1
|
Wu H, Guzior DV, Martin C, Neugebauer KA, Rzepka MM, Lumeng JC, Quinn RA, de Los Campos G. Longitudinal analyses of infants' microbiome and metabolome reveal microbes and metabolites with seemingly coordinated dynamics. Commun Biol 2024; 7:1506. [PMID: 39543263 DOI: 10.1038/s42003-024-07015-6] [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: 03/25/2024] [Accepted: 10/04/2024] [Indexed: 11/17/2024] Open
Abstract
Population studies have shown that the infant's microbiome and metabolome undergo significant changes in early childhood. However, no previous study has investigated how diverse these changes are across subjects and whether the subject-specific dynamics of some microbes correlate with the over-time dynamics of specific metabolites. Using mixed-effects models, and data from the ABC study, we investigated the early childhood dynamics of fecal microbiome and metabolome and identified 83 amplicon sequence variants (ASVs) and 753 metabolites with seemingly coordinated trajectories. Enrichment analysis of these microbes and molecules revealed eight ASV families and 23 metabolite groups involving 1032 ASV-metabolite pairs with their presence-absence changing in a coordinated fashion. Members of the Lachnospiraceae (464/1032) and metabolites related to cholestane steroids (309/1032) dominated proportional shifts within the fecal microbiome and metabolome as infants aged.
Collapse
Affiliation(s)
- Hao Wu
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Douglas V Guzior
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Christian Martin
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Mass Spectrometry and Metabolomics Core, Michigan State University, East Lansing, MI, USA
| | - Kerri A Neugebauer
- Department of Plant Soil and Microbiology, Michigan State University, East Lansing, MI, USA
| | - Madison M Rzepka
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Julie C Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA.
| | - Gustavo de Los Campos
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
- Department of Statistics and Probability, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
2
|
Iacucci M, Santacroce G, Majumder S, Morael J, Zammarchi I, Maeda Y, Ryan D, Di Sabatino A, Rescigno M, Aburto MR, Cryan JF, Ghosh S. Opening the doors of precision medicine: novel tools to assess intestinal barrier in inflammatory bowel disease and colitis-associated neoplasia. Gut 2024; 73:1749-1762. [PMID: 38851294 PMCID: PMC11422792 DOI: 10.1136/gutjnl-2023-331579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/18/2024] [Indexed: 06/10/2024]
Abstract
Mounting evidence underscores the pivotal role of the intestinal barrier and its convoluted network with diet and intestinal microbiome in the pathogenesis of inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CRC). Moreover, the bidirectional association of the intestinal barrier with the liver and brain, known as the gut-brain axis, plays a crucial role in developing complications, including extraintestinal manifestations of IBD and CRC metastasis. Consequently, barrier healing represents a crucial therapeutic target in these inflammatory-dependent disorders, with barrier assessment predicting disease outcomes, response to therapy and extraintestinal manifestations.New advanced technologies are revolutionising our understanding of the barrier paradigm, enabling the accurate assessment of the intestinal barrier and aiding in unravelling the complexity of the gut-brain axis. Cutting-edge endoscopic imaging techniques, such as ultra-high magnification endocytoscopy and probe-based confocal laser endomicroscopy, are new technologies allowing real-time exploration of the 'cellular' intestinal barrier. Additionally, novel advanced spatial imaging technology platforms, including multispectral imaging, upconversion nanoparticles, digital spatial profiling, optical spectroscopy and mass cytometry, enable a deep and comprehensive assessment of the 'molecular' and 'ultrastructural' barrier. In this promising landscape, artificial intelligence plays a pivotal role in standardising and integrating these novel tools, thereby contributing to barrier assessment and prediction of outcomes.Looking ahead, this integrated and comprehensive approach holds the promise of uncovering new therapeutic targets, breaking the therapeutic ceiling in IBD. Novel molecules, dietary interventions and microbiome modulation strategies aim to restore, reinforce, or modulate the gut-brain axis. These advancements have the potential for transformative and personalised approaches to managing IBD.
Collapse
Affiliation(s)
- Marietta Iacucci
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Giovanni Santacroce
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Snehali Majumder
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Jennifer Morael
- APC Microbiome Ireland, Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| | - Irene Zammarchi
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Yasuharu Maeda
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - David Ryan
- Department of Radiology, School of Medicine, University College Cork, Cork, Ireland
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Maria Rescigno
- IRCSS Humanitas Research Hospital, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Maria R Aburto
- APC Microbiome Ireland, Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| | - Subrata Ghosh
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Li Z, Gong R, Chu H, Zeng J, Chen C, Xu S, Hu L, Gao W, Zhang L, Yuan H, Cheng Z, Wang C, Du M, Zhu Q, Zhang L, Rong L, Hu X, Yang L. A universal plasma metabolites-derived signature predicts cardiovascular disease risk in MAFLD. Atherosclerosis 2024; 392:117526. [PMID: 38581738 DOI: 10.1016/j.atherosclerosis.2024.117526] [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: 12/30/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Metabolic associated fatty liver disease (MAFLD) is a novel concept proposed in 2020, which is more practical for identifying patients with fatty liver disease with high risk of disease progression. Fatty liver is a driver for extrahepatic complications, particularly cardiovascular diseases (CVD). Although the risk of CVD in MAFLD could be predicted by carotid ultrasound test, a very early stage prediction method before the formation of pathological damage is still lacking. METHODS Stool microbiomes and plasma metabolites were compared across 196 well-characterized participants encompassing normal controls, simple MAFLD patients, MAFLD patients with carotid artery pathological changes, and MAFLD patients with diagnosed coronary artery disease (CAD). 16S rDNA sequencing data and untargeted metabolomic profiles were interrogatively analyzed using differential abundance analysis and random forest (RF) machine learning algorithm to identify discriminatory gut microbiomes and metabolomic. RESULTS Characteristic microbial changes in MAFLD patients with CVD risk were represented by the increase of Clostridia and Firmicutes-to-Bacteroidetes ratios. Faecalibacterium was negatively correlated with mean-intima-media thickness (IMT), TC, and TG. Megamonas, Bacteroides, Parabacteroides, and Escherichia were positively correlated with the exacerbation of pathological indexes. MAFLD patients with CVD risk were characterized by the decrease of lithocholic acid taurine conjugate, and the increase of ethylvanillin propylene glycol acetal, both of which had close relationship with Ruminococcus and Gemmiger. Biotin l-sulfoxide had positive correlation with mean-IMT, TG, and weight. The general auxin pesticide beta-naphthoxyacetic acid and the food additive glucosyl steviol were both positively correlated with the increase of mean-IMT. The model combining the metabolite signatures with 9 clinical parameters accurately distinguished MAFLD with CVD risk in the proband and validation cohort. It was found that citral was the most important discriminative metabolite marker, which was validated by both in vitro and in vivo experiments. CONCLUSIONS Simple MAFLD patients and MAFLD patients with CVD risk had divergent gut microbes and plasma metabolites. The predictive model based on metabolites and 9 clinical parameters could effectively discriminate MAFLD patients with CVD risk at a very early stage.
Collapse
Affiliation(s)
- Zhonglin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Rui Gong
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Junchao Zeng
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Can Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Sanping Xu
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lilin Hu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Li Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Hang Yuan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Cheng Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Meng Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Qingjing Zhu
- Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Wuhan Medical Treatment Centre, Wuhan, 430070, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Lin Rong
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Xiaoqing Hu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
4
|
Sivalogan K, Liang D, Accardi C, Diaz-Artiga A, Hu X, Mollinedo E, Ramakrishnan U, Teeny SN, Tran V, Clasen TF, Thompson LM, Sinharoy SS. Human Milk Composition Is Associated with Maternal Body Mass Index in a Cross-Sectional, Untargeted Metabolomics Analysis of Human Milk from Guatemalan Mothers. Curr Dev Nutr 2024; 8:102144. [PMID: 38726027 PMCID: PMC11079463 DOI: 10.1016/j.cdnut.2024.102144] [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: 10/31/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024] Open
Abstract
Background Maternal overweight and obesity has been associated with poor lactation performance including delayed lactogenesis and reduced duration. However, the effect on human milk composition is less well understood. Objectives We evaluated the relationship of maternal BMI on the human milk metabolome among Guatemalan mothers. Methods We used data from 75 Guatemalan mothers who participated in the Household Air Pollution Intervention Network trial. Maternal BMI was measured between 9 and <20 weeks of gestation. Milk samples were collected at a single time point using aseptic collection from one breast at 6 mo postpartum and analyzed using high-resolution mass spectrometry. A cross-sectional untargeted high-resolution metabolomics analysis was performed by coupling hydrophilic interaction liquid chromatography (HILIC) and reverse phase C18 chromatography with mass spectrometry. Metabolic features associated with maternal BMI were determined by a metabolome-wide association study (MWAS), adjusting for baseline maternal age, education, and dietary diversity, and perturbations in metabolic pathways were identified by pathway enrichment analysis. Results The mean age of participants at baseline was 23.62 ± 3.81 y, and mean BMI was 24.27 ± 4.22 kg/m2. Of the total metabolic features detected by HILIC column (19,199 features) and by C18 column (11,594 features), BMI was associated with 1026 HILIC and 500 C18 features. Enriched pathways represented amino acid metabolism, galactose metabolism, and xenobiotic metabolic metabolism. However, no significant features were identified after adjusting for multiple comparisons using the Benjamini-Hochberg false discovery rate procedure (FDRBH < 0.2). Conclusions Findings from this untargeted MWAS indicate that maternal BMI is associated with metabolic perturbations of galactose metabolism, xenobiotic metabolism, and xenobiotic metabolism by cytochrome p450 and biosynthesis of amino acid pathways. Significant metabolic pathway alterations detected in human milk were associated with energy metabolism-related pathways including carbohydrate and amino acid metabolism.This trial was registered at clinicaltrials.gov as NCT02944682.
Collapse
Affiliation(s)
- Kasthuri Sivalogan
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Donghai Liang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Carolyn Accardi
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Anaite Diaz-Artiga
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Xin Hu
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Erick Mollinedo
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Usha Ramakrishnan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
- Department of Environmental Health, College of Public Health, University of Georgia, Athens, GA, United States
| | - Sami Nadeem Teeny
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - ViLinh Tran
- Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Thomas F Clasen
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Lisa M Thompson
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Sheela S Sinharoy
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| |
Collapse
|
5
|
Muller E, Shiryan I, Borenstein E. Multi-omic integration of microbiome data for identifying disease-associated modules. Nat Commun 2024; 15:2621. [PMID: 38521774 PMCID: PMC10960825 DOI: 10.1038/s41467-024-46888-3] [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: 01/22/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
Multi-omic studies of the human gut microbiome are crucial for understanding its role in disease across multiple functional layers. Nevertheless, integrating and analyzing such complex datasets poses significant challenges. Most notably, current analysis methods often yield extensive lists of disease-associated features (e.g., species, pathways, or metabolites), without capturing the multi-layered structure of the data. Here, we address this challenge by introducing "MintTea", an intermediate integration-based approach combining canonical correlation analysis extensions, consensus analysis, and an evaluation protocol. MintTea identifies "disease-associated multi-omic modules", comprising features from multiple omics that shift in concord and that collectively associate with the disease. Applied to diverse cohorts, MintTea captures modules with high predictive power, significant cross-omic correlations, and alignment with known microbiome-disease associations. For example, analyzing samples from a metabolic syndrome study, MintTea identifies a module with serum glutamate- and TCA cycle-related metabolites, along with bacterial species linked to insulin resistance. In another dataset, MintTea identifies a module associated with late-stage colorectal cancer, including Peptostreptococcus and Gemella species and fecal amino acids, in line with these species' metabolic activity and their coordinated gradual increase with cancer development. This work demonstrates the potential of advanced integration methods in generating systems-level, multifaceted hypotheses underlying microbiome-disease interactions.
Collapse
Affiliation(s)
- Efrat Muller
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Itamar Shiryan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Elhanan Borenstein
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel.
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
- Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
6
|
García-Manríquez N, Lozano C, Muñoz A, Morales MF, Giacaman RA. Anticaries properties of natural berries: systematic literature review. Nutr Rev 2024; 82:302-317. [PMID: 37352393 DOI: 10.1093/nutrit/nuad063] [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] [Indexed: 06/25/2023] Open
Abstract
CONTEXT Anticariogenic properties have been ascribed to polyphenolic compounds present in high concentrations in numerous fruits. Berries, in particular, have been reported as potentially having an inhibitory effect on the dental biofilm and subsequently on caries, but the evidence is unclear. OBJECTIVE The objective of this review was to explore the literature and summarize the evidence for berries having an inhibitory effect on the dental biofilm and an anticariogenic effect. DATA SOURCES Following Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines, the PubMed, Web of Science, and SCOPUS databases were scanned using predefined and accessible terms, with a search strategy based on a structured PICO question. DATA EXTRACTION After article selection, 23 studies met the inclusion criteria, most of them being in vitro studies. A risk assessment was performed, and data were extracted and presented in a table for qualitative analysis. DATA ANALYSIS Meta-analyses were conducted using standardized mean differences (SMDs) with a 95% confidence interval (CI) by Review manager 5.4. RESULTS Only 3 types of berries were found to have a reported anticaries effect: grape seed extract (GSE), cranberry, and sour cherry. Nine studies that fulfilled the eligibility criteria were subjected to quantitative analysis. Meta-analyses showed GSE was associated with enhanced remineralization of dental enamel (SMD = .96 95% CI [.45, 1.46], P < .0002) and of dentin (SMD = .65 95% CI [.13, 1.17], P = .01). Cranberry extracts positively influenced the cariogenic dental biofilm by decreasing the biofilm biomass (SMD = -2.23 95% CI [-4.40, -.05], P = .04), and biovolume (SMD = -2.86 95% CI [-4.34, -1.37], P = .0002), and increasing the biofilm pH (SMD = 7.9 95% CI [3.49, 12.31], P < .0004). CONCLUSION Within the limitations of this systematic review and metaanalysis, GSE and cranberries or their active compounds could represent an alternative for caries management. Further clinical trials are needed to verify this effect in a clinical setting. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42020223579.
Collapse
Affiliation(s)
- Natalia García-Manríquez
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca, Chile
| | - Carla Lozano
- Biochemistry and Oral Biology Laboratory, Research Institute for Dental Sciences, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Ana Muñoz
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca, Chile
| | - María Fernanda Morales
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca, Chile
| | - Rodrigo A Giacaman
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca, Chile
- Biochemistry and Oral Biology Laboratory, Research Institute for Dental Sciences, Faculty of Dentistry, University of Chile, Santiago, Chile
- In teruniversity Center for Healthy Aging, Chilean State Universities, Chile
| |
Collapse
|
7
|
Muller E, Shiryan I, Borenstein E. Multi-omic integration of microbiome data for identifying disease-associated modules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.03.547607. [PMID: 37461534 PMCID: PMC10349976 DOI: 10.1101/2023.07.03.547607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The human gut microbiome is a complex ecosystem with profound implications for health and disease. This recognition has led to a surge in multi-omic microbiome studies, employing various molecular assays to elucidate the microbiome's role in diseases across multiple functional layers. However, despite the clear value of these multi-omic datasets, rigorous integrative analysis of such data poses significant challenges, hindering a comprehensive understanding of microbiome-disease interactions. Perhaps most notably, multiple approaches, including univariate and multivariate analyses, as well as machine learning, have been applied to such data to identify disease-associated markers, namely, specific features (e.g., species, pathways, metabolites) that are significantly altered in disease state. These methods, however, often yield extensive lists of features associated with the disease without effectively capturing the multi-layered structure of multi-omic data or offering clear, interpretable hypotheses about underlying microbiome-disease mechanisms. Here, we address this challenge by introducing MintTea - an intermediate integration-based method for analyzing multi-omic microbiome data. MintTea combines a canonical correlation analysis (CCA) extension, consensus analysis, and an evaluation protocol to robustly identify disease-associated multi-omic modules. Each such module consists of a set of features from the various omics that both shift in concord, and collectively associate with the disease. Applying MintTea to diverse case-control cohorts with multi-omic data, we show that this framework is able to capture modules with high predictive power for disease, significant cross-omic correlations, and alignment with known microbiome-disease associations. For example, analyzing samples from a metabolic syndrome (MS) study, we found a MS-associated module comprising of a highly correlated cluster of serum glutamate- and TCA cycle-related metabolites, as well as bacterial species previously implicated in insulin resistance. In another cohort, we identified a module associated with late-stage colorectal cancer, featuring Peptostreptococcus and Gemella species and several fecal amino acids, in agreement with these species' reported role in the metabolism of these amino acids and their coordinated increase in abundance during disease development. Finally, comparing modules identified in different datasets, we detected multiple significant overlaps, suggesting common interactions between microbiome features. Combined, this work serves as a proof of concept for the potential benefits of advanced integration methods in generating integrated multi-omic hypotheses underlying microbiome-disease interactions and a promising avenue for researchers seeking systems-level insights into coherent mechanisms governing microbiome-related diseases.
Collapse
|
8
|
Lorenzo-Rebenaque L, Casto-Rebollo C, Diretto G, Frusciante S, Rodríguez JC, Ventero MP, Molina-Pardines C, Vega S, Marin C, Marco-Jiménez F. Modulation of Caecal Microbiota and Metabolome Profile in Salmonella-Infected Broilers by Phage Therapy. Int J Mol Sci 2023; 24:15201. [PMID: 37894882 PMCID: PMC10607084 DOI: 10.3390/ijms242015201] [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: 09/20/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteriophage therapy is considered one of the most promising tools to control zoonotic bacteria, such as Salmonella, in broiler production. Phages exhibit high specificity for their targeted bacterial hosts, causing minimal disruption to the niche microbiota. However, data on the gut environment's response to phage therapy in poultry are limited. This study investigated the influence of Salmonella phage on host physiology through caecal microbiota and metabolome modulation using high-throughput 16S rRNA gene sequencing and an untargeted metabolomics approach. We employed 24 caecum content samples and 24 blood serum samples from 4-, 5- and 6-week-old broilers from a previous study where Salmonella phages were administered via feed in Salmonella-infected broilers, which were individually weighed weekly. Phage therapy did not affect the alpha or beta diversity of the microbiota. Specifically, we observed changes in the relative abundance of 14 out of the 110 genera using the PLS-DA and Bayes approaches. On the other hand, we noted changes in the caecal metabolites (63 up-accumulated and 37 down-accumulated out of the 1113 caecal metabolites). Nevertheless, the minimal changes in blood serum suggest a non-significant physiological response. The application of Salmonella phages under production conditions modulates the caecal microbiome and metabolome profiles in broilers without impacting the host physiology in terms of growth performance.
Collapse
Affiliation(s)
- Laura Lorenzo-Rebenaque
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Calle Santiago Ramón y Cajal 20, Alfara del Patriarca, 45115 Valencia, Spain; (L.L.-R.); (S.V.); (C.M.)
| | - Cristina Casto-Rebollo
- Institute for Animal Science and Technology, Universitat Politècnica de València, 46022 Valencia, Spain;
| | - Gianfranco Diretto
- Italian Agency for New Technologies, Energy and Sustainable Development (ENEA), Biotechnology Laboratory, Centro Ricerche Casaccia, Via Anguillarese, 301, Santa Maria di Galeria, 00123 Rome, Italy; (G.D.); (S.F.)
| | - Sarah Frusciante
- Italian Agency for New Technologies, Energy and Sustainable Development (ENEA), Biotechnology Laboratory, Centro Ricerche Casaccia, Via Anguillarese, 301, Santa Maria di Galeria, 00123 Rome, Italy; (G.D.); (S.F.)
| | - Juan Carlos Rodríguez
- Microbiology Department, Dr. Balmis University General Hospital, Microbiology Division, Miguel Hernández University, ISABIAL, 03010 Alicante, Spain;
| | - María-Paz Ventero
- Microbiology Department, Dr. Balmis University General Hospital, ISABIAL, 03010 Alicante, Spain; (M.-P.V.); (C.M.-P.)
| | - Carmen Molina-Pardines
- Microbiology Department, Dr. Balmis University General Hospital, ISABIAL, 03010 Alicante, Spain; (M.-P.V.); (C.M.-P.)
| | - Santiago Vega
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Calle Santiago Ramón y Cajal 20, Alfara del Patriarca, 45115 Valencia, Spain; (L.L.-R.); (S.V.); (C.M.)
| | - Clara Marin
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Biomedical Research Institute, Faculty of Veterinary Medicine, Cardenal Herrera-CEU University, CEU Universities, Calle Santiago Ramón y Cajal 20, Alfara del Patriarca, 45115 Valencia, Spain; (L.L.-R.); (S.V.); (C.M.)
| | - Francisco Marco-Jiménez
- Institute for Animal Science and Technology, Universitat Politècnica de València, 46022 Valencia, Spain;
| |
Collapse
|
9
|
Hammond TC, Green SJ, Jacobs Y, Chlipala GE, Xing X, Heil S, Chen A, Aware C, Flemister A, Stromberg A, Balchandani P, Lin AL. Gut microbiome association with brain imaging markers, APOE genotype, calcium and vegetable intakes, and obesity in healthy aging adults. Front Aging Neurosci 2023; 15:1227203. [PMID: 37736325 PMCID: PMC10510313 DOI: 10.3389/fnagi.2023.1227203] [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: 05/22/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Introduction Advanced age is a significant factor in changes to brain physiology and cognitive functions. Recent research has highlighted the critical role of the gut microbiome in modulating brain functions during aging, which can be influenced by various factors such as apolipoprotein E (APOE) genetic variance, body mass index (BMI), diabetes, and dietary intake. However, the associations between the gut microbiome and these factors, as well as brain structural, vascular, and metabolic imaging markers, have not been well explored. Methods We recruited 30 community dwelling older adults between age 55-85 in Kentucky. We collected the medical history from the electronic health record as well as the Dietary Screener Questionnaire. We performed APOE genotyping with an oral swab, gut microbiome analysis using metagenomics sequencing, and brain structural, vascular, and metabolic imaging using MRI. Results Individuals with APOE e2 and APOE e4 genotypes had distinct microbiota composition, and higher level of pro-inflammatory microbiota were associated higher BMI and diabetes. In contrast, calcium- and vegetable-rich diets were associated with microbiota that produced short chain fatty acids leading to an anti-inflammatory state. We also found that important gut microbial butyrate producers were correlated with the volume of the thalamus and corpus callosum, which are regions of the brain responsible for relaying and processing information. Additionally, putative proinflammatory species were negatively correlated with GABA production, an inhibitory neurotransmitter. Furthermore, we observed that the relative abundance of bacteria from the family Eggerthellaceae, equol producers, was correlated with white matter integrity in tracts connecting the brain regions related to language, memory, and learning. Discussion These findings highlight the importance of gut microbiome association with brain health in aging population and could have important implications aimed at optimizing healthy brain aging through precision prebiotic, probiotic or dietary interventions.
Collapse
Affiliation(s)
- Tyler C. Hammond
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Stefan J. Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, IL, United States
| | - Yael Jacobs
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - George E. Chlipala
- Research Informatics Core, University of Illinois Chicago, Chicago, IL, United States
| | - Xin Xing
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Computer Science, University of Kentucky, Lexington, KY, United States
- Roy Blunt NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Radiology, University of Missouri, Columbia, MO, United States
| | - Sally Heil
- School of Medicine, University of Missouri, Columbia, MO, United States
| | - Anna Chen
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Chetan Aware
- Roy Blunt NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Radiology, University of Missouri, Columbia, MO, United States
| | - Abeoseh Flemister
- Roy Blunt NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Radiology, University of Missouri, Columbia, MO, United States
| | - Arnold Stromberg
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Priti Balchandani
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ai-Ling Lin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Roy Blunt NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Radiology, University of Missouri, Columbia, MO, United States
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, United States
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
10
|
Grosicki GJ, Pugh J, Wosinska L, Quilter K, Mattimoe D, Allen J, Joyce SA, O'Sullivan O, Close GL. Ultra-Endurance triathlon competition shifts fecal metabolome independent of changes to microbiome composition. J Appl Physiol (1985) 2023; 135:549-558. [PMID: 37391884 DOI: 10.1152/japplphysiol.00024.2023] [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: 01/13/2023] [Revised: 05/22/2023] [Accepted: 06/19/2023] [Indexed: 07/02/2023] Open
Abstract
Understanding changes to gut microbiota composition and metabolic output in response to acute exercise may be necessary for understanding the mechanisms mediating the long-term health and performance benefits of exercise. Our primary objective was to characterize acute changes in the fecal microbiome and metabolome following participation in an ultra-endurance (3.9 km swim, 180.2 km bike, 42.2 km run) triathlon. An exploratory aim was to determine associations between athlete-specific factors [race performance (i.e., completion time) and lifetime years of endurance training] with pre-race gut microbiota and metabolite profiles. Stool samples from 12 triathletes (9 males/3 females; 43 ± 14 yr, 23 ± 2 kg/m2) were collected ≤48 h before and the first bowel movement following race completion. Intra- and inter-individual diversity of bacterial species and individual bacterial taxa were unaltered following race completion (P > 0.05). However, significant reductions (P < 0.05) in free and secondary bile acids [deoxycholic acid (DCA), 12-keto-lithocholic acid (12-ketoLCA)] and short-chain fatty acids (butyric and pivalic acids), and significant increases (P < 0.05) in long-chain fatty acids (oleic and palmitoleic acids) were observed. Exploratory analyses revealed several associations between pre-race bacterial taxa and fecal metabolites with race performance and lifetime history of endurance training (P < 0.05). These findings suggest that 1) acute ultra-endurance exercise shifts microbial metabolism independent of changes to community composition and 2) athlete performance level and training history relate to resting-state gut microbial ecology.NEW & NOTEWORTHY This is the first study to characterize acute changes in gut microbial ecology and metabolism following an ultra-endurance triathlon. We demonstrate changes in gut microbial community function, but not structure, as well as several associations between gut microbiome and fecal metabolome characteristics with race completion time and lifetime history of endurance training. These data add to a small but growing body of literature seeking to characterize the acute and chronic effects of exercise on the gut microbial ecosystem.
Collapse
Affiliation(s)
- Gregory J Grosicki
- Biodynamics and Human Performance Center, Georgia Southern University - Armstrong Campus, Savannah, Georgia, United States
| | - Jamie Pugh
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Laura Wosinska
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Karina Quilter
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Darragh Mattimoe
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jacob Allen
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States
| | - Susan A Joyce
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Orla O'Sullivan
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Ireland
| | - Graeme L Close
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
11
|
Xu D, Wu Q, Liu W, Hu G, Meng H, Wang J. Therapeutic efficacy and underlying mechanisms of Gastrodia elata polysaccharides on dextran sulfate sodium-induced inflammatory bowel disease in mice: Modulation of the gut microbiota and improvement of metabolic disorders. Int J Biol Macromol 2023; 248:125919. [PMID: 37481182 DOI: 10.1016/j.ijbiomac.2023.125919] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory gastrointestinal disease, and an imbalance in the gut microbiota is a critical factor in its development. Gastrodia elata (G. elata), an Orchidaceae plant, is recognized for its nutritional and medicinal value. Studies have shown that G. elata polysaccharides (GBP) have anti-inflammatory properties that may ameliorate IBD. However, the therapeutic effects of GBP on gut microbiota metabolism remain unknown. Therefore, we aimed to examine the therapeutic potential of G. elata extract and GBP in dextran sulfate sodium (DSS)-induced IBD mice. GBP demonstrated the best therapeutic effect by reducing IBD symptoms in mice to the greatest extent. Administering GBP resulted in significant increases in the relative abundances of bacteria with potential anti-inflammatory effects, such as Ligilactobacillus and Alloprevotella, and decreases in the levels of bacteria associated with proinflammatory responses, such as Bacteroides and Escherichia-Shigella. Furthermore, 36 significant differential metabolites between the model and GBP groups were identified in feces, which were mainly enriched in amino acid metabolism, including tryptophan and cysteine, vitamin B6 metabolism and steroid hormone biosynthesis. Consequently, investigating the metabolic regulation of the gut microbiota is a promising approach to evaluate the therapeutic effect of GBP on IBD.
Collapse
Affiliation(s)
- Di Xu
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China.
| | - Qingyan Wu
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Wenya Liu
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Guannan Hu
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Huihui Meng
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Junsong Wang
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China.
| |
Collapse
|
12
|
Hill EB, Konigsberg IR, Ir D, Frank DN, Jambal P, Litkowski EM, Lange EM, Lange LA, Ostendorf DM, Scorsone JJ, Wayland L, Bing K, MacLean PS, Melanson EL, Bessesen DH, Catenacci VA, Stanislawski MA, Borengasser SJ. The Microbiome, Epigenome, and Diet in Adults with Obesity during Behavioral Weight Loss. Nutrients 2023; 15:3588. [PMID: 37630778 PMCID: PMC10458964 DOI: 10.3390/nu15163588] [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: 06/17/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 08/27/2023] Open
Abstract
Obesity has been linked to the gut microbiome, epigenome, and diet, yet these factors have not been studied together during obesity treatment. Our objective was to evaluate associations among gut microbiota (MB), DNA methylation (DNAme), and diet prior to and during a behavioral weight loss intervention. Adults (n = 47, age 40.9 ± 9.7 years, body mass index (BMI) 33.5 ± 4.5 kg/m2, 77% female) with data collected at baseline (BL) and 3 months (3 m) were included. Fecal MB was assessed via 16S sequencing and whole blood DNAme via the Infinium EPIC array. Food group and nutrient intakes and Healthy Eating Index (HEI) scores were calculated from 7-day diet records. Linear models were used to test for the effect of taxa relative abundance on DNAme and diet cross-sectionally at each time point, adjusting for confounders and a false discovery rate of 5%. Mean weight loss was 6.2 ± 3.9% at 3 m. At BL, one MB taxon, Ruminiclostridium, was associated with DNAme of the genes COL20A1 (r = 0.651, p = 0.029), COL18A1 (r = 0.578, p = 0.044), and NT5E (r = 0.365, p = 0.043). At 3 m, there were 14 unique MB:DNAme associations, such as Akkermansia with DNAme of GUSB (r = -0.585, p = 0.003), CRYL1 (r = -0.419, p = 0.007), C9 (r = -0.439, p = 0.019), and GMDS (r = -0.559, p = 0.046). Among taxa associated with DNAme, no significant relationships were seen with dietary intakes of relevant nutrients, food groups, or HEI scores. Our findings indicate that microbes linked to mucin degradation, short-chain fatty acid production, and body weight are associated with DNAme of phenotypically relevant genes. These relationships offer an initial understanding of the possible routes by which alterations in gut MB may influence metabolism during weight loss.
Collapse
Affiliation(s)
- Emily B. Hill
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (E.B.H.)
| | - Iain R. Konigsberg
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (I.R.K.)
| | - Diana Ir
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Purevsuren Jambal
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (E.B.H.)
| | - Elizabeth M. Litkowski
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (I.R.K.)
- Department of Epidemiology, University of Colorado School of Public Health, Aurora, CO 80045, USA
- Eastern Colorado Veterans Affairs Geriatric Research, Education, and Clinical Center, Aurora, CO 80045, USA
| | - Ethan M. Lange
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (I.R.K.)
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, CO 80045, USA
| | - Leslie A. Lange
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (I.R.K.)
- Eastern Colorado Veterans Affairs Geriatric Research, Education, and Clinical Center, Aurora, CO 80045, USA
| | - Danielle M. Ostendorf
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jared J. Scorsone
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Liza Wayland
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristen Bing
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul S. MacLean
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Edward L. Melanson
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel H. Bessesen
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Victoria A. Catenacci
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Maggie A. Stanislawski
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (I.R.K.)
| | - Sarah J. Borengasser
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (E.B.H.)
| |
Collapse
|
13
|
Bloemendaal M, Veniaminova E, Anthony DC, Gorlova A, Vlaming P, Khairetdinova A, Cespuglio R, Lesch KP, Arias Vasquez A, Strekalova T. Serotonin Transporter (SERT) Expression Modulates the Composition of the Western-Diet-Induced Microbiota in Aged Female Mice. Nutrients 2023; 15:3048. [PMID: 37447374 PMCID: PMC10346692 DOI: 10.3390/nu15133048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Background. The serotonin transporter (SERT), highly expressed in the gut and brain, is implicated in metabolic processes. A genetic variant of the upstream regulatory region of the SLC6A4 gene encoding SERT, the so-called short (s) allele, in comparison with the long (l) allele, results in the decreased function of this transporter, altered serotonergic regulation, an increased risk of psychiatric pathology and type-2 diabetes and obesity, especially in older women. Aged female mice with the complete (Sert-/-: KO) or partial (Sert+/-: HET) loss of SERT exhibit more pronounced negative effects following their exposure to a Western diet in comparison to wild-type (Sert+/+: WT) animals. Aims. We hypothesized that these effects might be mediated by an altered gut microbiota, which has been shown to influence serotonin metabolism. We performed V4 16S rRNA sequencing of the gut microbiota in 12-month-old WT, KO and HET female mice that were housed on a control or Western diet for three weeks. Results. The relative abundance of 11 genera was increased, and the abundance of 6 genera was decreased in the Western-diet-housed mice compared to the controls. There were correlations between the abundance of Streptococcus and Ruminococcaceae_UCG-014 and the expression of the pro-inflammatory marker Toll-like-Receptor 4 (Tlr4) in the dorsal raphe, as well as the expression of the mitochondrial activity marker perixome-proliferator-activated-receptor-cofactor-1b (Ppargc1b) in the prefrontal cortex. Although there was no significant impact of genotype on the microbiota in animals fed with the Control diet, there were significant interactions between diet and genotype. Following FDR correction, the Western diet increased the relative abundance of Intestinimonas and Atopostipes in the KO animals, which was not observed in the other groups. Erysipelatoclostridium abundance was increased by the Western diet in the WT group but not in HET or KO animals. Conclusions. The enhanced effects of a challenge with a Western diet in SERT-deficient mice include the altered representation of several gut genera, such as Intestinimonas, Atopostipes and Erysipelatoclostridium, which are also implicated in serotonergic and lipid metabolism. The manipulation of these genera may prove useful in individuals with the short SERT allele.
Collapse
Affiliation(s)
- Mirjam Bloemendaal
- Departments of Psychiatry & Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.V.); (A.A.V.)
| | - Ekaterina Veniaminova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
| | | | - Anna Gorlova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
| | - Priscilla Vlaming
- Departments of Psychiatry & Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.V.); (A.A.V.)
| | - Adel Khairetdinova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
| | - Raymond Cespuglio
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
- Neuroscience Research Center of Lyon, Claude-Bernard Lyon-1 University, 69500 Bron, France
| | - Klaus Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97080 Würzburg, Germany; (K.P.L.); (T.S.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Alejandro Arias Vasquez
- Departments of Psychiatry & Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.V.); (A.A.V.)
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97080 Würzburg, Germany; (K.P.L.); (T.S.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
14
|
Abstract
Cardiometabolic disease comprises cardiovascular and metabolic dysfunction and underlies the leading causes of morbidity and mortality, both within the United States and worldwide. Commensal microbiota are implicated in the development of cardiometabolic disease. Evidence suggests that the microbiome is relatively variable during infancy and early childhood, becoming more fixed in later childhood and adulthood. Effects of microbiota, both during early development, and in later life, may induce changes in host metabolism that modulate risk mechanisms and predispose toward the development of cardiometabolic disease. In this review, we summarize the factors that influence gut microbiome composition and function during early life and explore how changes in microbiota and microbial metabolism influence host metabolism and cardiometabolic risk throughout life. We highlight limitations in current methodology and approaches and outline state-of-the-art advances, which are improving research and building toward refined diagnosis and treatment options in microbiome-targeted therapies.
Collapse
Affiliation(s)
- Curtis L Gabriel
- Division of Gastroenterology, Hepatology and Nutrition (C.L.G.), Vanderbilt University Medical Center, Nashville
- Tennessee Center for AIDS Research (C.L.G.), Vanderbilt University Medical Center, Nashville
| | - Jane F Ferguson
- Division of Cardiovascular Medicine (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Microbiome Innovation Center (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Institute for Infection, Immunology, and Inflammation (J.F.F.), Vanderbilt University Medical Center, Nashville
| |
Collapse
|
15
|
Song Z, Ohnishi Y, Osada S, Gan L, Jiang J, Hu Z, Kumeta H, Kumaki Y, Yokoi Y, Nakamura K, Ayabe T, Yamauchi K, Aizawa T. Application of Benchtop NMR for Metabolomics Study Using Feces of Mice with DSS-Induced Colitis. Metabolites 2023; 13:metabo13050611. [PMID: 37233652 DOI: 10.3390/metabo13050611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/22/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Nuclear magnetic resonance (NMR)-based metabolomics, which comprehensively measures metabolites in biological systems and investigates their response to various perturbations, is widely used in research to identify biomarkers and investigate the pathogenesis of underlying diseases. However, further applications of high-field superconducting NMR for medical purposes and field research are restricted by its high cost and low accessibility. In this study, we applied a low-field, benchtop NMR spectrometer (60 MHz) employing a permanent magnet to characterize the alterations in the metabolic profile of fecal extracts obtained from dextran sodium sulfate (DSS)-induced ulcerative colitis model mice and compared them with the data acquired from high-field NMR (800 MHz). Nineteen metabolites were assigned to the 60 MHz 1H NMR spectra. Non-targeted multivariate analysis successfully discriminated the DSS-induced group from the healthy control group and showed high comparability with high-field NMR. In addition, the concentration of acetate, identified as a metabolite with characteristic behavior, could be accurately quantified using a generalized Lorentzian curve fitting method based on the 60 MHz NMR spectra.
Collapse
Affiliation(s)
- Zihao Song
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Yuki Ohnishi
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | | | - Li Gan
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Jiaxi Jiang
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Zhiyan Hu
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Hiroyuki Kumeta
- Advanced NMR Facility, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Yasuhiro Kumaki
- High-Resolution NMR Laboratory, Graduate School of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Yuki Yokoi
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Kiminori Nakamura
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Tokiyoshi Ayabe
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
| | - Kazuo Yamauchi
- Instrumental Analysis Section, Okinawa Institute of Science and Technology, Onna 904-0495, Japan
| | - Tomoyasu Aizawa
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo 060-0808, Japan
- Advanced NMR Facility, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0808, Japan
| |
Collapse
|
16
|
Thompson SC, Ford AL, Moothedan EJ, Stafford LS, Garrett TJ, Dahl WJ, Conesa A, Gonzalez CF, Lorca GL. Identification of food and nutrient components as predictors of Lactobacillus colonization. Front Nutr 2023; 10:1118679. [PMID: 37153913 PMCID: PMC10160632 DOI: 10.3389/fnut.2023.1118679] [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: 12/07/2022] [Accepted: 03/31/2023] [Indexed: 05/10/2023] Open
Abstract
A previous double-blind, randomized clinical trial of 42 healthy individuals conducted with Lactobacillus johnsonii N6.2 found that the probiotic's mechanistic tryptophan pathway was significantly modified when the data was stratified based on the individuals' lactic acid bacteria (LAB) stool content. These results suggest that confounding factors such as dietary intake which impact stool LAB content may affect the response to the probiotic treatment. Using dietary intake, serum metabolite, and stool LAB colony forming unit (CFU) data from a previous clinical trial, the relationships between diet, metabolic response, and fecal LAB were assessed. The diets of subject groups with high vs. low CFUs of LAB/g of wet stool differed in their intakes of monounsaturated fatty acids, vegetables, proteins, and dairy. Individuals with high LAB consumed greater amounts of cheese, fermented meats, soy, nuts and seeds, alcoholic beverages, and oils whereas individuals with low LAB consumed higher amounts of tomatoes, starchy vegetables, and poultry. Several dietary variables correlated with LAB counts; positive correlations were determined for nuts and seeds, fish high in N-3 fatty acids, soy, and processed meats, and negative correlations to consumption of vegetables including tomatoes. Using machine learning, predictors of LAB count included cheese, nuts and seeds, fish high in N-3 fatty acids, and erucic acid. Erucic acid alone accurately predicted LAB categorization, and was shown to be utilized as a sole fatty acid source by several Lactobacillus species regardless of their mode of fermentation. Several metabolites were significantly upregulated in each group based on LAB titers, notably polypropylene glycol, caproic acid, pyrazine, and chondroitin sulfate; however, none were correlated with the dietary intake variables. These findings suggest that dietary variables may drive the presence of LAB in the human gastrointestinal tract and potentially impact response to probiotic interventions.
Collapse
Affiliation(s)
- Sharon C. Thompson
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Amanda L. Ford
- Department of Food Science and Human Nutrition, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Elijah J. Moothedan
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Lauren S. Stafford
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Timothy J. Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Wendy J. Dahl
- Department of Food Science and Human Nutrition, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Ana Conesa
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
- Institute for Integrative Systems Biology, Spanish National Research CouncilValencia, Spain
| | - Claudio F. Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Graciela L. Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
17
|
Ishimwe JA, Ferguson JF, Kirabo A. Sex Differences in Fatty Acid Metabolism and Blood Pressure Response to Dietary Salt in Humans. CARDIOGENETICS 2023; 13:33-46. [PMID: 38605973 PMCID: PMC11008634 DOI: 10.3390/cardiogenetics13010005] [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] [Indexed: 04/13/2024] Open
Abstract
Salt sensitivity is a trait in which high dietary sodium (Na+) intake causes an increase in blood pressure (BP). We previously demonstrated that in the gut, elevated dietary Na+ causes dysbiosis. The mechanistic interplay between excess dietary Na+-induced alteration in the gut microbiome and sex differences is less understood. The goal of this study was to identify novel metabolites in sex differences and blood pressure in response to a high dietary Na+ intake. We performed stool and plasma metabolomics analysis and measured the BP of human volunteers with salt intake above or below the American Heart Association recommendations. We also performed RNA sequencing on human monocytes treated with high salt in vitro. The relationship between BP and dietary Na+ intake was different in women and men. Network analysis revealed that fatty acids as top subnetworks differentially changed with salt intake. We found that women with high dietary Na+ intake have high levels of arachidonic acid related metabolism, suggesting a role in sex differences of the blood pressure response to Na+. The exposure of monocytes to high salt in vitro upregulates the transcription of fatty acid receptors and arachidonic acid-related genes. These findings provide potentially novel insights into metabolic changes underlying gut dysbiosis and inflammation in salt sensitivity of BP.
Collapse
Affiliation(s)
- Jeanne A. Ishimwe
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Nashville, TN 37235, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Medical Center, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
18
|
Comparison of fecal and blood metabolome reveals inconsistent associations of the gut microbiota with cardiometabolic diseases. Nat Commun 2023; 14:571. [PMID: 36732517 PMCID: PMC9894915 DOI: 10.1038/s41467-023-36256-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Blood metabolome is commonly used in human studies to explore the associations of gut microbiota-derived metabolites with cardiometabolic diseases. Here, in a cohort of 1007 middle-aged and elderly adults with matched fecal metagenomic (149 species and 214 pathways) and paired fecal and blood targeted metabolomics data (132 metabolites), we find disparate associations with taxonomic composition and microbial pathways when using fecal or blood metabolites. For example, we observe that fecal, but not blood butyric acid significantly associates with both gut microbiota and prevalent type 2 diabetes. These findings are replicated in an independent validation cohort involving 103 adults. Our results suggest that caution should be taken when inferring microbiome-cardiometabolic disease associations from either blood or fecal metabolome data.
Collapse
|
19
|
Cheon S, Kim G, Bae JH, Lee DH, Seong H, Kim DH, Han JS, Lim SY, Han NS. Comparative analysis of prebiotic effects of four oligosaccharides using in vitro gut model: digestibility, microbiome, and metabolome changes. FEMS Microbiol Ecol 2023; 99:6979797. [PMID: 36623850 PMCID: PMC9875365 DOI: 10.1093/femsec/fiad002] [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: 04/12/2022] [Revised: 11/09/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Fructooligosaccharides (FOS), Ad-fructooligosaccharides (Ad-FOS), resistant maltodextrin (RMD), and maltooligosaccharides (MOS) are commercially available prebiotic oligosaccharides. In this study, the effects of prebiotics on the human gut microbial ecosystem were evaluated using an in vitro gut model. FOS and Ad-FOS showed tolerance to digestion, whereas RMD and MOS showed moderate digestion by digestive enzymes. In in vitro fecal fermentation, Bifidobacterium spp. increased in the following order: FOS, Ad-FOS, MOS, and RMD, whereas Bacteroides spp. increased in RMD medium. Bacteroides xylanisolvens exhibited cross-feeding by enabling the growth of other beneficial bacteria during co-culture in RMD medium. In metabolome analysis, total short-chain fatty acids (SCFAs) were highly produced in the following order: RMD, FOS, MOS, and Ad-FOS; acetate in the order of FOS, MOS/RMD, and Ad-FOS; butyrate in the order of RMD, MOS, FOS, and Ad-FOS; and propionate only in RMD. In addition, the conversion of betaine to trimethylamine was rarely affected in the following order: MOS, RMD, FOS, and Ad-FOS. Lastly, the four oligosaccharides inhibited the adhesion of pathogenic Escherichia coli to human epithelial cells to a similar extent. The comparative analysis results obtained in this study will provide comprehensive information of these substances to manufacturers and customers.
Collapse
Affiliation(s)
| | | | - Jae-Han Bae
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Dong Hyeon Lee
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hyunbin Seong
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Da Hye Kim
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Jung-Sook Han
- Samyang Corp. 295 Pangyo-ro, Samyang Corporation Food Biotech R&D Center, Bundang-gu, Seongnam-si Gyeonggi-do 13488, Republic of Korea
| | - Su-Youn Lim
- Samyang Corp. 295 Pangyo-ro, Samyang Corporation Food Biotech R&D Center, Bundang-gu, Seongnam-si Gyeonggi-do 13488, Republic of Korea
| | - Nam Soo Han
- Corresponding author: Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea. Tel: +82-43-261-2567; Fax: +82-43-271-4412; E-mail:
| |
Collapse
|
20
|
Jang HH, Noh H, Kim G, Cho SY, Kim HJ, Choe JS, Kim J, Scalbert A, Gunter MJ, Kwon O, Kim H. Differences in dietary patterns related to metabolic health by gut microbial enterotypes of Korean adults. Front Nutr 2023; 9:1045397. [PMID: 36687725 PMCID: PMC9853283 DOI: 10.3389/fnut.2022.1045397] [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: 09/15/2022] [Accepted: 12/13/2022] [Indexed: 01/07/2023] Open
Abstract
Diet has a profound impact on the progression of metabolic syndrome (MetS) into various diseases. The gut microbiota could modulate the effect of diet on metabolic health. We examined whether dietary patterns related to MetS differed according to gut microbial enterotypes among 348 Korean adults aged 18-60 years recruited between 2018∼2021 in a cross-sectional study. The enterotype of each participant was identified based on 16S rRNA gut microbiota data. The main dietary pattern predicting MetS (MetS-DP) of each enterotype was derived using reduced-rank regression (RRR) models. In the RRR models, 27 food group intakes assessed by a semi-quantitative food frequency questionnaire and MetS prediction markers including triglyceride to high-density lipoprotein cholesterol (TG/HDL) ratio and homeostatic model assessment for insulin resistance (HOMA-IR) were used as predictor and response variables, respectively. The MetS-DP extracted in Bacteroides enterotype (B-type) was characterized by high consumption of refined white rice and low consumption of eggs, vegetables, and mushrooms. The MetS-DP derived among Prevotella enterotype (P-type) was characterized by a high intake of sugary food and low intakes of bread, fermented legumes, and fermented vegetables. The MetS-DP of B-type was positively associated with metabolic unhealthy status (OR T3 vs. T1 = 3.5; 95% CI = 1.5-8.2), comparing the highest tertile to the lowest tertile. Although it was not significantly associated with overall metabolic unhealthy status, the MetS-DP of P-type was positively associated with hyperglycemia risk (OR T3 vs. T1 = 6.2; 95% CI = 1.6-24.3). These results suggest that MetS-DP may differ according to the gut microbial enterotype of each individual. If such associations are found to be causal, personalized nutrition guidelines based on the enterotypes could be recommended to prevent MetS.
Collapse
Affiliation(s)
- Hwan-Hee Jang
- National Institute of Agricultural Sciences, Rural Development Administration (NAS-RDA), Wanju, Jeollabuk-do, Republic of Korea
| | - Hwayoung Noh
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
- Department of Cancer Prevention and Environment, Léon Bérard Cancer Center, L’Institut National de la Santé et de la Recherche Médicale (INSERM) U1296, Lyon, France
| | - Gichang Kim
- National Institute of Agricultural Sciences, Rural Development Administration (NAS-RDA), Wanju, Jeollabuk-do, Republic of Korea
| | - Su-Yeon Cho
- National Institute of Agricultural Sciences, Rural Development Administration (NAS-RDA), Wanju, Jeollabuk-do, Republic of Korea
| | - Hyeon-Jeong Kim
- National Institute of Agricultural Sciences, Rural Development Administration (NAS-RDA), Wanju, Jeollabuk-do, Republic of Korea
| | - Jeong-Sook Choe
- National Institute of Agricultural Sciences, Rural Development Administration (NAS-RDA), Wanju, Jeollabuk-do, Republic of Korea
| | - Jeongseon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Augustin Scalbert
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Oran Kwon
- Graduate Program in System Health Science and Engineering, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Hyesook Kim
- Graduate Program in System Health Science and Engineering, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Leão I, de Carvalho TB, Henriques V, Ferreira C, Sampaio-Maia B, Manaia CM. Pseudomonadota in the oral cavity: a glimpse into the environment-human nexus. Appl Microbiol Biotechnol 2023; 107:517-534. [PMID: 36567346 PMCID: PMC9842593 DOI: 10.1007/s00253-022-12333-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/27/2022]
Abstract
The phylum Pseudomonadota is amongst the most represented in the environment, with a comparatively lower prevalence in the human oral cavity. The ubiquity of Pseudomonadota and the fact that the oral cavity is the most likely entry portal of bacteria from external sources underlie the need to better understand its occurrence in the interface environment-humans. Yet, the relevance oral Pseudomonadota is largely underexplored in the scientific literature, a gap that this review aims at addressing by making, for the first time, an overview of the diversity and ecology of Pseudomonadota in the oral cavity. The screening of scientific literature and human microbiome databases unveiled 1328 reports of Pseudomonadota in the oral cavity. Most of these belonged to the classes Beta- and Gammaproteobacteria, mainly to the families Neisseriaceae, Campylobacteriaceae, and Pasteurelaceae. Others also regularly reported include genera such as Enterobacter, Klebsiella, Acinetobacter, Escherichia, Burkholderia, or Citrobacter, whose members have high potential to acquire virulence and antibiotic resistance genes. This review provides evidence that clinically relevant environmental Pseudomonadota may colonize humans via oral cavity. The need for further investigation about Pseudomonadota at the environment-oral cavity interface and their role as vectors potentially involved in virulence and antibiotic resistance transmission is demonstrated. KEY POINTS: • Neisseriaceae, Campylobacteriaceae, and Pasteurelaceae are part of the core oral microbiome • Enterobacteriaceae, Acinetobacter, or Burkholderia are frequent in the oral microbiome • Gut dysbiosis may be associated with colonization by ubiquitous oral Pseudomonadota.
Collapse
Affiliation(s)
- Inês Leão
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Teresa Bento de Carvalho
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Valentina Henriques
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Catarina Ferreira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Benedita Sampaio-Maia
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| | - Célia M. Manaia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| |
Collapse
|
22
|
Abstract
Trillions of microbes are indigenous to the human gastrointestinal tract, together forming an ecological community known as the gut microbiota. The gut microbiota is involved in dietary digestion to produce various metabolites. In healthy condition, microbial metabolites have unneglectable roles in regulating host physiology and intestinal homeostasis. However, increasing studies have reported the correlation between metabolites and the development of colorectal cancer (CRC), with the identification of oncometabolites. Meanwhile, metabolites can also influence the efficacy of cancer treatments. In this review, metabolites derived from microbes-mediated metabolism of dietary carbohydrates, proteins, and cholesterol, are introduced. The roles of pro-tumorigenic (secondary bile acids and polyamines) and anti-tumorigenic (short-chain fatty acids and indole derivatives) metabolites in CRC development are then discussed. The impacts of metabolites on chemotherapy and immunotherapy are further elucidated. Collectively, given the importance of microbial metabolites in CRC, therapeutic approaches that target metabolites may be promising to improve patient outcome.
Collapse
Affiliation(s)
- Yali Liu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| |
Collapse
|
23
|
Haffner JJ, Katemauswa M, Kagone TS, Hossain E, Jacobson D, Flores K, Parab AR, Obregon-Tito AJ, Tito RY, Reyes LM, Troncoso-Corzo L, Guija-Poma E, Meda N, Carabin H, Honap TP, Sankaranarayanan K, Lewis CM, McCall LI. Untargeted Fecal Metabolomic Analyses across an Industrialization Gradient Reveal Shared Metabolites and Impact of Industrialization on Fecal Microbiome-Metabolome Interactions. mSystems 2022; 7:e0071022. [PMID: 36416540 PMCID: PMC9765122 DOI: 10.1128/msystems.00710-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
The metabolome is a central determinant of human phenotypes and includes the plethora of small molecules produced by host and microbiome or taken up from exogenous sources. However, studies of the metabolome have so far focused predominantly on urban, industrialized populations. Through an untargeted metabolomic analysis of 90 fecal samples from human individuals from Africa and the Americas-the birthplace and the last continental expansion of our species, respectively-we characterized a shared human fecal metabolome. The majority of detected metabolite features were ubiquitous across populations, despite any geographic, dietary, or behavioral differences. Such shared metabolite features included hyocholic acid and cholesterol. However, any characterization of the shared human fecal metabolome is insufficient without exploring the influence of industrialization. Here, we show chemical differences along an industrialization gradient, where the degree of industrialization correlates with metabolomic changes. We identified differential metabolite features such as amino acid-conjugated bile acids and urobilin as major metabolic correlates of these behavioral shifts. Additionally, coanalyses with over 5,000 publicly available human fecal samples and cooccurrence probability analyses with the gut microbiome highlight connections between the human fecal metabolome and gut microbiome. Our results indicate that industrialization significantly influences the human fecal metabolome, but diverse human lifestyles and behavior still maintain a shared human fecal metabolome. This study represents the first characterization of the shared human fecal metabolome through untargeted analyses of populations along an industrialization gradient. IMPORTANCE As the world becomes increasingly industrialized, understanding the biological consequences of these lifestyle shifts and what it means for past, present, and future human health is critical. Indeed, industrialization is associated with rises in allergic and autoimmune health conditions and reduced microbial diversity. Exploring these health effects on a chemical level requires consideration of human lifestyle diversity, but understanding the significance of any differences also requires knowledge of what molecular components are shared between human groups. Our study reveals the key chemistry of the human gut as defined by varied industrialization-based differences and ubiquitous shared features. Ultimately, these novel findings extend our knowledge of human molecular biology, especially as it is influenced by lifestyle and behavior, and provide steps toward understanding how human biology has changed over our species' history.
Collapse
Affiliation(s)
- Jacob J. Haffner
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Mitchelle Katemauswa
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Thérèse S. Kagone
- Burkina Faso Ministry of Health, Ouagadougou, Kadiogo, Burkina Faso
- Centre MURAZ Research Institute, Bobo-Dioulasso, Burkina Faso
| | - Ekram Hossain
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - David Jacobson
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Karina Flores
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Adwaita R. Parab
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Alexandra J. Obregon-Tito
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Raul Y. Tito
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | | | | | - Emilio Guija-Poma
- Centro de Investigación de Bioquímica y Nutrición, Facultad de Medicina Humana, Universidad de San Martín de Porres, Lima, Perú
| | - Nicolas Meda
- Burkina Faso Ministry of Health, Ouagadougou, Kadiogo, Burkina Faso
| | - Hélène Carabin
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
- Département de Médecine Sociale et Préventive, École de Santé Publique de l’Université de Montréal, Montréal, Quebec, Canada
- Centre de Recherche en Santé Publique (CReSP) de l’Université de Montréal et du CIUSS du Centre Sud de Montréal, Montréal, Quebec, Canada
| | - Tanvi P. Honap
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Krithivasan Sankaranarayanan
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Cecil M. Lewis
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
24
|
Lee BY, Ordovás JM, Parks EJ, Anderson CAM, Barabási AL, Clinton SK, de la Haye K, Duffy VB, Franks PW, Ginexi EM, Hammond KJ, Hanlon EC, Hittle M, Ho E, Horn AL, Isaacson RS, Mabry PL, Malone S, Martin CK, Mattei J, Meydani SN, Nelson LM, Neuhouser ML, Parent B, Pronk NP, Roche HM, Saria S, Scheer FAJL, Segal E, Sevick MA, Spector TD, Van Horn L, Varady KA, Voruganti VS, Martinez MF. Research gaps and opportunities in precision nutrition: an NIH workshop report. Am J Clin Nutr 2022; 116:1877-1900. [PMID: 36055772 PMCID: PMC9761773 DOI: 10.1093/ajcn/nqac237] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/06/2022] [Accepted: 08/30/2022] [Indexed: 02/01/2023] Open
Abstract
Precision nutrition is an emerging concept that aims to develop nutrition recommendations tailored to different people's circumstances and biological characteristics. Responses to dietary change and the resulting health outcomes from consuming different diets may vary significantly between people based on interactions between their genetic backgrounds, physiology, microbiome, underlying health status, behaviors, social influences, and environmental exposures. On 11-12 January 2021, the National Institutes of Health convened a workshop entitled "Precision Nutrition: Research Gaps and Opportunities" to bring together experts to discuss the issues involved in better understanding and addressing precision nutrition. The workshop proceeded in 3 parts: part I covered many aspects of genetics and physiology that mediate the links between nutrient intake and health conditions such as cardiovascular disease, Alzheimer disease, and cancer; part II reviewed potential contributors to interindividual variability in dietary exposures and responses such as baseline nutritional status, circadian rhythm/sleep, environmental exposures, sensory properties of food, stress, inflammation, and the social determinants of health; part III presented the need for systems approaches, with new methods and technologies that can facilitate the study and implementation of precision nutrition, and workforce development needed to create a new generation of researchers. The workshop concluded that much research will be needed before more precise nutrition recommendations can be achieved. This includes better understanding and accounting for variables such as age, sex, ethnicity, medical history, genetics, and social and environmental factors. The advent of new methods and technologies and the availability of considerably more data bring tremendous opportunity. However, the field must proceed with appropriate levels of caution and make sure the factors listed above are all considered, and systems approaches and methods are incorporated. It will be important to develop and train an expanded workforce with the goal of reducing health disparities and improving precision nutritional advice for all Americans.
Collapse
Affiliation(s)
- Bruce Y Lee
- Health Policy and Management, City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA
| | - José M Ordovás
- USDA-Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Elizabeth J Parks
- Nutrition and Exercise Physiology, University of Missouri School of Medicine, MO, USA
| | | | - Albert-László Barabási
- Network Science Institute and Department of Physics, Northeastern University, Boston, MA, USA
| | | | - Kayla de la Haye
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Valerie B Duffy
- Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Paul W Franks
- Novo Nordisk Foundation, Hellerup, Denmark, Copenhagen, Denmark, and Lund University Diabetes Center, Sweden
- The Lund University Diabetes Center, Malmo, SwedenInsert Affiliation Text Here
| | - Elizabeth M Ginexi
- National Institutes of Health, Office of Behavioral and Social Sciences Research, Bethesda, MD, USA
| | - Kristian J Hammond
- Computer Science, Northwestern University McCormick School of Engineering, IL, USA
| | - Erin C Hanlon
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Michael Hittle
- Epidemiology and Clinical Research, Stanford University, Stanford, CA, USA
| | - Emily Ho
- Public Health and Human Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Abigail L Horn
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | | | | | - Susan Malone
- Rory Meyers College of Nursing, New York University, New York, NY, USA
| | - Corby K Martin
- Ingestive Behavior Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Josiemer Mattei
- Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Simin Nikbin Meydani
- USDA-Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Lorene M Nelson
- Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | | | - Brendan Parent
- Grossman School of Medicine, New York University, New York, NY, USA
| | | | - Helen M Roche
- UCD Conway Institute, School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Suchi Saria
- Johns Hopkins University, Baltimore, MD, USA
| | - Frank A J L Scheer
- Brigham and Women's Hospital, Boston, MA, USA
- Medicine and Neurology, Harvard Medical School, Boston, MA, USA
| | - Eran Segal
- Computer Science and Applied Math, Weizmann Institute of Science, Rehovot, Israel
| | - Mary Ann Sevick
- Grossman School of Medicine, New York University, New York, NY, USA
| | - Tim D Spector
- Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Linda Van Horn
- Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Krista A Varady
- Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Venkata Saroja Voruganti
- Nutrition and Nutrition Research Institute, Gillings School of Public Health, The University of North Carolina, Chapel Hill, NC, USA
| | - Marie F Martinez
- Health Policy and Management, City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA
| |
Collapse
|
25
|
Maki KA, Ganesan SM, Meeks B, Farmer N, Kazmi N, Barb JJ, Joseph PV, Wallen GR. The role of the oral microbiome in smoking-related cardiovascular risk: a review of the literature exploring mechanisms and pathways. J Transl Med 2022; 20:584. [PMID: 36503487 PMCID: PMC9743777 DOI: 10.1186/s12967-022-03785-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. Oral health is associated with smoking and cardiovascular outcomes, but there are gaps in knowledge of many mechanisms connecting smoking to cardiovascular risk. Therefore, the aim of this review is to synthesize literature on smoking and the oral microbiome, and smoking and cardiovascular risk/disease, respectively. A secondary aim is to identify common associations between the oral microbiome and cardiovascular risk/disease to smoking, respectively, to identify potential shared oral microbiome-associated mechanisms. We identified several oral bacteria across varying studies that were associated with smoking. Atopobium, Gemella, Megasphaera, Mycoplasma, Porphyromonas, Prevotella, Rothia, Treponema, and Veillonella were increased, while Bergeyella, Haemophilus, Lautropia, and Neisseria were decreased in the oral microbiome of smokers versus non-smokers. Several bacteria that were increased in the oral microbiome of smokers were also positively associated with cardiovascular outcomes including Porphyromonas, Prevotella, Treponema, and Veillonella. We review possible mechanisms that may link the oral microbiome to smoking and cardiovascular risk including inflammation, modulation of amino acids and lipids, and nitric oxide modulation. Our hope is this review will inform future research targeting the microbiome and smoking-related cardiovascular disease so possible microbial targets for cardiovascular risk reduction can be identified.
Collapse
Affiliation(s)
- Katherine A. Maki
- grid.410305.30000 0001 2194 5650Translational Biobehavioral and Health Disparities Branch, National Institutes of Health, Clinical Center, 10 Center Drive, Building 10, Bethesda, MD 20814 USA
| | - Sukirth M. Ganesan
- grid.214572.70000 0004 1936 8294Department of Periodontics, The University of Iowa College of Dentistry and Dental Clinics, 801 Newton Rd., Iowa City, IA 52242 USA
| | - Brianna Meeks
- grid.411024.20000 0001 2175 4264University of Maryland, School of Social Work, Baltimore, MD USA
| | - Nicole Farmer
- grid.410305.30000 0001 2194 5650Translational Biobehavioral and Health Disparities Branch, National Institutes of Health, Clinical Center, 10 Center Drive, Building 10, Bethesda, MD 20814 USA
| | - Narjis Kazmi
- grid.410305.30000 0001 2194 5650Translational Biobehavioral and Health Disparities Branch, National Institutes of Health, Clinical Center, 10 Center Drive, Building 10, Bethesda, MD 20814 USA
| | - Jennifer J. Barb
- grid.410305.30000 0001 2194 5650Translational Biobehavioral and Health Disparities Branch, National Institutes of Health, Clinical Center, 10 Center Drive, Building 10, Bethesda, MD 20814 USA
| | - Paule V. Joseph
- grid.420085.b0000 0004 0481 4802National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD USA ,grid.280738.60000 0001 0035 9863National Institute of Nursing Research, National Institutes of Health, Bethesda, MD USA
| | - Gwenyth R. Wallen
- grid.410305.30000 0001 2194 5650Translational Biobehavioral and Health Disparities Branch, National Institutes of Health, Clinical Center, 10 Center Drive, Building 10, Bethesda, MD 20814 USA
| |
Collapse
|
26
|
Carson TL, Buro AW, Miller D, Peña A, Ard JD, Lampe JW, Yi N, Lefkowitz E, William VDP, Morrow C, Wilson L, Barnes S, Demark-Wahnefried W. Rationale and study protocol for a randomized controlled feeding study to determine the structural- and functional-level effects of diet-specific interventions on the gut microbiota of non-Hispanic black and white adults. Contemp Clin Trials 2022; 123:106968. [PMID: 36265810 PMCID: PMC10095329 DOI: 10.1016/j.cct.2022.106968] [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: 04/27/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Colorectal cancer (CRC), the third leading cause of cancer-related deaths in the US, has been associated with an overrepresentation or paucity of several microbial taxa in the gut microbiota, but causality has not been established. Black men and women have among the highest CRC incidence and mortality rates of any racial/ethnic group. This study will examine the impact of the Dietary Approaches to Stop Hypertension (DASH) diet on gut microbiota and fecal metabolites associated with CRC risk. METHODS A generally healthy sample of non-Hispanic Black and white adults (n = 112) is being recruited to participate in a parallel-arm randomized controlled feeding study. Participants are randomized to receive the DASH diet or a standard American diet for a 28-day period. Fecal samples are collected weekly throughout the study to analyze changes in the gut microbiota using 16 s rRNA and selected metagenomics. Differences in bacterial alpha and beta diversity and taxa that have been associated with CRC (Bacteroides, Fusobacterium, Clostridium, Lactobacillus, Bifidobacterium, Ruminococcus, Porphyromonas, Succinivibrio) are being evaluated. Covariate measures include body mass index, comorbidities, medication history, physical activity, stress, and demographic characteristics. CONCLUSION Our findings will provide preliminary evidence for the DASH diet as an approach for cultivating a healthier gut microbiota across non-Hispanic Black and non-Hispanic White adults. These results can impact clinical, translational, and population-level approaches for modification of the gut microbiota to reduce risk of chronic diseases including CRC. TRIAL REGISTRATION This study was registered on ClinicalTrials.gov, identifier NCT04538482, on September 4, 2020 (https://clinicaltrials.gov/ct2/show/NCT04538482).
Collapse
Affiliation(s)
- Tiffany L Carson
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States of America.
| | - Acadia W Buro
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States of America
| | - Darci Miller
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States of America
| | - Alissa Peña
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States of America
| | - Jamy D Ard
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Johanna W Lampe
- Public Health Science Division, Fred Hutchinson Cancer Center, Seattle, WA, United States of America
| | - Nengjun Yi
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Elliot Lefkowitz
- Center for Clinical and Translational Sciences, University of Alabama at Birmingham, Birmingham, AL, United States of America; Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Van Der Pol William
- Center for Clinical and Translational Sciences, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Casey Morrow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Landon Wilson
- Department of Pharmacology and Toxicology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America; Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America; Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Wendy Demark-Wahnefried
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| |
Collapse
|
27
|
Hammond TC, Powell E, Green SJ, Chlipala G, Frank J, Yackzan AT, Yanckello LM, Chang YH, Xing X, Heil S, Springer JE, Pennypacker K, Stromberg A, Sawaki L, Lin AL. Functional recovery outcomes following acute stroke is associated with abundance of gut microbiota related to inflammation, butyrate and secondary bile acid. FRONTIERS IN REHABILITATION SCIENCES 2022; 3:1017180. [PMID: 36386777 PMCID: PMC9644110 DOI: 10.3389/fresc.2022.1017180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022]
Abstract
Accumulating evidence suggests that gut microbes modulate brain plasticity via the bidirectional gut-brain axis and play a role in stroke rehabilitation. However, the microbial species alterations associated with stroke and their correlation with functional outcome measures following acute stroke remain unknown. Here we measure post-stroke gut dysbiosis and how it correlates with gut permeability and cognitive functions in 12 stroke participants, 18 controls with risk factors for stroke, and 12 controls without risk factors. Stool samples were used to measure the microbiome with whole genome shotgun sequencing and leaky gut markers. We genotyped APOE status and measured diet composition and motor, cognitive, and emotional status using NIH Toolbox. We used linear regression methods to identify gut microbial associations with cognitive and emotional assessments. We did not find significance differences between the two control groups. In contrast, the bacteria populations of the Stroke group were statistically dissimilar from the control groups. Relative abundance analysis revealed notable decreases in butyrate-producing microbial taxa, secondary bile acid-producing taxa, and equol-producing taxa. The Stroke group had higher levels of the leaky gut marker alpha-1-antitrypsin in the stool than either of the groups and several taxa including Roseburia species (a butyrate producer) were negatively correlated with alpha-1-antitrypsin. Stroke participants scored lower on memory testing than those in the two control groups. Stroke participants with more Roseburia performed better on the picture vocabulary task; more Bacteroides uniformis (a butyrate producer) and less Escherichia coli (a pro-inflammatory species) reported higher levels of self-efficacy. Intakes of fiber, fruit and vegetable were lower, but sweetened beverages were higher, in the Stroke group compared with controls. Vegetable consumption was correlated with many bacterial changes among the participants, but only the species Clostridium bolteae, a pro-inflammatory species, was significantly associated with stroke. Our findings indicate that stroke is associated with a higher abundance of proinflammatory species and a lower abundance of butyrate producers and secondary bile acid producers. These altered microbial communities are associated with poorer functional performances. Future studies targeting the gut microbiome should be developed to elucidate whether its manipulation could optimize rehabilitation and boost recovery.
Collapse
Affiliation(s)
- Tyler C. Hammond
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Elizabeth Powell
- Department of Physical Medicine and Rehabilitation, University of Kentucky, Lexington, KY, United States
| | - Stefan J. Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, IL, United States
| | - George Chlipala
- Research Informatics Core, University of Illinois Chicago, Chicago, IL, United States
| | - Jacqueline Frank
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Center for Advanced Stroke Science, Department of Neurology, University of Kentucky, Lexington, KY, United States
| | - Andrew T. Yackzan
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Lucille M. Yanckello
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Ya-Hsuan Chang
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Xin Xing
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Computer Science, University of Kentucky, Lexington, KY, United States
| | - Sally Heil
- School of Medicine, University of Missouri, Columbia, MO, United States
| | - Joe E. Springer
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Keith Pennypacker
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Center for Advanced Stroke Science, Department of Neurology, University of Kentucky, Lexington, KY, United States
| | - Arnold Stromberg
- Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Lumy Sawaki
- Department of Physical Medicine and Rehabilitation, University of Kentucky, Lexington, KY, United States
| | - Ai-Ling Lin
- Department of Radiology, University of Missouri, Columbia, MO, United States
- Institute for Data Science & Informatics, University of Missouri, Columbia, MOUnited States
- Department of Biological Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
28
|
Bykova M, Hou Y, Eng C, Cheng F. Quantitative trait locus (xQTL) approaches identify risk genes and drug targets from human non-coding genomes. Hum Mol Genet 2022; 31:R105-R113. [PMID: 36018824 PMCID: PMC9989738 DOI: 10.1093/hmg/ddac208] [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: 07/26/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Advances and reduction of costs in various sequencing technologies allow for a closer look at variations present in the non-coding regions of the human genome. Correlating non-coding variants with large-scale multi-omic data holds the promise not only of a better understanding of likely causal connections between non-coding DNA and expression of traits but also identifying potential disease-modifying medicines. Genome-phenome association studies have created large datasets of DNA variants that are associated with multiple traits or diseases, such as Alzheimer's disease; yet, the functional consequences of variants, in particular of non-coding variants, remain largely unknown. Recent advances in functional genomics and computational approaches have led to the identification of potential roles of DNA variants, such as various quantitative trait locus (xQTL) techniques. Multi-omic assays and analytic approaches toward xQTL have identified links between genetic loci and human transcriptomic, epigenomic, proteomic and metabolomic data. In this review, we first discuss the recent development of xQTL from multi-omic findings. We then highlight multimodal analysis of xQTL and genetic data for identification of risk genes and drug targets using Alzheimer's disease as an example. We finally discuss challenges and future research directions (e.g. artificial intelligence) for annotation of non-coding variants in complex diseases.
Collapse
Affiliation(s)
- Marina Bykova
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|
29
|
Chatelaine HA, Ramazani CA, Spencer K, Olivo‐Marston S, Bailey MT, McElroy J, Hatzakis E, Mathé EA, Kopec RE. Dietary Energy Intake and Presence of Aberrant Crypt Foci Are Associated with Phospholipid, Purine, and Taurine Metabolite Abundances in C57BL/6N Mouse Colon. Mol Nutr Food Res 2022; 66:e2200180. [PMID: 35969485 PMCID: PMC9787839 DOI: 10.1002/mnfr.202200180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/12/2022] [Indexed: 12/31/2022]
Abstract
SCOPE Colon metabolomes associated with high-fat (H) versus energy-restricted (E) diets in early colorectal cancer (CRC) models have never been directly compared. The objectives of this study are to elucidate metabolites associated with diet, aberrant crypt foci (ACF), and diet:ACF interaction, using a lifetime murine model. METHODS AND RESULTS Three-week-old mice consumed control (C), E, or H initiation diets for 18 weeks. ACF formation is initiated weeks 16-21 with azoxymethane injections, followed by progression diet crossover (to C, E, or H) through week 60. Colon extracts are analyzed using ultra-high-performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). Metabolites associated with diet, ACF, or diet:ACF are determined using regression models (FDR-adjusted p-value <0.05). No metabolites are significantly associated with initiation diets, but concentrations of acylcarnitines and phospholipids are associated with C, E, and H progression diets. Purines, taurine, and phospholipids are associated with ACF presence. No significant associations between metabolites and diet:ACF interaction are observed. CONCLUSIONS These results suggest that recent, rather than early-life, diet is more closely associated with the colon metabolome, particularly lipid metabolism. Results from this study also provide candidate biomarkers of early CRC development and provide support for the importance of early diet on influencing pre-CRC risk.
Collapse
Affiliation(s)
- Haley A. Chatelaine
- OSU Interdisciplinary Nutrition PhD Program (OSUN)Department of Human SciencesThe Ohio State University1787 Neil AveColumbusOH43210United States
- Division of Preclinical Innovation Informatics CoreNational Center for Advancing Translational Sciences9800 Medical Center DriveRockvilleMD20850USA
| | - Cynthia A. Ramazani
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Big Data for Indiana State UniversityIndiana State UniversityTerre HauteIN47807USA
| | - Kyle Spencer
- Division of Preclinical Innovation Informatics CoreNational Center for Advancing Translational Sciences9800 Medical Center DriveRockvilleMD20850USA
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Nationwide Children's HospitalColumbusOH43205USA
| | - Susan Olivo‐Marston
- Division of EpidemiologyCollege of Public HealthThe Ohio State UniversityColumbusOH43210USA
- Southern Illinois University School of MedicineSpringfieldIL62794USA
| | - Michael T. Bailey
- Nationwide Children's HospitalColumbusOH43205USA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOH43210USA
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOH43205USA
- Oral and GI Research Affinity GroupNationwide Children's HospitalColumbusOH43205USA
| | - Joseph McElroy
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
| | - Emmanuel Hatzakis
- Department of Food Science and TechnologyThe Ohio State UniversityColumbusOH43210USA
- Foods for Health Discovery ThemeThe Ohio State UniversityColumbusOH43210USA
| | - Ewy A. Mathé
- Division of Preclinical Innovation Informatics CoreNational Center for Advancing Translational Sciences9800 Medical Center DriveRockvilleMD20850USA
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOH43210USA
- Translational Data Analytics InstituteThe Ohio State UniversityColumbusOH43210USA
| | - Rachel E. Kopec
- OSU Interdisciplinary Nutrition PhD Program (OSUN)Department of Human SciencesThe Ohio State University1787 Neil AveColumbusOH43210United States
- Foods for Health Discovery ThemeThe Ohio State UniversityColumbusOH43210USA
| |
Collapse
|
30
|
Cox IJ, Peña Rodríguez M, Fagan A, Rojas-Lara MV, Le Guennec A, Rodriguez-Alvarez F, McGeorge S, Escalona-Nandez I, Torre A, Bajaj JS. Stool microbiota show greater linkages with plasma metabolites compared to salivary microbiota in a multinational cirrhosis cohort. Liver Int 2022; 42:2274-2282. [PMID: 35635305 DOI: 10.1111/liv.15329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/03/2022] [Accepted: 05/27/2022] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Cirrhosis is associated with changes in gut microbiota in both saliva and stool. The relative linkage patterns of stool versus saliva microbiota with systemic metabolomics are unclear and may differ across countries. We hypothesized that stool microbiota have greater linkages with plasma metabolites than saliva microbiota, which may depend on country of origin. METHODS Age-balanced controls and outpatient patients with cirrhosis, compensated and decompensated, from the USA and Mexico (MX) underwent plasma collection and dietary recall. Plasma metabolomics were analysed using nuclear magnetic resonance spectroscopy. Microbiota in stool and saliva samples were analysed using 16S rRNA analyses. Correlation network differences between both saliva and stool gut microbiota and plasma metabolites were compared between subject groupings and within/between countries. RESULTS A total of 313 age-balanced subjects-135 USA (47 control, 48 compensated and 40 decompensated) and 178 MX (71 control, 56 compensated and 51 decompensated)-were enrolled. Cirrhosis severity, including lactulose and rifaximin use, were comparable. Plasma metabolites differed with advancing cirrhosis, between countries and according to 90-day hospitalizations. Correlation networks demonstrated more microbiome-metabolite linkages in stool compared to saliva in both populations, although there were no salivary correlation metrics across decompensated subjects in either country. Stool Lactobacillus showed a positive correlation to plasma lactate in decompensated cirrhosis from MX but not USA. CONCLUSIONS Stool microbiota were more extensively linked with systemic metabolites than were saliva microbiota, irrespective of cirrhosis severity and country. These changes were more prominent in decompensated cirrhosis and were centred around plasma lactate, which might reflect the interaction of diet and lactulose therapy.
Collapse
Affiliation(s)
- Isobel Jane Cox
- The Roger Williams Institute of Hepatology London, Foundation for Liver Research, London, UK.,Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Marcela Peña Rodríguez
- Laboratory for the Diagnosis of Emerging and Reemerging Diseases (LaDEER), University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Andrew Fagan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, Virginia, USA
| | - Mayra V Rojas-Lara
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Adrien Le Guennec
- Randall Centre for Cell & Molecular Biophysics and Centre for Biomolecular Spectroscopy, King's College London, London, UK
| | | | - Sara McGeorge
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, Virginia, USA
| | - Ivonne Escalona-Nandez
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Aldo Torre
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, Virginia, USA
| |
Collapse
|
31
|
Ramos-Lopez O, Martinez JA, Milagro FI. Holistic Integration of Omics Tools for Precision Nutrition in Health and Disease. Nutrients 2022; 14:nu14194074. [PMID: 36235725 PMCID: PMC9572439 DOI: 10.3390/nu14194074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
The combination of multiple omics approaches has emerged as an innovative holistic scope to provide a more comprehensive view of the molecular and physiological events underlying human diseases (including obesity, dyslipidemias, fatty liver, insulin resistance, and inflammation), as well as for elucidating unique and specific metabolic phenotypes. These omics technologies include genomics (polymorphisms and other structural genetic variants), epigenomics (DNA methylation, histone modifications, long non-coding RNA, telomere length), metagenomics (gut microbiota composition, enterotypes), transcriptomics (RNA expression patterns), proteomics (protein quantities), and metabolomics (metabolite profiles), as well as interactions with dietary/nutritional factors. Although more evidence is still necessary, it is expected that the incorporation of integrative omics could be useful not only for risk prediction and early diagnosis but also for guiding tailored dietary treatments and prognosis schemes. Some challenges include ethical and regulatory issues, the lack of robust and reproducible results due to methodological aspects, the high cost of omics methodologies, and high-dimensional data analyses and interpretation. In this review, we provide examples of system biology studies using multi-omics methodologies to unravel novel insights into the mechanisms and pathways connecting the genotype to clinically relevant traits and therapy outcomes for precision nutrition applications in health and disease.
Collapse
Affiliation(s)
- Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
- Correspondence:
| | - J. Alfredo Martinez
- Precision Nutrition and Cardiometabolic Health, IMDEA Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain
| | - Fermin I. Milagro
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain
- Center for Nutrition Research, University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
32
|
Hasavci D, Blank T. Age-dependent effects of gut microbiota metabolites on brain resident macrophages. Front Cell Neurosci 2022; 16:944526. [PMID: 36072564 PMCID: PMC9441744 DOI: 10.3389/fncel.2022.944526] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, development of age-related diseases, such as Alzheimer's and Parkinson's disease, as well as other brain disorders, including anxiety, depression, and schizophrenia have been shown to be associated with changes in the gut microbiome. Several factors can induce an alteration in the bacterial composition of the host's gastrointestinal tract. Besides dietary changes and frequent use of antibiotics, the microbiome is also profoundly affected by aging. Levels of microbiota-derived metabolites are elevated in older individuals with age-associated diseases and cognitive defects compared to younger, healthy age groups. The identified metabolites with higher concentration in aged hosts, which include choline and trimethylamine, are known risk factors for age-related diseases. While the underlying mechanisms and pathways remain elusive for the most part, it has been shown, that these metabolites are able to trigger the innate immunity in the central nervous system by influencing development and activation status of brain-resident macrophages. The macrophages residing in the brain comprise parenchymal microglia and non-parenchymal macrophages located in the perivascular spaces, meninges, and the choroid plexus. In this review, we highlight the impact of age on the composition of the microbiome and microbiota-derived metabolites and their influence on age-associated diseases caused by dysfunctional brain-resident macrophages.
Collapse
Affiliation(s)
| | - Thomas Blank
- Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Saliva microbiome, dietary, and genetic markers are associated with suicidal ideation in university students. Sci Rep 2022; 12:14306. [PMID: 35995968 PMCID: PMC9395396 DOI: 10.1038/s41598-022-18020-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 08/03/2022] [Indexed: 11/09/2022] Open
Abstract
Here, salivary microbiota and major histocompatibility complex (MHC) human leukocyte antigen (HLA) alleles were compared between 47 (12.6%) young adults with recent suicidal ideation (SI) and 325 (87.4%) controls without recent SI. Several bacterial taxa were correlated with SI after controlling for sleep issues, diet, and genetics. Four MHC class II alleles were protective for SI including DRB1*04, which was absent in every subject with SI while present in 21.7% of controls. Increased incidence of SI was observed with four other MHC class II alleles and two MHC class I alleles. Associations between these HLA alleles and salivary bacteria were also identified. Furthermore, rs10437629, previously associated with attempted suicide, was correlated here with SI and the absence of Alloprevotella rava, a producer of an organic acid known to promote brain energy homeostasis. Hence, microbial-genetic associations may be important players in the diathesis-stress model for suicidal behaviors.
Collapse
|
34
|
Yang L, Dai Y, He H, Liu Z, Liao S, Zhang Y, Liao G, An Z. Integrative analysis of gut microbiota and fecal metabolites in metabolic associated fatty liver disease patients. Front Microbiol 2022; 13:969757. [PMID: 36071958 PMCID: PMC9441872 DOI: 10.3389/fmicb.2022.969757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveMetabolic associated fatty liver disease (MAFLD) affects nearly a quarter of the world’s population. Our study aimed to characterize the gut microbiome and overall changes in the fecal and serum metabolomes in MAFLD patients.MethodsThirty-two patients diagnosed with MAFLD and 30 healthy individuals (control group, CG) were included in this study, the basic clinical characteristics and laboratory test results including routine biochemistry, etc. were recorded for all, and their serum and fecal samples were collected. A portion of the fecal samples was subjected to 16S rDNA sequencing, and the other portion of the fecal samples and serum samples were subjected to non-targeted metabolomic detection based on liquid chromatography-mass spectrometry (LC–MS). Statistical analysis of clinical data was performed using SPSS software package version 25.0 (SPSS Inc., Chicago, IL, United States). The analysis of 16S rDNA sequencing results was mainly performed by R software (V. 2.15.3), and the metabolomics data analysis was mainly performed by CD 3.1 software. Two-tailed p value < 0.05 was considered statistically significant.ResultsThe 16S sequencing data suggested that the species richness and diversity of MAFLD patients were reduced compared with controls. At the phylum level, the relative abundance of Bacteroidota, Pseudomonadota, and Fusobacteriota increased and Bacillota decreased in MAFLD patients. At the genus level, the relative abundances of Prevotella, Bacteroides, Escherichia-Shigella, etc. increased. 2,770 metabolites were detected in stool samples and 1,245 metabolites were detected in serum samples. The proportion of differential lipid metabolites in serum (49%) was higher than that in feces (21%). There were 22 differential metabolites shared in feces and serum. And the association analysis indicated that LPC 18:0 was positively correlated with Christensenellaceae_R-7_group, Oscillospiraceae_UCG-002; neohesperidin was also positively correlated with Peptoniphilus, Phycicoccus, and Stomatobaculum.ConclusionMicrobial sequencing data suggested decreased species richness and diversity and altered β-diversity in feces. Metabolomic analysis identified overall changes in fecal and serum metabolites dominated by lipid molecules. And the association analysis with gut microbes provided potentially pivotal gut microbiota-metabolite combinations in MAFLD patients, which might provide new clues for further research on the disease mechanism and the development of new diagnostic markers and treatments.
Collapse
Affiliation(s)
- Lidan Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yuzhao Dai
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - He He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shenling Liao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Zhang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Information Management, Department of Stomatology Informatics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Ga Liao,
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Zhenmei An,
| |
Collapse
|
35
|
Long-Term Mastication Changed Salivary Metabolomic Profiles. Metabolites 2022; 12:metabo12070660. [PMID: 35888784 PMCID: PMC9322701 DOI: 10.3390/metabo12070660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 11/29/2022] Open
Abstract
Saliva is an ideal biofluid for monitoring oral and systemic health. Repeated mastication is a typical physical stimulus that improves salivary flow and oral hygiene. Recent metabolomic studies have shown the potential of salivary metabolomic components for various disease monitoring systems. Here, we evaluated the effect of long-term mastication on salivary metabolomic profiles. Young women with good oral hygiene (20.8 ± 0.3 years, n = 17) participated. They were prohibited from chewing gum during control periods (4 weeks each) and were instructed to chew a piece of gum base seven times a day for 10 min each time during the intervention period. Paired samples of unstimulated whole saliva collected on the last day of the control and intervention period were compared. Liquid chromatography−time-of-flight mass spectrometry successfully quantified 85 metabolites, of which 41 showed significant differences (p < 0.05, Wilcoxon paired test corrected by false discovery rate). Except for a few metabolites, such as citrate, most metabolites showed lower concentrations after the intervention. The pathways related to glycogenic amino acids, such as alanine, arginine, and glutamine, altered considerably. This study suggests that long-term mastication induces unstimulated salivary component-level changes.
Collapse
|
36
|
Ke A, Parreira VR, Farber JM, Goodridge L. Inhibition of Cronobacter sakazakii in an infant simulator of the human intestinal microbial ecosystem using a potential synbiotic. Front Microbiol 2022; 13:947624. [PMID: 35910651 PMCID: PMC9335077 DOI: 10.3389/fmicb.2022.947624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/27/2022] [Indexed: 12/22/2022] Open
Abstract
Powdered infant formula (PIF) can be contaminated with Cronobacter sakazakii, which can cause severe illnesses in infants. Synbiotics, a combination of probiotics and prebiotics, could act as an alternative control measure for C. sakazakii contamination in PIF and within the infant gut, but synbiotics have not been well studied for their ability to inhibit C. sakazakii. Using a Simulator of the Human Intestinal Microbial Ecosystem (SHIME®) inoculated with infant fecal matter, we demonstrated that a potential synbiotic, consisting of six lactic acid bacteria (LAB) strains and Vivinal GOS, can inhibit the growth of C. sakazakii in an infant possibly through either the production of antimicrobial metabolites like acetate, increasing species diversity within the SHIME compartments to compete for nutrients or a combination of mechanisms. Using a triple SHIME set-up, i.e., three identical SHIME compartments, the first SHIME (SHIME 1) was designated as the control SHIME in the absence of a treatment, whereas SHIME 2 and 3 were the treated SHIME over 2, 1-week treatment periods. The addition of the potential synbiotic (LAB + VGOS) resulted in a significant decrease in C. sakazakii levels within 1 week (p < 0.05), but in the absence of a treatment the significant decline took 2 weeks (p < 0.05), and the LAB treatment did not decrease C. sakazakii levels (p ≥ 0.05). The principal component analysis showed a distinction between metabolomic profiles for the control and LAB treatment, but similar profiles for the LAB + VGOS treatment. The addition of the potential synbiotic (LAB + VGOS) in the first treatment period slightly increased species diversity (p ≥ 0.05) compared to the control and LAB, which may have had an effect on the survival of C. sakazakii throughout the treatment period. Our results also revealed that the relative abundance of Bifidobacterium was negatively correlated with Cronobacter when no treatments were added (ρ = −0.96; p < 0.05). These findings suggest that C. sakazakii could be inhibited by the native gut microbiota, and inhibition can be accelerated by the potential synbiotic treatment.
Collapse
|
37
|
Lee JY, Hwang HW, Jin HS, Lee JE, Kang NJ, Lee DW. A Genomics-Based Semirational Approach for Expanding the Postbiotic Potential of Collagen Peptides Using Lactobacillaceae. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:8365-8376. [PMID: 35758868 DOI: 10.1021/acs.jafc.2c01251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Food-derived bioactive peptides (BPs) have received considerable attention as postbiotics for human gut health. Here we used a genomics-based semirational approach to expand the postbiotic potential of collagen peptides (CPs) produced from probiotic fermentation. In silico digestion revealed distinct BPs embedded in fish collagen in a protease-dependent manner. Anaerobic digestion of collagen by representative Lactobacillaceae species revealed differential substrate utilization and collagen degradation patterns. Nanoliquid chromatography-mass spectrometry analysis of CPs showed that each species exhibited different cleavage patterns and unique peptide profiles. Remarkably, the 1-10 kDa CPs produced by Lacticaseibacillus paracasei showed agonistic activities toward G protein-coupled receptor 35 (GPR35). These CPs could repair intestinal epithelium through the GPR35-mediated extracellular signal-regulated protein kinase (ERK) 1/2 signaling pathway, suggesting that probiotic-aided collagen hydrolysates can serve as postbiotics for host-microbe interactions. Therefore, this study provides an effective strategy for the rapid screening of CPs for gut health in the gastrointestinal tract.
Collapse
Affiliation(s)
- Ji-Young Lee
- Department of Biotechnology, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, South Korea
| | - Hye Won Hwang
- Department of Bioindustrial Engineering, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, South Korea
| | - Hyeon-Su Jin
- Department of Biotechnology, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, South Korea
| | - Jae-Eun Lee
- Department of Biotechnology, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, South Korea
| | - Nam Joo Kang
- School of Food Science and Biotechnology, Kyungpook National University, Daehak-ro 80, Buk-gu, Daegu 41566, South Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, South Korea
- Department of Bioindustrial Engineering, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, South Korea
| |
Collapse
|
38
|
Shute A, Bihan DG, Lewis IA, Nasser Y. Metabolomics: The Key to Unraveling the Role of the Microbiome in Visceral Pain Neurotransmission. Front Neurosci 2022; 16:917197. [PMID: 35812241 PMCID: PMC9260117 DOI: 10.3389/fnins.2022.917197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022] Open
Abstract
Inflammatory bowel disease (IBD), comprising Crohn's disease and Ulcerative colitis, is a relapsing and remitting disease of the gastrointestinal tract, presenting with chronic inflammation, ulceration, gastrointestinal bleeding, and abdominal pain. Up to 80% of patients suffering from IBD experience acute pain, which dissipates when the underlying inflammation and tissue damage resolves. However, despite achieving endoscopic remission with no signs of ongoing intestinal inflammation or damage, 30-50% of IBD patients in remission experience chronic abdominal pain, suggesting altered sensory neuronal processing in this disorder. Furthermore, effective treatment for chronic pain is limited such that 5-25% of IBD outpatients are treated with narcotics, with associated morbidity and mortality. IBD patients commonly present with substantial alterations to the microbial community structure within the gastrointestinal tract, known as dysbiosis. The same is also true in irritable bowel syndrome (IBS), a chronic disorder characterized by altered bowel habits and abdominal pain, in the absence of inflammation. An emerging body of literature suggests that the gut microbiome plays an important role in visceral hypersensitivity. Specific microbial metabolites have an intimate relationship with host receptors that are highly expressed on host cell and neurons, suggesting that microbial metabolites play a key role in visceral hypersensitivity. In this review, we will discuss the techniques used to analysis the metabolome, current potential metabolite targets for visceral hypersensitivity, and discuss the current literature that evaluates the role of the post-inflammatory microbiota and metabolites in visceral hypersensitivity.
Collapse
Affiliation(s)
- Adam Shute
- Department of Medicine, Cumming School of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Dominique G. Bihan
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Ian A. Lewis
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Yasmin Nasser
- Department of Medicine, Cumming School of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
39
|
Boudar Z, Sehli S, El Janahi S, Al Idrissi N, Hamdi S, Dini N, Brim H, Amzazi S, Nejjari C, Lloyd-Puryear M, Ghazal H. Metagenomics Approaches to Investigate the Neonatal Gut Microbiome. Front Pediatr 2022; 10:886627. [PMID: 35799697 PMCID: PMC9253679 DOI: 10.3389/fped.2022.886627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
Early infancy is critical for the development of an infant's gut flora. Many factors can influence microbiota development during the pre- and postnatal periods, including maternal factors, antibiotic exposure, mode of delivery, dietary patterns, and feeding type. Therefore, investigating the connection between these variables and host and microbiome interactions in neonatal development would be of great interest. As the "unculturable" era of microbiome research gives way to an intrinsically multidisciplinary field, microbiome research has reaped the advantages of technological advancements in next-generation sequencing, particularly 16S rRNA gene amplicon and shotgun sequencing, which have considerably expanded our knowledge about gut microbiota development during early life. Using omics approaches to explore the neonatal microbiome may help to better understand the link between the microbiome and newborn diseases. Herein, we summarized the metagenomics methods and tools used to advance knowledge on the neonatal microbiome origin and evolution and how the microbiome shapes early and late individuals' lives for health and disease. The way to overcome limitations in neonatal microbiome studies will be discussed.
Collapse
Affiliation(s)
- Zakia Boudar
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sofia Sehli
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sara El Janahi
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Najib Al Idrissi
- Department of Surgery, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Salsabil Hamdi
- Laboratory of Genomics and Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Nouzha Dini
- Mother and Child Department, Cheikh Khalifa International University Hospital, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Hassan Brim
- Department of Pathology, Howard University, Washington, DC, United States
| | - Saaïd Amzazi
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, and Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Chakib Nejjari
- Department of Epidemiology and Biostatistics, International School of Public Health, Mohammed VI University of Health Sciences, Casablanca, Morocco
- Department of Epidemiology and Public Health, Faculty of Medicine, University Sidi Mohammed Ben Abdellah, Fez, Morocco
| | | | - Hassan Ghazal
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
- National Center for Scientific and Technical Research, Rabat, Morocco
| |
Collapse
|
40
|
Hua H, Meydan C, Afshin EE, Lili LN, D’Adamo CR, Rickard N, Dudley JT, Price ND, Zhang B, Mason CE. A Wipe-Based Stool Collection and Preservation Kit for Microbiome Community Profiling. Front Immunol 2022; 13:889702. [PMID: 35711426 PMCID: PMC9196042 DOI: 10.3389/fimmu.2022.889702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
While a range of methods for stool collection exist, many require complicated, self-directed protocols and stool transfer. In this study, we introduce and validate a novel, wipe-based approach to fecal sample collection and stabilization for metagenomics analysis. A total of 72 samples were collected across four different preservation types: freezing at -20°C, room temperature storage, a commercial DNA preservation kit, and a dissolvable wipe used with DESS (dimethyl sulfoxide, ethylenediaminetetraacetic acid, sodium chloride) solution. These samples were sequenced and analyzed for taxonomic abundance metrics, bacterial metabolic pathway classification, and diversity analysis. Overall, the DESS wipe results validated the use of a wipe-based capture method to collect stool samples for microbiome analysis, showing an R2 of 0.96 for species across all kingdoms, as well as exhibiting a maintenance of Shannon diversity (3.1-3.3) and species richness (151-159) compared to frozen samples. Moreover, DESS showed comparable performance to the commercially available preservation kit (R2 of 0.98), and samples consistently clustered by subject across each method. These data support that the DESS wipe method can be used for stable, room temperature collection and transport of human stool specimens.
Collapse
Affiliation(s)
- Hui Hua
- Thorne HealthTech, New York, NY, United States
| | - Cem Meydan
- Thorne HealthTech, New York, NY, United States
| | | | | | - Christopher R. D’Adamo
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | | | - Nathan D. Price
- Thorne HealthTech, New York, NY, United States
- Institute for Systems Biology, Seattle, WA, United States
| | - Bodi Zhang
- Thorne HealthTech, New York, NY, United States
| | - Christopher E. Mason
- Thorne HealthTech, New York, NY, United States
- The WorldQuant Initiative for Quantitative Prediction, New York, NY, United States
| |
Collapse
|
41
|
Bombin A, Yan S, Bombin S, Mosley JD, Ferguson JF. Obesity influences composition of salivary and fecal microbiota and impacts the interactions between bacterial taxa. Physiol Rep 2022; 10:e15254. [PMID: 35384379 PMCID: PMC8980904 DOI: 10.14814/phy2.15254] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/04/2022] [Accepted: 03/17/2022] [Indexed: 04/23/2023] Open
Abstract
Obesity is an increasing global health concern and is associated with a broad range of morbidities. The gut microbiota are increasingly recognized as important contributors to obesity and cardiometabolic health. This study aimed to characterize oral and gut microbial communities, and evaluate host: microbiota interactions between clinical obesity classifications. We performed 16S rRNA sequencing on fecal and salivary samples, global metabolomics profiling on plasma and stool samples, and dietary profiling in 135 healthy individuals. We grouped individuals by obesity status, based on body mass index (BMI), including lean (BMI 18-124.9), overweight (BMI 25-29.9), or obese (BMI ≥30). We analyzed differences in microbiome composition, community inter-relationships, and predicted microbial function by obesity status. We found that salivary bacterial communities of lean and obese individuals were compositionally and phylogenetically distinct. An increase in obesity status was positively associated with strong correlations between bacterial taxa, particularly with bacterial groups implicated in metabolic disorders including Fretibacterium, and Tannerella. Consumption of sweeteners, especially xylitol, significantly influenced compositional and phylogenetic diversities of salivary and fecal bacterial communities. In addition, obesity groups exhibited differences in predicted bacterial metabolic activity, which was correlated with host's metabolite concentrations. Overall, obesity was associated with distinct changes in bacterial community dynamics, particularly in saliva. Consideration of microbiome community structure and inclusion of salivary samples may improve our ability to understand pathways linking microbiota to obesity and cardiometabolic disease.
Collapse
Affiliation(s)
- Andrei Bombin
- Division of Clinical PharmacologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Shun Yan
- Department of GeneticsThe University of AlabamaBirminghamAlabamaUSA
| | - Sergei Bombin
- Department of Biological SciencesThe University of AlabamaTuscaloosaAlabamaUSA
| | - Jonathan D. Mosley
- Division of Clinical PharmacologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jane F. Ferguson
- Division of Cardiovascular MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Microbiome Innovation Center (VMIC)NashvilleTennesseeUSA
| |
Collapse
|
42
|
Sharma R. Emerging Interrelationship Between the Gut Microbiome and Cellular Senescence in the Context of Aging and Disease: Perspectives and Therapeutic Opportunities. Probiotics Antimicrob Proteins 2022; 14:648-663. [PMID: 34985682 PMCID: PMC8728710 DOI: 10.1007/s12602-021-09903-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2021] [Indexed: 12/12/2022]
Abstract
The significance of diversity, composition, and functional attributes of the gut microbiota in shaping human health is well recognized. Studies have shown that gut microbiota is closely linked to human aging, and changes in the gut microbiome can predict human survival and longevity. In addition, a causal relationship between gut microbiota dysbiosis and chronic age-related disorders is also becoming apparent. Recent advances in our understanding of the cellular and molecular aspects of biological aging have revealed a cellular senescence-centric view of the aging process. However, the association between the gut microbiome and cellular senescence is only beginning to be understood. The present review provides an integrative view of the evolving relationship between the gut microbiome and cellular senescence in aging and disease. Evidence relating to microbiome-mediated modulation of senescent cells, as well as senescent cells-mediated changes in intestinal homeostasis and diseases, have been discussed. Unanswered questions and future research directions have also been deliberated to truly ascertain the relationship between the gut microbiome and cellular senescence for developing microbiome-based age-delaying and longevity-promoting therapies.
Collapse
Affiliation(s)
- Rohit Sharma
- Faculty of Applied Sciences & Biotechnology, Shoolini University, Solan, 173229, India.
| |
Collapse
|
43
|
Pettersen VK, Antunes LCM, Dufour A, Arrieta MC. Inferring early-life host and microbiome functions by mass spectrometry-based metaproteomics and metabolomics. Comput Struct Biotechnol J 2021; 20:274-286. [PMID: 35024099 PMCID: PMC8718658 DOI: 10.1016/j.csbj.2021.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 12/17/2022] Open
Abstract
Humans have a long-standing coexistence with microorganisms. In particular, the microbial community that populates the human gastrointestinal tract has emerged as a critical player in governing human health and disease. DNA and RNA sequencing techniques that map taxonomical composition and genomic potential of the gut community have become invaluable for microbiome research. However, deriving a biochemical understanding of how activities of the gut microbiome shape host development and physiology requires an expanded experimental design that goes beyond these approaches. In this review, we explore advances in high-throughput techniques based on liquid chromatography-mass spectrometry. These omics methods for the identification of proteins and metabolites have enabled direct characterisation of gut microbiome functions and the crosstalk with the host. We discuss current metaproteomics and metabolomics workflows for producing functional profiles, the existing methodological challenges and limitations, and recent studies utilising these techniques with a special focus on early life gut microbiome.
Collapse
Affiliation(s)
- Veronika Kuchařová Pettersen
- Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
- Pediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Centre for New Antibacterial Strategies, UiT The Arctic University of Norway, Tromsø, Norway
| | - Luis Caetano Martha Antunes
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
- National Institute of Science and Technology of Innovation on Diseases of Neglected Populations, Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Antoine Dufour
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada
| | - Marie-Claire Arrieta
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
44
|
A Meta-Omics Analysis Unveils the Shift in Microbial Community Structures and Metabolomics Profiles in Mangrove Sediments Treated with a Selective Actinobacterial Isolation Procedure. Molecules 2021; 26:molecules26237332. [PMID: 34885912 PMCID: PMC8658942 DOI: 10.3390/molecules26237332] [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: 10/06/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
Mangrove sediment ecosystems in the coastal areas of the Yucatan peninsula are unique environments, influenced by their karstic origin and connection with the world’s largest underground river. The microbial communities residing in these sediments are influenced by the presence of mangrove roots and the trading chemistry for communication between sediment bacteria and plant roots can be targeted for secondary metabolite research. To explore the secondary metabolite production potential of microbial community members in mangrove sediments at the “El Palmar” natural reserve in Sisal, Yucatan, a combined meta-omics approach was applied. The effects of a cultivation medium reported to select for actinomycetes within mangrove sediments’ microbial communities was also analyzed. The metabolome of the microbial communities was analyzed by high-resolution liquid chromatography-tandem mass spectrometry, and molecular networking analysis was used to investigate if known natural products and their variants were present. Metagenomic results suggest that the sediments from “El Palmar” harbor a stable bacterial community independently of their distance from mangrove tree roots. An unexpected decrease in the observed abundance of actinomycetes present in the communities occurred when an antibiotic-amended medium considered to be actinomycete-selective was applied for a 30-day period. However, the use of this antibiotic-amended medium also enhanced production of secondary metabolites within the microbial community present relative to the water control, suggesting the treatment selected for antibiotic-resistant bacteria capable of producing a higher number of secondary metabolites. Secondary metabolite mining of “El Palmar” microbial community metagenomes identified polyketide synthase and non-ribosomal peptide synthetases’ biosynthetic genes in all analyzed metagenomes. The presence of these genes correlated with the annotation of several secondary metabolites from the Global Natural Product Social Molecular Networking database. These results highlight the biotechnological potential of the microbial communities from “El Palmar”, and show the impact selective media had on the composition of communities of actinobacteria.
Collapse
|
45
|
Bagheri M, Shah RD, Mosley JD, Ferguson JF. A metabolome and microbiome wide association study of healthy eating index points to the mechanisms linking dietary pattern and metabolic status. Eur J Nutr 2021; 60:4413-4427. [PMID: 34057579 PMCID: PMC8572162 DOI: 10.1007/s00394-021-02599-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Healthy eating index (HEI), a measure of diet quality, associates with metabolic health outcomes; however, the molecular basis is unclear. We conducted a multi-omic study to examine whether HEI associates with the circulatory and gut metabolome and investigated the gut microbiome-HEI interaction on circulating and gut metabolites. METHODS Through a cross-sectional study, we evaluated diet quality in healthy individuals [the ABO Glycoproteomics in Platelets and Endothelial Cells (ABO) Study, n = 73], metabolites (measured at Metabolon Inc.) in plasma (n = 800) and gut (n = 767) and the gut microbiome at enterotype and microbial taxa (n = 296) levels. Pathway analysis was conducted using Metaboanalyst 4.0. We performed multi-variable linear regression to explore both the HEI-metabolites and HEI-microbiome associations and how metabolites were affected by the HEI-microbiome interaction. In the Fish oils and Adipose Inflammation Reduction (FAIR) Study (n = 25), analyses on HEI and plasma metabolites were replicated. Estimates of findings from both studies were pooled in random-effects meta-analysis. RESULTS The HEI-2015 was associated with 74 plasma and 73 gut metabolites (mostly lipids) and with 47 metabolites in the meta-analysis of the ABO and FAIR Studies. Compared to Enterotype-1 participants, those with Enterotype-2 had higher diet quality (p = 0.01). We also identified 9 microbial genera associated with HEI, and 35 plasma and 40 gut metabolites linked to the HEI-gut microbiome interaction. Pathways involved in the metabolism of polar lipids, amino acids and caffeine strongly associated with diet quality. However, the HEI-microbiome interaction not only influenced the pathways involved in the metabolism of branch-chain amino acids, it also affected upstream pathways including nucleotide metabolism and amino acids biosynthesis. CONCLUSIONS Our multi-omic analysis demonstrated that changes in metabolism, measured by either circulatory/gut metabolites or metabolic pathways, are influenced by not only diet quality but also gut microbiome alterations shaped by the quality of diet consumed. Future work is needed to explore the causality in the interplay between HEI and gut-microbiome composition in metabolism.
Collapse
Affiliation(s)
- Minoo Bagheri
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB 354B, Nashville, TN, 37232, USA
| | - Rachana D Shah
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan D Mosley
- Division of Clinical Pharmacology, Department of Medicine and Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB 354B, Nashville, TN, 37232, USA.
| |
Collapse
|
46
|
Samieri C, Yassine HN, Melo van Lent D, Lefèvre-Arbogast S, van de Rest O, Bowman GL, Scarmeas N. Personalized nutrition for dementia prevention. Alzheimers Dement 2021; 18:1424-1437. [PMID: 34757699 DOI: 10.1002/alz.12486] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
The role of nutrition has been investigated for decades under the assumption of one-size-fits-all. Yet there is heterogeneity in metabolic and neurobiological responses to diet. Thus a more personalized approach may better fit biological reality and have increased efficacy to prevent dementia. Personalized nutrition builds on the food exposome, defined as the history of diet-related exposures over the lifetime, and on its interactions with the genome and other biological characteristics (eg, metabolism, the microbiome) to shape health. We review current advances of personalized nutrition in dementia research. We discuss key questions, success milestones, and future roadmap from observational epidemiology to clinical studies through basic science. A personalized nutrition approach based on the best prescription for the most appropriate target population in the most relevant time-window has the potential to strengthen dementia-prevention efforts.
Collapse
Affiliation(s)
- Cécilia Samieri
- Univ. Bordeaux, ISPED, Inserm, Bordeaux Population Health Research Center, Bordeaux, France
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine USC, Los Angeles, California, USA.,Department of Neurology, Keck School of Medicine USC, Los Angeles, California, USA
| | - Debora Melo van Lent
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Ondine van de Rest
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands
| | - Gene L Bowman
- Department of Neurology and Layton Aging and Alzheimer's Disease Center, Oregon Health and Science University, Portland, Oregon, USA.,Helfgott Research Institute, National University of Natural Medicine, Portland, Oregon, USA
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Taub Institute for Research in Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, USA
| |
Collapse
|
47
|
The impact of the Th17:Treg axis on the IgA-Biome across the glycemic spectrum. PLoS One 2021; 16:e0258812. [PMID: 34669745 PMCID: PMC8528330 DOI: 10.1371/journal.pone.0258812] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022] Open
Abstract
Secretory IgA (SIgA) is released into mucosal surfaces where its function extends beyond that of host defense to include the shaping of resident microbial communities by mediating exclusion/inclusion of respective microbes and regulating bacterial gene expression. In this capacity, SIgA acts as the fulcrum on which host immunity and the health of the microbiota are balanced. We recently completed an analysis of the gut and salivary IgA-Biomes (16S rDNA sequencing of SIgA-coated/uncoated bacteria) in Mexican-American adults that identified IgA-Biome differences across the glycemic spectrum. As Th17:Treg ratio imbalances are associated with gut microbiome dysbiosis and chronic inflammatory conditions such as type 2 diabetes, the present study extends our prior work by examining the impact of Th17:Treg ratios (pro-inflammatory:anti-inflammatory T-cell ratios) and the SIgA response (Th17:Treg-SIgA axis) in shaping microbial communities. Examining the impact of Th17:Treg ratios (determined by epigenetic qPCR lymphocyte subset quantification) on the IgA-Biome across diabetes phenotypes identified a proportional relationship between Th17:Treg ratios and alpha diversity in the stool IgA-Biome of those with dysglycemia, significant changes in community composition of the stool and salivary microbiomes across glycemic profiles, and genera preferentially abundant by T-cell inflammatory phenotype. This is the first study to associate epigenetically quantified Th17:Treg ratios with both the larger and SIgA-fractionated microbiome, assess these associations in the context of a chronic inflammatory disease, and offers a novel frame through which to evaluate mucosal microbiomes in the context of host responses and inflammation.
Collapse
|
48
|
DE-Oliveira GJM, Schieferdecker MEM, Campos ACL. ARE ENTEROTYPES IN OBESE MODIFIED BY BARIATRIC SURGERY, THE USE OF PROBIOTIC SUPPLEMENTS AND FOOD HABITS? ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2021; 34:e1601. [PMID: 34669890 PMCID: PMC8521827 DOI: 10.1590/0102-672020210002e1601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/11/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Studies suggest that bariatric surgery, use of probiotic supplements and the dietary pattern can change enterotypes, as well as the entire microbial population. OBJECTIVE To verify the influence of bariatric surgery, the use of probiotic supplements and eating habits on enterotypes in obese patients. METHODS Articles published between the 2015 and 2020 were searched in Lilacs and PubMed with the headings: probiotics, eating behavior, food consumption, food, diet, microbiota, gastrointestinal microbiome, bariatric surgery, gastric bypass and the keyword enterotype in Portuguese, English and Spanish. RESULTS Of the 260 articles found, only studies carried out in obese adults relating changes in the enterotype after bariatric surgery or use of probiotics or dietary patterns and original articles were selected. In the end, eight papers on enterotype change and bariatric surgery were selected and categorized, four on the relationship between food consumption and microbiota and one on the effects of probiotics on enterotypes. CONCLUSION The microbial structure is widely modified after bariatric surgery, since the use of probiotic supplement does not bring lasting changes. Enterotypes appear to be shaped by long-term dietary patterns, can modulate how nutrients are metabolized and can be a useful biomarker to improve clinical management.
Collapse
|
49
|
Sharma V, Sharma V, Shahjouei S, Li J, Chaudhary D, Khan A, Wolk DM, Zand R, Abedi V. At the Intersection of Gut Microbiome and Stroke: A Systematic Review of the Literature. Front Neurol 2021; 12:729399. [PMID: 34630304 PMCID: PMC8498333 DOI: 10.3389/fneur.2021.729399] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Ischemic and hemorrhagic stroke are associated with a high rate of long-term disability and death. Recent investigations focus efforts to better understand how alterations in gut microbiota composition influence clinical outcomes. A key metabolite, trimethylamine N-oxide (TMAO), is linked to multiple inflammatory, vascular, and oxidative pathways. The current biochemical underpinnings of microbial effects on stroke remain largely understudied. The goal of our study is to explore the current literature to explain the interactions between the human gut microbiome and stroke progression, recovery, and outcome. We also provide a descriptive review of TMAO. Methods: A systematic literature search of published articles between January 1, 1990, and March 22, 2020, was performed on the PubMed database to identify studies addressing the role of the microbiome and TMAO in the pathogenesis and recovery of acute stroke. Our initial investigation focused on human subject studies and was further expanded to include animal studies. Relevant articles were included, regardless of study design. The analysis included reviewers classifying and presenting selected articles by study design and sample size in a chart format. Results: A total of 222 titles and abstracts were screened. A review of the 68 original human subject articles resulted in the inclusion of 24 studies in this review. To provide further insight into TMAO as a key player, an additional 40 articles were also reviewed and included. Our findings highlighted that alterations in richness and abundance of gut microbes and increased plasma TMAO play an important role in vascular events and outcomes. Our analysis revealed that restoration of a healthy gut, through targeted TMAO-reducing therapies, could provide alternative secondary prevention for at-risk patients. Discussion: Biochemical interactions between the gut microbiome and inflammation, resulting in metabolic derangements, can affect stroke progression and outcomes. Clinical evidence supports the importance of TMAO in modulating underlying stroke risk factors. Lack of standardization and distinct differences in sample sizes among studies are major limitations.
Collapse
Affiliation(s)
- Vishakha Sharma
- Kansas City University College of Osteopathic Medicine, Kansas City, MO, United States
| | - Vaibhav Sharma
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | - Shima Shahjouei
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States
| | - Jiang Li
- Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA, United States
| | - Durgesh Chaudhary
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States
| | - Ayesha Khan
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States.,Geisinger Health System, Geisinger Northeast Internal Medicine Residency, Wilkes Barre, PA, United States
| | - Donna M Wolk
- Department of Laboratory Medicine, Geisinger Health System, Diagnostic Medicine Institute, Danville, PA, United States
| | - Ramin Zand
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States
| | - Vida Abedi
- Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA, United States.,Department of Public Health Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
50
|
Muller E, Algavi YM, Borenstein E. A meta-analysis study of the robustness and universality of gut microbiome-metabolome associations. MICROBIOME 2021; 9:203. [PMID: 34641974 PMCID: PMC8507343 DOI: 10.1186/s40168-021-01149-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 08/18/2021] [Indexed: 05/15/2023]
Abstract
BACKGROUND Microbiome-metabolome studies of the human gut have been gaining popularity in recent years, mostly due to accumulating evidence of the interplay between gut microbes, metabolites, and host health. Statistical and machine learning-based methods have been widely applied to analyze such paired microbiome-metabolome data, in the hope of identifying metabolites that are governed by the composition of the microbiome. Such metabolites can be likely modulated by microbiome-based interventions, offering a route for promoting gut metabolic health. Yet, to date, it remains unclear whether findings of microbially associated metabolites in any single study carry over to other studies or cohorts, and how robust and universal are microbiome-metabolites links. RESULTS In this study, we addressed this challenge by performing a comprehensive meta-analysis to identify human gut metabolites that can be predicted based on the composition of the gut microbiome across multiple studies. We term such metabolites "robustly well-predicted". To this end, we processed data from 1733 samples from 10 independent human gut microbiome-metabolome studies, focusing initially on healthy subjects, and implemented a machine learning pipeline to predict metabolite levels in each dataset based on the composition of the microbiome. Comparing the predictability of each metabolite across datasets, we found 97 robustly well-predicted metabolites. These include metabolites involved in important microbial pathways such as bile acid transformations and polyamines metabolism. Importantly, however, other metabolites exhibited large variation in predictability across datasets, suggesting a cohort- or study-specific relationship between the microbiome and the metabolite. Comparing taxonomic contributors to different models, we found that some robustly well-predicted metabolites were predicted by markedly different sets of taxa across datasets, suggesting that some microbially associated metabolites may be governed by different members of the microbiome in different cohorts. We finally examined whether models trained on a control group of a given study successfully predicted the metabolite's level in the disease group of the same study, identifying several metabolites where the model was not transferable, indicating a shift in microbial metabolism in disease-associated dysbiosis. CONCLUSIONS Combined, our findings provide a better understanding of the link between the microbiome and metabolites and allow researchers to put identified microbially associated metabolites within the context of other studies. Video abstract.
Collapse
Affiliation(s)
- Efrat Muller
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Yadid M. Algavi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elhanan Borenstein
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Santa Fe Institute, Santa Fe, NM USA
| |
Collapse
|