunology Laboratory, Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), Barcelona Science Park, c/Baldiri Reixac 4-8, Tower R, Barcelona, Catalonia, Spain
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
34
|
Dadi P, Pauling CW, Shrivastava A, Shah DD. Synthesis of versatile neuromodulatory molecules by a gut microbial glutamate decarboxylase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583032. [PMID: 38915512 PMCID: PMC11195143 DOI: 10.1101/2024.03.02.583032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Dysbiosis of the microbiome correlates with many neurological disorders, yet very little is known about the chemistry that controls the production of neuromodulatory molecules by gut microbes. Here, we found that an enzyme glutamate decarboxylase (BfGAD) of a gut microbe Bacteroides fragilis forms multiple neuromodulatory molecules such as γ-aminobutyric acid (GABA), hypotaurine, taurine, homotaurine, and β-alanine. We evolved BfGAD and doubled its taurine productivity. Additionally, we increased its specificity towards the substrate L-glutamate. Here, we provide a chemical strategy via which the BfGAD activity could be fine-tuned. In future, this strategy could be used to modulate the production of neuromodulatory molecules by gut microbes.
Collapse
Affiliation(s)
- Pavani Dadi
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281
- School of Life Sciences, Arizona State University, Tempe, AZ 85281
| | - Clint W. Pauling
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306
| | - Abhishek Shrivastava
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281
- School of Life Sciences, Arizona State University, Tempe, AZ 85281
| | - Dhara D. Shah
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306
| |
Collapse
|
35
|
Liu M, Peng R, Tian C, Shi J, Ma J, Shi R, Qi X, Zhao R, Guan H. Effects of the gut microbiota and its metabolite short-chain fatty acids on endometriosis. Front Cell Infect Microbiol 2024; 14:1373004. [PMID: 38938880 PMCID: PMC11208329 DOI: 10.3389/fcimb.2024.1373004] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
In recent years, a growing body of research has confirmed that the gut microbiota plays a major role in the maintenance of human health and disease. A gut microbiota imbalance can lead to the development of many diseases, such as pregnancy complications, adverse pregnancy outcomes, polycystic ovary syndrome, endometriosis, and cancer. Short-chain fatty acids are metabolites of specific intestinal bacteria and are crucial for maintaining intestinal homeostasis and regulating metabolism and immunity. Endometriosis is the result of cell proliferation, escape from immune surveillance, and invasive metastasis. There is a strong correlation between the anti-proliferative and anti-inflammatory effects of short-chain fatty acids produced by gut microbes and the development of endometriosis. Given that the mechanism of action of gut microbiota and Short-chain fatty acids in endometriosis remain unclear, this paper aims to provide a comprehensive review of the complex interactions between intestinal flora, short-chain fatty acids and endometriosis. In addition, we explored potential microbial-based treatment strategies for endometriosis, providing new insights into the future development of diagnostic tests and prevention and treatment methods for endometriosis.
Collapse
Affiliation(s)
- Menghe Liu
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Ru Peng
- Department of Obstetrics and Gynecology, Hohhot Maternal and Child Health Care Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Chunfang Tian
- Department of Oncology, Inner Mongolia Traditional Chinese Medicine Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jianping Shi
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jiannan Ma
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Ruiwen Shi
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiao Qi
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Rongwei Zhao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haibin Guan
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| |
Collapse
|
36
|
Fernandez-Cantos MV, Babu AF, Hanhineva K, Kuipers OP. Identification of metabolites produced by six gut commensal Bacteroidales strains using non-targeted LC-MS/MS metabolite profiling. Microbiol Res 2024; 283:127700. [PMID: 38518452 DOI: 10.1016/j.micres.2024.127700] [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: 12/15/2023] [Revised: 03/05/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
As the most abundant gram-negative bacterial order in the gastrointestinal tract, Bacteroidales bacteria have been extensively studied for their contribution to various aspects of gut health. These bacteria are renowned for their involvement in immunomodulation and their remarkable capacity to break down complex carbohydrates and fibers. However, the human gut microbiota is known to produce many metabolites that ultimately mediate important microbe-host and microbe-microbe interactions. To gain further insights into the metabolites produced by the gut commensal strains of this order, we examined the metabolite composition of their bacterial cell cultures in the stationary phase. Based on their abundance in the gastrointestinal tract and their relevance in health and disease, we selected a total of six bacterial strains from the relevant genera Bacteroides, Phocaeicola, Parabacteroides, and Segatella. We grew these strains in modified Gifu anaerobic medium (mGAM) supplemented with mucin, which resembles the gut microbiota's natural environment. Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based metabolite profiling revealed 179 annotated metabolites that had significantly differential abundances between the studied bacterial strains and the control growth medium. Most of them belonged to classes such as amino acids and derivatives, organic acids, and nucleot(s)ides. Of particular interest, Segatella copri DSM 18205 (previously referred to as Prevotella copri) produced substantial quantities of the bioactive metabolites phenylethylamine, tyramine, tryptamine, and ornithine. Parabacteroides merdae CL03T12C32 stood out due to its ability to produce cadaverine, histamine, acetylputrescine, and deoxycarnitine. In addition, we found that strains of the genera Bacteroides, Phocaeicola, and Parabacteroides accumulated considerable amounts of proline-hydroxyproline, a collagen-derived bioactive dipeptide. Collectively, these findings offer a more detailed comprehension of the metabolic potential of these Bacteroidales strains, contributing to a better understanding of their role within the human gut microbiome in health and disease.
Collapse
Affiliation(s)
- Maria Victoria Fernandez-Cantos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Ambrin Farizah Babu
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, Kuopio 70210, Finland
| | - Kati Hanhineva
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, Kuopio 70210, Finland; Department of Life Technologies, Food Sciences Unit, University of Turku, Turku 20014, Finland
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
37
|
Barcan AS, Barcan RA, Vamanu E. Therapeutic Potential of Fungal Polysaccharides in Gut Microbiota Regulation: Implications for Diabetes, Neurodegeneration, and Oncology. J Fungi (Basel) 2024; 10:394. [PMID: 38921380 PMCID: PMC11204944 DOI: 10.3390/jof10060394] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/04/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
This review evaluates the therapeutic effects of polysaccharides derived from mushroom species that have medicinal and edible properties. The fungal polysaccharides were recently studied, focusing on their modulation of the gut microbiota and their impact on various diseases. The study covers both clinical and preclinical studies, detailing the results and highlighting the significant influence of these polysaccharides on gut microbiota modulation. It discusses the potential health benefits derived from incorporating these polysaccharides into the diet for managing chronic diseases such as diabetes, neurodegenerative disorders, and cancer. Furthermore, the review emphasizes the interaction between fungal polysaccharides and the gut microbiota, underscoring their role in modulating the gut microbial community. It presents a systematic analysis of the findings, demonstrating the substantial impact of fungal polysaccharides on gut microbiota composition and function, which may contribute to their therapeutic effects in various chronic conditions. We conclude that the modulation of the gut microbiota by these polysaccharides may play a crucial role in mediating their therapeutic effects, offering a promising avenue for further research and potential applications in disease prevention and treatment.
Collapse
Affiliation(s)
- Alexandru Stefan Barcan
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Graham Kerr Building, Glasgow G12 8QQ, UK
| | | | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
| |
Collapse
|
38
|
Lillie IM, Booth CE, Horvath AE, Mondragon M, Engevik MA, Horvath TD. Characterizing arginine, ornithine, and putrescine pathways in enteric pathobionts. Microbiologyopen 2024; 13:e1408. [PMID: 38560776 PMCID: PMC10982811 DOI: 10.1002/mbo3.1408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 03/10/2024] [Accepted: 03/16/2024] [Indexed: 04/04/2024] Open
Abstract
Arginine-ornithine metabolism plays a crucial role in bacterial homeostasis, as evidenced by numerous studies. However, the utilization of arginine and the downstream products of its metabolism remain undefined in various gut bacteria. To bridge this knowledge gap, we employed genomic screening to pinpoint relevant metabolic targets. We also devised a targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomics method to measure the levels of arginine, its upstream precursors, and downstream products in cell-free conditioned media from enteric pathobionts, including Escherichia coli, Klebsiella aerogenes, K. pneumoniae, Pseudomonas fluorescens, Acinetobacter baumannii, Streptococcus agalactiae, Staphylococcus epidermidis, S. aureus, and Enterococcus faecalis. Our findings revealed that all selected bacterial strains consumed glutamine, glutamate, and arginine, and produced citrulline, ornithine, and GABA in our chemically defined medium. Additionally, E. coli, K. pneumoniae, K. aerogenes, and P. fluorescens were found to convert arginine to agmatine and produce putrescine. Interestingly, arginine supplementation promoted biofilm formation in K. pneumoniae, while ornithine supplementation enhanced biofilm formation in S. epidermidis. These findings offer a comprehensive insight into arginine-ornithine metabolism in enteric pathobionts.
Collapse
Affiliation(s)
- Ian M. Lillie
- Department of Materials Science & EngineeringCornell UniversityIthacaNew YorkUSA
- Department of Pathology & ImmunologyBaylor College of MedicineHoustonTexasUSA
- Department of PathologyTexas Children's HospitalHoustonTexasUSA
| | - Charles E. Booth
- Department of Regenerative Medicine & Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Adelaide E. Horvath
- Department of Regenerative Medicine & Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Department of Biology & BiochemistryUniversity of HoustonHoustonTexasUSA
- Department of MathematicsUniversity of HoustonHoustonTexasUSA
| | - Matthew Mondragon
- Department of Regenerative Medicine & Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Melinda A. Engevik
- Department of Regenerative Medicine & Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Department of Microbiology & ImmunologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Thomas D. Horvath
- Department of Pathology & ImmunologyBaylor College of MedicineHoustonTexasUSA
- Department of PathologyTexas Children's HospitalHoustonTexasUSA
| |
Collapse
|
39
|
Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, Foo JB, Ong YS, How CW, Khaw KY. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther 2024; 9:37. [PMID: 38360862 PMCID: PMC10869798 DOI: 10.1038/s41392-024-01743-1] [Citation(s) in RCA: 215] [Impact Index Per Article: 215.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/02/2024] [Accepted: 01/14/2024] [Indexed: 02/17/2024] Open
Abstract
The human gastrointestinal tract is populated with a diverse microbial community. The vast genetic and metabolic potential of the gut microbiome underpins its ubiquity in nearly every aspect of human biology, including health maintenance, development, aging, and disease. The advent of new sequencing technologies and culture-independent methods has allowed researchers to move beyond correlative studies toward mechanistic explorations to shed light on microbiome-host interactions. Evidence has unveiled the bidirectional communication between the gut microbiome and the central nervous system, referred to as the "microbiota-gut-brain axis". The microbiota-gut-brain axis represents an important regulator of glial functions, making it an actionable target to ameliorate the development and progression of neurodegenerative diseases. In this review, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases. As the gut microbiome provides essential cues to microglia, astrocytes, and oligodendrocytes, we examine the communications between gut microbiota and these glial cells during healthy states and neurodegenerative diseases. Subsequently, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases using a metabolite-centric approach, while also examining the role of gut microbiota-related neurotransmitters and gut hormones. Next, we examine the potential of targeting the intestinal barrier, blood-brain barrier, meninges, and peripheral immune system to counteract glial dysfunction in neurodegeneration. Finally, we conclude by assessing the pre-clinical and clinical evidence of probiotics, prebiotics, and fecal microbiota transplantation in neurodegenerative diseases. A thorough comprehension of the microbiota-gut-brain axis will foster the development of effective therapeutic interventions for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian Sheng Loh
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Wen Qi Mak
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Li Kar Stella Tan
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Chu Xin Ng
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Hong Hao Chan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Shiau Hueh Yeow
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| | - Kooi Yeong Khaw
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
40
|
Shin JH, Tillotson G, MacKenzie TN, Warren CA, Wexler HM, Goldstein EJC. Bacteroides and related species: The keystone taxa of the human gut microbiota. Anaerobe 2024; 85:102819. [PMID: 38215933 DOI: 10.1016/j.anaerobe.2024.102819] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Microbial communities play a significant role in maintaining ecosystems in a healthy homeostasis. Presently, in the human gastrointestinal tract, there are certain taxonomic groups of importance, though there is no single species that plays a keystone role. Bacteroides spp. are known to be major players in the maintenance of eubiosis in the human gastrointestinal tract. Here we review the critical role that Bacteroides play in the human gut, their potential pathogenic role outside of the gut, and their various methods of adapting to the environment, with a focus on data for B. fragilis and B. thetaiotaomicron. Bacteroides are anaerobic non-sporing Gram negative organisms that are also resistant to bile acids, generally thriving in the gut and having a beneficial relationship with the host. While they are generally commensal organisms, some Bacteroides spp. can be opportunistic pathogens in scenarios of GI disease, trauma, cancer, or GI surgery, and cause infection, most commonly intra-abdominal infection. B. fragilis can develop antimicrobial resistance through multiple mechanisms in large part due to its plasticity and fluid genome. Bacteroidota (formerly, Bacteroidetes) have a very broad metabolic potential in the GI microbiota and can rapidly adapt their carbohydrate metabolism to the available nutrients. Gastrointestinal Bacteroidota species produce short-chain fatty acids such as succinate, acetate, butyrate, and occasionally propionate, as the major end-products, which have wide-ranging and many beneficial influences on the host. Bacteroidota, via bile acid metabolism, also play a role in in colonization-resistance of other organisms, including Clostridioides difficile, and maintenance of gut integrity.
Collapse
Affiliation(s)
- Jae Hyun Shin
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| | | | | | - Cirle A Warren
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| | - Hannah M Wexler
- GLAVAHCS, Los Angeles, CA, USA; David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | | |
Collapse
|
41
|
Mullowney MW, Fiebig A, Schnizlein MK, McMillin M, Rose AR, Koval J, Rubin D, Dalal S, Sogin ML, Chang EB, Sidebottom AM, Crosson S. Microbially catalyzed conjugation of GABA and tyramine to bile acids. J Bacteriol 2024; 206:e0042623. [PMID: 38174933 PMCID: PMC10810215 DOI: 10.1128/jb.00426-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Bile acids (BAs) are cholesterol-derived molecules that aid in digestion and nutrient absorption, regulate host metabolic processes, and influence physiology of the gut microbiota. Both the host and its microbiome contribute to enzymatic modifications that shape the chemical diversity of BAs in the gut. Several bacterial species have been reported to conjugate standard amino acids to BAs, but it was not known if bacteria conjugate BAs to other amine classes. Here, we show that Bacteroides fragilis strain P207, isolated from a bacterial bloom in the J-pouch of a patient with ulcerative colitis pouchitis, conjugates standard amino acids and the neuroactive amines γ-aminobutyric acid (GABA) and tyramine to deoxycholic acid. We extended this analysis to other human gut isolates and identified species that are competent to conjugate GABA and tyramine to primary and secondary BAs, and further identified diverse BA-GABA and BA-tyramine amides in human stool. A longitudinal metabolomic analysis of J-pouch contents of the patient from whom B. fragilis P207 was isolated revealed highly reduced levels of secondary bile acids and a shifting BA amide profile before, during, and after onset of pouchitis, including temporal changes in several BA-GABA amides. Treatment of pouchitis with ciprofloxacin was associated with a marked reduction of nearly all BA amides in the J-pouch. Our study expands the known repertoire of conjugated bile acids produced by bacteria to include BA conjugates to GABA and tyramine and demonstrates that these molecules are present in the human gut. IMPORTANCE BAs are modified in multiple ways by host enzymes and the microbiota to produce a chemically diverse set of molecules that assist in the digestive process and impact many physiological functions. This study reports the discovery of bacterial species that conjugate the neuroactive amines, GABA and tyramine, to primary and secondary BAs. We further present evidence that BA-GABA and BA-tyramine conjugates are present in the human gut, and document a shifting BA-GABA profile in a human pouchitis patient before, during, and after inflammation and antibiotic treatment. GABA and tyramine are common metabolic products of the gut microbiota and potent neuroactive molecules. GABA- and tyramine-conjugated BAs may influence receptor-mediated regulatory mechanisms of humans and their gut microbes, and absorption of these molecules and their entry into enterohepatic circulation may impact host physiology at distal tissue sites. This study defines new conjugated bile acids in the human gut.
Collapse
Affiliation(s)
| | - Aretha Fiebig
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Matthew K. Schnizlein
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Mary McMillin
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Amber R. Rose
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Jason Koval
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - David Rubin
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Sushila Dalal
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | - Eugene B. Chang
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | - Sean Crosson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
42
|
Meng X, Shu Q. Novel primers to identify a wider diversity of butyrate-producing bacteria. World J Microbiol Biotechnol 2024; 40:76. [PMID: 38252387 DOI: 10.1007/s11274-023-03872-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024]
Abstract
Butyrate-producing bacteria are a functionally important part of the intestinal tract flora, and the resulting butyric acid is essential for maintaining host intestinal health, regulating the immune system, and influencing energy metabolism. However, butyrate-producing bacteria have not been defined as a coherent phylogenetic group. They are primarily identified using primers for key genes in the butyrate-producing pathway, and their use has been limited to the Bacillota and Bacteroidetes phyla. To overcome this limitation, we developed functional gene primers able to identify butyrate-producing bacteria through the butyrate kinase gene, which encodes the enzyme involved in the final step of the butyrate-producing pathway. Genomes extracted from human and rat feces were used to amplify the target genes through PCR. The obtained sequences were analyzed using BLASTX to construct a developmental tree using the MEGA software. The newly designed butyrate kinase gene primers allowed to recognize a wider diversity of butyrate-producing bacteria than that recognized using currently available primers. Specifically, butyrate-producing bacteria from the Synergistota and Spirochaetota phyla were identified for the first time using these primers. Thus, the developed primers provide a more accurate method for researchers and doctors to identify potential butyrate-producing bacteria and deepen our understanding of butyrate-producing bacterial species.
Collapse
Affiliation(s)
- Xianbin Meng
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Qinglong Shu
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| |
Collapse
|
43
|
Dop C, Auvin S, Mondot S, Lepage P, Ilhan ZE. Longitudinal exposure to antiseizure medications shape gut-derived microbiome, resistome, and metabolome landscape. ISME COMMUNICATIONS 2024; 4:ycae123. [PMID: 39526134 PMCID: PMC11544314 DOI: 10.1093/ismeco/ycae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
The influence of chronically administered host-targeted drugs on the gut microbiome remains less understood compared to antibiotics. We investigated repetitive exposure effects of three common antiseizure medications [carbamazepine (CBZ), valproic acid, and levetiracetam] on the gut microbial composition, resistome, and metabolome using microcosms constructed from feces of young children. Microcosms were established by cultivating feces for 24 h (C0). These microcosms were daily transferred into fresh media for seven cycles (C1-C7) with antiseizure medications or carrier molecules, followed by four cycles without any drugs (C8-C11). The microbial dynamics and resistome of microcosms at C0, C1, C7, and C11 were assessed with 16S ribosomal ribonucleic acid gene sequencing or shotgun metagenome sequencing and real-time quantitative polymerase chain reaction analysis of the antimicrobial resistance genes, respectively. Metabolites of CBZ-treated and control microcosms at C0, C1, and C7 were evaluated using non-targeted metabolomics. Our findings revealed that the serial transfer approach longitudinally altered the microcosm composition. Among the medications, CBZ had the most substantial impact on the structure and metabolism of the feces-derived microcosms. The microbiome composition partially recovered during the drug-free period. Specifically, Bacteroides and Flavonifractor were depleted and Escherichia and Clostridium were enriched. Additionally, repetitive CBZ exposure increased the abundance and expression of genes related to various antibiotic resistance mechanisms, more specifically, efflux pumps and antibiotic target alteration. CBZ-induced changes in the microbiome were mirrored in the metabolome, with reductions in the citric acid cycle metabolites, glutamine, and spermidine, alongside increased levels of vitamin B6. Our study suggests that repetitive CBZ exposure may negatively impact gut microbial homeostasis and metabolism.
Collapse
Affiliation(s)
- Camille Dop
- Université Paris-Saclay, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Micalis Institute, Domaine de Vilvert, Jouy-en-Josas, France
| | - Stéphane Auvin
- Université de Paris, Institut National de la Santé et de la Recherche Médicale (INSERM), NeuroDiderot, Paris, France
- Pediatric Neurology Department, Assistance Publique-Hôpitaux de Paris (APHP), Robert Debré University Hospital, Paris, France
- Institut Universitaire de France, Paris, France
| | - Stanislas Mondot
- Université Paris-Saclay, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Micalis Institute, Domaine de Vilvert, Jouy-en-Josas, France
| | - Patricia Lepage
- Université Paris-Saclay, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Micalis Institute, Domaine de Vilvert, Jouy-en-Josas, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), AgroParisTech, Micalis Institute, Domaine de Vilvert, Jouy-en-Josas, France
| |
Collapse
|
44
|
Roussel C, Sola M, Lessard-Lord J, Nallabelli N, Généreux P, Cavestri C, Azeggouar Wallen O, Villano R, Raymond F, Flamand N, Silvestri C, Di Marzo V. Human gut microbiota and their production of endocannabinoid-like mediators are directly affected by a dietary oil. Gut Microbes 2024; 16:2335879. [PMID: 38695302 PMCID: PMC11067990 DOI: 10.1080/19490976.2024.2335879] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) and the gut microbiome affect each other. We investigated the impact of supplementation with Buglossoides arvensis oil (BO), rich in stearidonic acid (SDA), on the human gut microbiome. Employing the Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), we simulated the ileal and ascending colon microbiomes of four donors. Our results reveal two distinct microbiota clusters influenced by BO, exhibiting shared and contrasting shifts. Notably, Bacteroides and Clostridia abundance underwent similar changes in both clusters, accompanied by increased propionate production in the colon. However, in the ileum, cluster 2 displayed a higher metabolic activity in terms of BO-induced propionate levels. Accordingly, a triad of bacterial members involved in propionate production through the succinate pathway, namely Bacteroides, Parabacteroides, and Phascolarctobacterium, was identified particularly in this cluster, which also showed a surge of second-generation probiotics, such as Akkermansia, in the colon. Finally, we describe for the first time the capability of gut bacteria to produce N-acyl-ethanolamines, and particularly the SDA-derived N-stearidonoyl-ethanolamine, following BO supplementation, which also stimulated the production of another bioactive endocannabinoid-like molecule, commendamide, in both cases with variations across individuals. Spearman correlations enabled the identification of bacterial genera potentially involved in endocannabinoid-like molecule production, such as, in agreement with previous reports, Bacteroides in the case of commendamide. This study suggests that the potential health benefits on the human microbiome of certain dietary oils may be amenable to stratified nutrition strategies and extend beyond n-3 PUFAs to include microbiota-derived endocannabinoid-like mediators.
Collapse
Affiliation(s)
- Charlène Roussel
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Mathilde Sola
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Jacob Lessard-Lord
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
| | - Nayudu Nallabelli
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Pamela Généreux
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
| | - Camille Cavestri
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
| | - Oumaima Azeggouar Wallen
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Rosaria Villano
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli (Napoli), Italy
| | - Frédéric Raymond
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Nicolas Flamand
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Cristoforo Silvestri
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Vincenzo Di Marzo
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| |
Collapse
|
45
|
Moradian H, Gabriel T, Barrau M, Roblin X, Paul S. New methods to unveil host-microbe interaction mechanisms along the microbiota-gut-brain-axis. Gut Microbes 2024; 16:2351520. [PMID: 38717832 PMCID: PMC11086032 DOI: 10.1080/19490976.2024.2351520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Links between the gut microbiota and human health have been supported throughout numerous studies, such as the development of neurological disease disorders. This link is referred to as the "microbiota-gut-brain axis" and is the focus of an emerging field of research. Microbial-derived metabolites and gut and neuro-immunological metabolites regulate this axis in health and many diseases. Indeed, assessing these signals, whether induced by microbial metabolites or neuro-immune mediators, could significantly increase our knowledge of the microbiota-gut-brain axis. However, this will require the development of appropriate techniques and potential models. Methods for studying the induced signals originating from the microbiota remain crucial in this field. This review discusses the methods and techniques available for studies of microbiota-gut-brain interactions. We highlight several much-debated elements of these methodologies, including the widely used in vivo and in vitro models, their implications, and perspectives in the field based on a systematic review of PubMed. Applications of various animal models (zebrafish, mouse, canine, rat, rabbit) to microbiota-gut-brain axis research with practical examples of in vitro methods and innovative approaches to studying gut-brain communications are highlighted. In particular, we extensively discuss the potential of "organ-on-a-chip" devices and their applications in this field. Overall, this review sheds light on the most widely used models and methods, guiding researchers in the rational choice of strategies for studies of microbiota-gut-brain interactions.
Collapse
Affiliation(s)
- Habibullah Moradian
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
| | - Tristan Gabriel
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
| | - Mathilde Barrau
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Xavier Roblin
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
46
|
Lin H, Peddada SD. Multigroup analysis of compositions of microbiomes with covariate adjustments and repeated measures. Nat Methods 2024; 21:83-91. [PMID: 38158428 PMCID: PMC10776411 DOI: 10.1038/s41592-023-02092-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/17/2023] [Indexed: 01/03/2024]
Abstract
Microbiome differential abundance analysis methods for two groups are well-established in the literature. However, many microbiome studies involve more than two groups, sometimes even ordered groups such as stages of a disease, and require different types of comparison. Standard pairwise comparisons are inefficient in terms of power and false discovery rates. In this Article, we propose a general framework, ANCOM-BC2, for performing a wide range of multigroup analyses with covariate adjustments and repeated measures. We illustrate our methodology through two real datasets. The first example explores the effects of aridity on the soil microbiome, and the second example investigates the effects of surgical interventions on the microbiome of patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Huang Lin
- Biostatistics and Computational Biology Branch, NIEHS, NIH, Research Triangle Park, NC, USA
- Department of Epidemiology and Biostatistics, University of Maryland, College Park, MD, USA
| | - Shyamal Das Peddada
- Biostatistics and Computational Biology Branch, NIEHS, NIH, Research Triangle Park, NC, USA.
| |
Collapse
|
47
|
Bonham KS, Fahur Bottino G, McCann SH, Beauchemin J, Weisse E, Barry F, Cano Lorente R, The RESONANCE Consortium, Huttenhower C, Bruchhage M, D’Sa V, Deoni S, Klepac-Ceraj V. Gut-resident microorganisms and their genes are associated with cognition and neuroanatomy in children. SCIENCE ADVANCES 2023; 9:eadi0497. [PMID: 38134274 PMCID: PMC10745691 DOI: 10.1126/sciadv.adi0497] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023]
Abstract
Emerging evidence implicates gut microbial metabolism in neurodevelopmental disorders, but its influence on typical neurodevelopment has not been explored in detail. We investigated the relationship between the microbiome and neuroanatomy and cognition of 381 healthy children, demonstrating that differences in microbial taxa and genes are associated with overall cognitive function and the size of brain regions. Using a combination of statistical and machine learning models, we showed that species including Alistipes obesi, Blautia wexlerae, and Ruminococcus gnavus were enriched or depleted in children with higher cognitive function scores. Microbial metabolism of short-chain fatty acids was also associated with cognitive function. In addition, machine models were able to predict the volume of brain regions from microbial profiles, and taxa that were important in predicting cognitive function were also important for predicting individual brain regions and specific subscales of cognitive function. These findings provide potential biomarkers of neurocognitive development and may enable development of targets for early detection and intervention.
Collapse
Affiliation(s)
- Kevin S. Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | | | | | | | - Elizabeth Weisse
- Department of Psychology, University of Stavanger, Stavanger, Norway
| | | | | | | | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Associate Member, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Muriel Bruchhage
- Department of Psychology, University of Stavanger, Stavanger, Norway
| | - Viren D’Sa
- Rhode Island Hospital, Providence, RI, USA
| | - Sean Deoni
- Rhode Island Hospital, Providence, RI, USA
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| |
Collapse
|
48
|
Gao X, Yang W, Li S, Liu S, Yang W, Song S, Sheng J, Zhao Y, Tian Y. Moringa oleifera leaf alleviates functional constipation via regulating the gut microbiota and the enteric nervous system in mice. Front Microbiol 2023; 14:1315402. [PMID: 38192287 PMCID: PMC10773919 DOI: 10.3389/fmicb.2023.1315402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Moringa oleifera Lam. leaf is not only a new food resource in China, but also a traditional medicinal plant. It is commonly used in the folk to alleviate constipation, but its laxative mechanism is not fully understood. Hence we investigated it in loperamide-induced functional constipation (FC) mice. The results showed that MOAE significantly regulated not only gastrointestinal hormones and neurotransmitters in serum but also important gastrointestinal motility factors in the enteric nervous system (ENS)-interstitial cells of Cajal (ICCs)-smooth muscle cell (SMC) network. Meanwhile, MOAE attenuated intestinal inflammation, increased cecal short-chain fatty acid levels and colonic antimicrobial peptide expression, and improved the impaired intestinal barrier function in loperamide-induced FC mice. In addition, MOAE also increased fecal water content by inhibiting the mRNA expression of colonic aquaporins (Aqp3 and Aqp4) in FC mice. Interestingly and importantly, MOAE affected the intestinal microbiota by inhibiting some key "constipation-causing" microbiota, such as Bacteroidaceae, Clostridiaceae, Bacteroides, and Ruminococcus, and promoting the growth of other important "constipation-curing" microbiota, such as Butyricoccus, Tyzzerella, and Desulfovibrio. These important taxa are significantly associated with a variety of indicators of constipation. These findings suggest that MOAE can promote defecation through its rich chemical composition to modulate the ENS-ICCs-SMCs network and the gut microecosystem.
Collapse
Affiliation(s)
- Xiaoyu Gao
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming, China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Weiqian Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Department of Hotel Management, Chongqing Vocational Institute of Tourism, Chongqing, China
| | - Sijin Li
- College of Pu’er Tea West Yunnan University of Applied Sciences, Puer, China
| | - Shuangfeng Liu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Weixing Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Shuang Song
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jun Sheng
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Yan Zhao
- Division of Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yang Tian
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
49
|
Mullowney MW, Fiebig A, Schnizlein MK, McMillin M, Rose AR, Koval J, Rubin D, Dalal S, Sogin ML, Chang EB, Sidebottom AM, Crosson S. Microbially-catalyzed conjugation of GABA and tyramine to bile acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559407. [PMID: 37808758 PMCID: PMC10557584 DOI: 10.1101/2023.09.25.559407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Bile acids (BAs) are cholesterol-derived molecules that aid in digestion and nutrient absorption, regulate host metabolic processes, and influence physiology of the gut microbiota. Both the host and its microbiome contribute to enzymatic modifications that shape the chemical diversity of BAs in the gut. Several bacterial species have been reported to conjugate standard amino acids to BAs, but it was not known if bacteria conjugate BAs to other amine classes. Here, we show that Bacteroides fragilis strain P207, isolated from a bacterial bloom in the J-pouch of a patient with ulcerative colitis (UC) pouchitis, conjugates standard amino acids and the neuroactive amines γ-aminobutyric acid (GABA) and tyramine to deoxycholic acid. We extended this analysis to other human gut isolates and identified species that are competent to conjugate GABA and tyramine to primary and secondary BAs, and further identified diverse BA-GABA and BA-tyramine amides in human stool. A longitudinal metabolomic analysis of J-pouch contents of the patient from whom B. fragilis P207 was isolated revealed highly reduced levels of secondary bile acids and a shifting BA amide profile before, during, and after onset of pouchitis, including temporal changes in several BA-GABA amides. Treatment of pouchitis with ciprofloxacin was associated with a marked reduction of nearly all BA amides in the J-pouch. Our study expands the known repertoire of conjugated bile acids produced by bacteria to include BA conjugates to GABA and tyramine and demonstrates that these molecules are present in the human gut.
Collapse
Affiliation(s)
- Michael W Mullowney
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Aretha Fiebig
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Matthew K Schnizlein
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Mary McMillin
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Amber R Rose
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Jason Koval
- Department of Medicine, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - David Rubin
- Department of Medicine, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Sushila Dalal
- Department of Medicine, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | | | - Eugene B Chang
- Department of Medicine, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Ashley M Sidebottom
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Sean Crosson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
50
|
Zhang C, Yu L, Ma C, Jiang S, Zhang Y, Wang S, Tian F, Xue Y, Zhao J, Zhang H, Liu L, Chen W, Huang S, Zhang J, Zhai Q. A key genetic factor governing arabinan utilization in the gut microbiome alleviates constipation. Cell Host Microbe 2023; 31:1989-2006.e8. [PMID: 37992712 DOI: 10.1016/j.chom.2023.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 11/24/2023]
Abstract
Impaired gastrointestinal motility is associated with gut dysbiosis. Probiotics, such as Bifidobacteria, can improve this bowel disorder; however, efficacy is strain-dependent. We determine that a genetic factor, the abfA cluster governing arabinan utilization, in Bifidobacterium longum impacts treatment efficacy against functional constipation (FC). In mice with FC, B. longum, but not an abfA mutant, improved gastrointestinal transit time, an affect that was dependent upon dietary arabinan. abfA genes were identified in other commensal bacteria, whose effects in ameliorating murine FC were similarly abfA-dependent. In a double-blind, randomized, placebo-controlled clinical trial, supplementation with abfA-cluster-carrying B. longum, but not an abfA-deficient strain, enriched arabinan-utilization residents, increased beneficial metabolites, and improved FC symptoms. Across human cohorts, abfA-cluster abundance can predict FC, and transplantation of abfA cluster-enriched human microbiota to FC-induced germ-free mice improved gut motility. Collectively, these findings demonstrate a role for microbial abfA cluster in ameliorating FC, establishing principles for genomics-directed probiotic therapies.
Collapse
Affiliation(s)
- Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chenchen Ma
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, China
| | - Shuaiming Jiang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, China
| | - Yufeng Zhang
- Faculty of Dentistry, University of Hong Kong, Hong Kong SAR, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Liming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shi Huang
- Faculty of Dentistry, University of Hong Kong, Hong Kong SAR, China.
| | - Jiachao Zhang
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|