101
|
Zheng S, Zhou L, Hoene M, Peter A, Birkenfeld AL, Weigert C, Liu X, Zhao X, Xu G, Lehmann R. A New Biomarker Profiling Strategy for Gut Microbiome Research: Valid Association of Metabolites to Metabolism of Microbiota Detected by Non-Targeted Metabolomics in Human Urine. Metabolites 2023; 13:1061. [PMID: 37887386 PMCID: PMC10608496 DOI: 10.3390/metabo13101061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/09/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/28/2023] Open
Abstract
The gut microbiome is of tremendous relevance to human health and disease, so it is a hot topic of omics-driven biomedical research. However, a valid identification of gut microbiota-associated molecules in human blood or urine is difficult to achieve. We hypothesize that bowel evacuation is an easy-to-use approach to reveal such metabolites. A non-targeted and modifying group-assisted metabolomics approach (covering 40 types of modifications) was applied to investigate urine samples collected in two independent experiments at various time points before and after laxative use. Fasting over the same time period served as the control condition. As a result, depletion of the fecal microbiome significantly affected the levels of 331 metabolite ions in urine, including 100 modified metabolites. Dominating modifications were glucuronidations, carboxylations, sulfations, adenine conjugations, butyrylations, malonylations, and acetylations. A total of 32 compounds, including common, but also unexpected fecal microbiota-associated metabolites, were annotated. The applied strategy has potential to generate a microbiome-associated metabolite map (M3) of urine from healthy humans, and presumably also other body fluids. Comparative analyses of M3 vs. disease-related metabolite profiles, or therapy-dependent changes may open promising perspectives for human gut microbiome research and diagnostics beyond analyzing feces.
Collapse
Affiliation(s)
- Sijia Zheng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (S.Z.); (L.Z.); (X.L.); (X.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (S.Z.); (L.Z.); (X.L.); (X.Z.)
| | - Miriam Hoene
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tuebingen, Germany; (M.H.); (A.P.); (C.W.)
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tuebingen, Germany; (M.H.); (A.P.); (C.W.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 90451 Neuherberg, Germany
| | - Andreas L. Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 90451 Neuherberg, Germany
- Internal Medicine 4, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Cora Weigert
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tuebingen, Germany; (M.H.); (A.P.); (C.W.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 90451 Neuherberg, Germany
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (S.Z.); (L.Z.); (X.L.); (X.Z.)
| | - Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (S.Z.); (L.Z.); (X.L.); (X.Z.)
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (S.Z.); (L.Z.); (X.L.); (X.Z.)
| | - Rainer Lehmann
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tuebingen, Germany; (M.H.); (A.P.); (C.W.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 90451 Neuherberg, Germany
| |
Collapse
|
102
|
Fuller H, Zhu Y, Nicholas J, Chatelaine HA, Drzymalla EM, Sarvestani AK, Julián-Serrano S, Tahir UA, Sinnott-Armstrong N, Raffield LM, Rahnavard A, Hua X, Shutta KH, Darst BF. Metabolomic epidemiology offers insights into disease aetiology. Nat Metab 2023; 5:1656-1672. [PMID: 37872285 PMCID: PMC11164316 DOI: 10.1038/s42255-023-00903-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/30/2023] [Accepted: 09/06/2023] [Indexed: 10/25/2023]
Abstract
Metabolomic epidemiology is the high-throughput study of the relationship between metabolites and health-related traits. This emerging and rapidly growing field has improved our understanding of disease aetiology and contributed to advances in precision medicine. As the field continues to develop, metabolomic epidemiology could lead to the discovery of diagnostic biomarkers predictive of disease risk, aiding in earlier disease detection and better prognosis. In this Review, we discuss key advances facilitated by the field of metabolomic epidemiology for a range of conditions, including cardiometabolic diseases, cancer, Alzheimer's disease and COVID-19, with a focus on potential clinical utility. Core principles in metabolomic epidemiology, including study design, causal inference methods and multi-omic integration, are briefly discussed. Future directions required for clinical translation of metabolomic epidemiology findings are summarized, emphasizing public health implications. Further work is needed to establish which metabolites reproducibly improve clinical risk prediction in diverse populations and are causally related to disease progression.
Collapse
Affiliation(s)
- Harriett Fuller
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yiwen Zhu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jayna Nicholas
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Haley A Chatelaine
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Emily M Drzymalla
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Afrand K Sarvestani
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | | | - Usman A Tahir
- Department of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Xinwei Hua
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Katherine H Shutta
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Burcu F Darst
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
103
|
Widjaja F, Rietjens IMCM. From-Toilet-to-Freezer: A Review on Requirements for an Automatic Protocol to Collect and Store Human Fecal Samples for Research Purposes. Biomedicines 2023; 11:2658. [PMID: 37893032 PMCID: PMC10603957 DOI: 10.3390/biomedicines11102658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/29/2023] Open
Abstract
The composition, viability and metabolic functionality of intestinal microbiota play an important role in human health and disease. Studies on intestinal microbiota are often based on fecal samples, because these can be sampled in a non-invasive way, although procedures for sampling, processing and storage vary. This review presents factors to consider when developing an automated protocol for sampling, processing and storing fecal samples: donor inclusion criteria, urine-feces separation in smart toilets, homogenization, aliquoting, usage or type of buffer to dissolve and store fecal material, temperature and time for processing and storage and quality control. The lack of standardization and low-throughput of state-of-the-art fecal collection procedures promote a more automated protocol. Based on this review, an automated protocol is proposed. Fecal samples should be collected and immediately processed under anaerobic conditions at either room temperature (RT) for a maximum of 4 h or at 4 °C for no more than 24 h. Upon homogenization, preferably in the absence of added solvent to allow addition of a buffer of choice at a later stage, aliquots obtained should be stored at either -20 °C for up to a few months or -80 °C for a longer period-up to 2 years. Protocols for quality control should characterize microbial composition and viability as well as metabolic functionality.
Collapse
Affiliation(s)
- Frances Widjaja
- Division of Toxicology, Wageningen University & Research, 6708 WE Wageningen, The Netherlands;
| | | |
Collapse
|
104
|
Chang CC, Liu TC, Lu CJ, Chiu HC, Lin WN. Machine learning strategy for identifying altered gut microbiomes for diagnostic screening in myasthenia gravis. Front Microbiol 2023; 14:1227300. [PMID: 37829445 PMCID: PMC10565662 DOI: 10.3389/fmicb.2023.1227300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/23/2023] [Accepted: 09/06/2023] [Indexed: 10/14/2023] Open
Abstract
Myasthenia gravis (MG) is a neuromuscular junction disease with a complex pathophysiology and clinical variation for which no clear biomarker has been discovered. We hypothesized that because changes in gut microbiome composition often occur in autoimmune diseases, the gut microbiome structures of patients with MG would differ from those without, and supervised machine learning (ML) analysis strategy could be trained using data from gut microbiota for diagnostic screening of MG. Genomic DNA from the stool samples of MG and those without were collected and established a sequencing library by constructing amplicon sequence variants (ASVs) and completing taxonomic classification of each representative DNA sequence. Four ML methods, namely least absolute shrinkage and selection operator, extreme gradient boosting (XGBoost), random forest, and classification and regression trees with nested leave-one-out cross-validation were trained using ASV taxon-based data and full ASV-based data to identify key ASVs in each data set. The results revealed XGBoost to have the best predicted performance. Overlapping key features extracted when XGBoost was trained using the full ASV-based and ASV taxon-based data were identified, and 31 high-importance ASVs (HIASVs) were obtained, assigned importance scores, and ranked. The most significant difference observed was in the abundance of bacteria in the Lachnospiraceae and Ruminococcaceae families. The 31 HIASVs were used to train the XGBoost algorithm to differentiate individuals with and without MG. The model had high diagnostic classification power and could accurately predict and identify patients with MG. In addition, the abundance of Lachnospiraceae was associated with limb weakness severity. In this study, we discovered that the composition of gut microbiomes differed between MG and non-MG subjects. In addition, the proposed XGBoost model trained using 31 HIASVs had the most favorable performance with respect to analyzing gut microbiomes. These HIASVs selected by the ML model may serve as biomarkers for clinical use and mechanistic study in the future. Our proposed ML model can identify several taxonomic markers and effectively discriminate patients with MG from those without with a high accuracy, the ML strategy can be applied as a benchmark to conduct noninvasive screening of MG.
Collapse
Affiliation(s)
- Che-Cheng Chang
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Neurology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Tzu-Chi Liu
- Graduate Institute of Business Administration, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chi-Jie Lu
- Graduate Institute of Business Administration, Fu Jen Catholic University, New Taipei City, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Information Management, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hou-Chang Chiu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Neurology, Taipei Medical University, Shuang-Ho Hospital, New Taipei City, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
105
|
Juśkiewicz J, Ognik K, Fotschki J, Napiórkowska D, Cholewińska E, Grzelak-Błaszczyk K, Krauze M, Fotschki B. The Effects of Dietary Chromium Supplementation along with Discontinuing a High-Fat Diet on the Microbial Enzymatic Activity and the Production of SCFAs in the Faeces of Rats. Nutrients 2023; 15:3962. [PMID: 37764746 PMCID: PMC10534834 DOI: 10.3390/nu15183962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/25/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The present study assessed the changes in faecal microbial activity in obese Wistar rats fed high-fat or low-fat diets supplemented with various forms of chromium (picolinate or nanoparticles). The 18-week study was divided into two phases: an introductory period (9 weeks; obesity status induction via a high-fat diet) and an experimental period (9 weeks; maintained on a high-fat diet or switched to a low-fat diet and Cr supplementation). During the experimental period (10-18 weeks of feeding), samples of fresh faeces were collected on chosen days. The bacterial enzymatic activity and short-chain fatty acids (SCFAs) concentration were assessed to characterise the dynamism of the changes in faecal microbial metabolic activity under the applied dietary treatments. The results indicated that faecal microbial metabolic activity displayed several adaptation mechanisms in response to modifications in dietary conditions, and a beneficial outcome resulted from a pro-healthy dietary habit change, that is, switching from a high-fat to a low-fat diet. Dietary supplementation with chromium nanoparticles further modulated the aforementioned microbial activity, i.e., diminished the extracellular and total enzymatic activities, while the effect of chromium picolinate addition was negligible. Both the high-fat diet and the addition of chromium nanoparticles reduced SCFA concentrations and increased the faecal pH values.
Collapse
Affiliation(s)
- Jerzy Juśkiewicz
- Division of Food Science, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland; (J.F.); (D.N.); (B.F.)
| | - Katarzyna Ognik
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland; (K.O.); (E.C.); (M.K.)
| | - Joanna Fotschki
- Division of Food Science, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland; (J.F.); (D.N.); (B.F.)
| | - Dorota Napiórkowska
- Division of Food Science, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland; (J.F.); (D.N.); (B.F.)
| | - Ewelina Cholewińska
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland; (K.O.); (E.C.); (M.K.)
| | - Katarzyna Grzelak-Błaszczyk
- Institute of Food Technology and Analysis, Łódź University of Technology, Stefanowskiego 2/22, 90-537 Łódź, Poland;
| | - Magdalena Krauze
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland; (K.O.); (E.C.); (M.K.)
| | - Bartosz Fotschki
- Division of Food Science, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland; (J.F.); (D.N.); (B.F.)
| |
Collapse
|
106
|
Wuyts S, Alves R, Zimmermann‐Kogadeeva M, Nishijima S, Blasche S, Driessen M, Geyer PE, Hercog R, Kartal E, Maier L, Müller JB, Garcia Santamarina S, Schmidt TSB, Sevin DC, Telzerow A, Treit PV, Wenzel T, Typas A, Patil KR, Mann M, Kuhn M, Bork P. Consistency across multi-omics layers in a drug-perturbed gut microbial community. Mol Syst Biol 2023; 19:e11525. [PMID: 37485738 PMCID: PMC10495815 DOI: 10.15252/msb.202311525] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/09/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Multi-omics analyses are used in microbiome studies to understand molecular changes in microbial communities exposed to different conditions. However, it is not always clear how much each omics data type contributes to our understanding and whether they are concordant with each other. Here, we map the molecular response of a synthetic community of 32 human gut bacteria to three non-antibiotic drugs by using five omics layers (16S rRNA gene profiling, metagenomics, metatranscriptomics, metaproteomics and metabolomics). We find that all the omics methods with species resolution are highly consistent in estimating relative species abundances. Furthermore, different omics methods complement each other for capturing functional changes. For example, while nearly all the omics data types captured that the antipsychotic drug chlorpromazine selectively inhibits Bacteroidota representatives in the community, the metatranscriptome and metaproteome suggested that the drug induces stress responses related to protein quality control. Metabolomics revealed a decrease in oligosaccharide uptake, likely caused by Bacteroidota depletion. Our study highlights how multi-omics datasets can be utilized to reveal complex molecular responses to external perturbations in microbial communities.
Collapse
Affiliation(s)
- Sander Wuyts
- European Molecular Biology LaboratoryHeidelbergGermany
| | - Renato Alves
- European Molecular Biology LaboratoryHeidelbergGermany
| | | | | | - Sonja Blasche
- European Molecular Biology LaboratoryHeidelbergGermany
- Medical Research Council Toxicology UnitCambridgeUK
| | | | - Philipp E Geyer
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Rajna Hercog
- European Molecular Biology LaboratoryHeidelbergGermany
| | - Ece Kartal
- European Molecular Biology LaboratoryHeidelbergGermany
| | - Lisa Maier
- European Molecular Biology LaboratoryHeidelbergGermany
| | - Johannes B Müller
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Sarela Garcia Santamarina
- European Molecular Biology LaboratoryHeidelbergGermany
- Present address:
MOSTMICRO Unit, Instituto de Tecnologia Quimica e BiologicaUniversidade Nova de LisboaOeirasPortugal
| | | | | | - Anja Telzerow
- European Molecular Biology LaboratoryHeidelbergGermany
| | - Peter V Treit
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Tobias Wenzel
- European Molecular Biology LaboratoryHeidelbergGermany
- Present address:
Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological SciencesPontificia Universidad Catolica de ChileSantiagoChile
| | | | - Kiran R Patil
- European Molecular Biology LaboratoryHeidelbergGermany
- Medical Research Council Toxicology UnitCambridgeUK
| | - Matthias Mann
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
- Proteomics Program, NNF Center for Protein Research, Faculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Michael Kuhn
- European Molecular Biology LaboratoryHeidelbergGermany
| | - Peer Bork
- European Molecular Biology LaboratoryHeidelbergGermany
- Max Delbrück Centre for Molecular MedicineBerlinGermany
- Yonsei Frontier Lab (YFL)Yonsei UniversitySeoulSouth Korea
- Department of Bioinformatics, BiocenterUniversity of WürzburgWürzburgGermany
| |
Collapse
|
107
|
He S, Sun L, Chen J, Ouyang Y. Recent Advances and Perspectives in Relation to the Metabolomics-Based Study of Diabetic Retinopathy. Metabolites 2023; 13:1007. [PMID: 37755287 PMCID: PMC10536395 DOI: 10.3390/metabo13091007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/13/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Diabetic retinopathy (DR), a prevalent microvascular complication of diabetes, is a major cause of acquired blindness in adults. Currently, a clinical diagnosis of DR primarily relies on fundus fluorescein angiography, with a limited availability of effective biomarkers. Metabolomics, a discipline dedicated to scrutinizing the response of various metabolites within living organisms, has shown noteworthy advancements in uncovering metabolic disorders and identifying key metabolites associated with DR in recent years. Consequently, this review aims to present the latest advancements in metabolomics techniques and comprehensively discuss the principal metabolic outcomes derived from analyzing blood, vitreous humor, aqueous humor, urine, and fecal samples.
Collapse
Affiliation(s)
| | | | | | - Yang Ouyang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.H.)
| |
Collapse
|
108
|
Takeuchi T, Kubota T, Nakanishi Y, Tsugawa H, Suda W, Kwon ATJ, Yazaki J, Ikeda K, Nemoto S, Mochizuki Y, Kitami T, Yugi K, Mizuno Y, Yamamichi N, Yamazaki T, Takamoto I, Kubota N, Kadowaki T, Arner E, Carninci P, Ohara O, Arita M, Hattori M, Koyasu S, Ohno H. Gut microbial carbohydrate metabolism contributes to insulin resistance. Nature 2023; 621:389-395. [PMID: 37648852 PMCID: PMC10499599 DOI: 10.1038/s41586-023-06466-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/25/2022] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Insulin resistance is the primary pathophysiology underlying metabolic syndrome and type 2 diabetes1,2. Previous metagenomic studies have described the characteristics of gut microbiota and their roles in metabolizing major nutrients in insulin resistance3-9. In particular, carbohydrate metabolism of commensals has been proposed to contribute up to 10% of the host's overall energy extraction10, thereby playing a role in the pathogenesis of obesity and prediabetes3,4,6. Nevertheless, the underlying mechanism remains unclear. Here we investigate this relationship using a comprehensive multi-omics strategy in humans. We combine unbiased faecal metabolomics with metagenomics, host metabolomics and transcriptomics data to profile the involvement of the microbiome in insulin resistance. These data reveal that faecal carbohydrates, particularly host-accessible monosaccharides, are increased in individuals with insulin resistance and are associated with microbial carbohydrate metabolisms and host inflammatory cytokines. We identify gut bacteria associated with insulin resistance and insulin sensitivity that show a distinct pattern of carbohydrate metabolism, and demonstrate that insulin-sensitivity-associated bacteria ameliorate host phenotypes of insulin resistance in a mouse model. Our study, which provides a comprehensive view of the host-microorganism relationships in insulin resistance, reveals the impact of carbohydrate metabolism by microbiota, suggesting a potential therapeutic target for ameliorating insulin resistance.
Collapse
Affiliation(s)
- Tadashi Takeuchi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Tetsuya Kubota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Division of Diabetes and Metabolism, The Institute for Medical Science Asahi Life Foundation, Tokyo, Japan.
- Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan.
| | - Yumiko Nakanishi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
| | - Hiroshi Tsugawa
- Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science (CSRS), Yokohama, Japan
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Wataru Suda
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Andrew Tae-Jun Kwon
- Laboratory for Applied Regulatory Genomics Network Analysis, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Junshi Yazaki
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Shino Nemoto
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Yoshiki Mochizuki
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Toshimori Kitami
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Katsuyuki Yugi
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Institute for Advanced Biosciences, Keio University, Fujisawa, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiko Mizuno
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
- Development Bank of Japan, Tokyo, Japan
| | - Nobutake Yamamichi
- Center for Epidemiology and Preventive Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | | | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, Ami Town, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Erik Arner
- Laboratory for Applied Regulatory Genomics Network Analysis, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Fondazione Human Technopole, Milan, Italy
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
| | - Masahira Hattori
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
109
|
Ruocco C, Malavazos AE, Ragni M, Carruba MO, Valerio A, Iacobellis G, Nisoli E. Amino acids contribute to adaptive thermogenesis. New insights into the mechanisms of action of recent drugs for metabolic disorders are emerging. Pharmacol Res 2023; 195:106892. [PMID: 37619907 DOI: 10.1016/j.phrs.2023.106892] [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] [Academic Contribution Register] [Received: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Adaptive thermogenesis is the heat production by muscle contractions (shivering thermogenesis) or brown adipose tissue (BAT) and beige fat (non-shivering thermogenesis) in response to external stimuli, including cold exposure. BAT and beige fat communicate with peripheral organs and the brain through a variegate secretory and absorption processes - controlling adipokines, microRNAs, extracellular vesicles, and metabolites - and have received much attention as potential therapeutic targets for managing obesity-related disorders. The sympathetic nervous system and norepinephrine-releasing adipose tissue macrophages (ATM) activate uncoupling protein 1 (UCP1), expressed explicitly in brown and beige adipocytes, dissolving the electrochemical gradient and uncoupling tricarboxylic acid cycle and the electron transport chain from ATP production. Mounting evidence has attracted attention to the multiple effects of dietary and endogenously synthesised amino acids in BAT thermogenesis and metabolic phenotype in animals and humans. However, the mechanisms implicated in these processes have yet to be conclusively characterized. In the present review article, we aim to define the principal investigation areas in this context, including intestinal microbiota constitution, adipose autophagy modulation, and secretome and metabolic fluxes control, which lead to increased brown/beige thermogenesis. Finally, also based on our recent epicardial adipose tissue results, we summarise the evidence supporting the notion that the new dual and triple agonists of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon (GCG) receptor - with never before seen weight loss and insulin-sensitizing efficacy - promote thermogenic-like amino acid profiles in BAT with robust heat production and likely trigger sympathetic activation and adaptive thermogenesis by controlling amino acid metabolism and ATM expansion in BAT and beige fat.
Collapse
Affiliation(s)
- Chiara Ruocco
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alexis Elias Malavazos
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Piazza Edmondo Malan, 2, San Donato Milanese, 20097 Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, via della Commenda, 10, 20122 Milan, Italy
| | - Maurizio Ragni
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Michele O Carruba
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa, 11, 25123 Brescia, Italy
| | - Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL, USA
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy.
| |
Collapse
|
110
|
Celano G, Calabrese FM, Riezzo G, D’Attoma B, Ignazzi A, Di Chito M, Sila A, De Nucci S, Rinaldi R, Linsalata M, Vacca M, Apa CA, Angelis MD, Giannelli G, De Pergola G, Russo F. Effects of a Very-Low-Calorie Ketogenic Diet on the Fecal and Urinary Volatilome in an Obese Patient Cohort: A Preliminary Investigation. Nutrients 2023; 15:3752. [PMID: 37686784 PMCID: PMC10490432 DOI: 10.3390/nu15173752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Several recent studies deepened the strong connection between gut microbiota and obesity. The effectiveness of the very-low-calorie ketogenic diet (VLCKD) has been measured in terms of positive impact on the host homeostasis, but little is known of the modification exerted on the intestinal metabolome. To inspect this complex relationship, we analyzed both fecal and urinary metabolome in terms of volatile organic compounds (VOCs) by the GC-MS method in 25 obese patients that were under VLCKD for eight weeks. Partial least square discriminant analysis evidenced specific urinary and fecal metabolites whose profile can be considered a signature of a partial restore toward the host eubiosis. Specifically, among various keystone VOCs, the decreased concentration of four statistically significant fecal esters (i.e., propanoic acid pentyl ester, butanoic acid hexyl ester, butanoic acid pentyl ester, and pentanoic acid butyl ester) supports the positive effect of VLCKD treatment. Our pilot study results suggest a potential positive effect of VLCKD intervention affecting fecal and urinary volatilome profiles from obese patients. Meta-omics techniques including the study of genes and transcripts will help in developing new interventions useful in preventing or treating obesity and its associated health problems.
Collapse
Affiliation(s)
- Giuseppe Celano
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (M.V.); (C.A.A.); (M.D.A.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (M.V.); (C.A.A.); (M.D.A.)
| | - Giuseppe Riezzo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Benedetta D’Attoma
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Antonia Ignazzi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Martina Di Chito
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Annamaria Sila
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Sara De Nucci
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Roberta Rinaldi
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Michele Linsalata
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Mirco Vacca
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (M.V.); (C.A.A.); (M.D.A.)
| | - Carmen Aurora Apa
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (M.V.); (C.A.A.); (M.D.A.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (M.V.); (C.A.A.); (M.D.A.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy;
| | - Giovanni De Pergola
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| |
Collapse
|
111
|
Choy CT, Siu PLK, Zhou J, Wong CH, Lee YW, Chan HW, Tsui JCC, Lo CJY, Loo SKF, Tsui SKW. Improvements in Gut Microbiome Composition Predict the Clinical Efficacy of a Novel Synbiotics Formula in Children with Mild to Moderate Atopic Dermatitis. Microorganisms 2023; 11:2175. [PMID: 37764019 PMCID: PMC10536305 DOI: 10.3390/microorganisms11092175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/09/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disease with a significant association with various type-2 inflammation-related comorbidities. Ongoing research suggests the crucial involvement of gut microbiome, especially in childhood onset AD, and hence, probiotics have emerged as a potential non-steroid-based therapeutics option to complement existing AD management plans. In order to delineate the impact of probiotics in the gut microbiome of pediatric AD patients from southern China, targeted 16S rRNA sequencing and thorough bioinformatic analysis were performed to analyze the gut microbiome profiles of 24 AD children after taking an orally administered novel synbiotics formula with triple prebiotics for 8 weeks. A notable improvement in Eczema Area and Severity Index (EASI) (p = 0.008) was observed after taking an 8-week course of probiotics, with no adverse effects observed. The relative abundances of key microbial drivers including Bacteroides fragilis and Lactobacillus acidophilus were significantly increased at week 8. We also found that the positive responsiveness towards an 8-week course of probiotics was associated with improvements in the gut microbiome profile with a higher relative abundance of probiotic species. Over-represented functional abundance pathways related to vitamin B synthesis and peptidoglycan recycling may imply the underlying mechanism. In summary, our study suggests how the gut microbial landscape shifts upon probiotic supplementation in AD children, and provides preliminary evidence to support targeted probiotic supplementation for the management of childhood AD.
Collapse
Affiliation(s)
- Chi Tung Choy
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Pui Ling Kella Siu
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Junwei Zhou
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Chi Ho Wong
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Yuk Wai Lee
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Ho Wang Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | | | - Claudia Jun Yi Lo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Steven King Fan Loo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
- Hong Kong Institute of Integrative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
- Dermatology Centre, CUHK Medical Centre, The Chinese University of Hong Kong, Hong Kong
| | - Stephen Kwok Wing Tsui
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
- Centre for Microbial Genomics and Proteomics, The Chinese University of Hong Kong, Hong Kong
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
112
|
Hensen T, Fässler D, O’Mahony L, Albrich WC, Barda B, Garzoni C, Kleger GR, Pietsch U, Suh N, Hertel J, Thiele I. The Effects of Hospitalisation on the Serum Metabolome in COVID-19 Patients. Metabolites 2023; 13:951. [PMID: 37623894 PMCID: PMC10456321 DOI: 10.3390/metabo13080951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
COVID-19, a systemic multi-organ disease resulting from infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is known to result in a wide array of disease outcomes, ranging from asymptomatic to fatal. Despite persistent progress, there is a continued need for more accurate determinants of disease outcomes, including post-acute symptoms after COVID-19. In this study, we characterised the serum metabolomic changes due to hospitalisation and COVID-19 disease progression by mapping the serum metabolomic trajectories of 71 newly hospitalised moderate and severe patients in their first week after hospitalisation. These 71 patients were spread out over three hospitals in Switzerland, enabling us to meta-analyse the metabolomic trajectories and filter consistently changing metabolites. Additionally, we investigated differential metabolite-metabolite trajectories between fatal, severe, and moderate disease outcomes to find prognostic markers of disease severity. We found drastic changes in serum metabolite concentrations for 448 out of the 901 metabolites. These results included markers of hospitalisation, such as environmental exposures, dietary changes, and altered drug administration, but also possible markers of physiological functioning, including carboxyethyl-GABA and fibrinopeptides, which might be prognostic for worsening lung injury. Possible markers of disease progression included altered urea cycle metabolites and metabolites of the tricarboxylic acid (TCA) cycle, indicating a SARS-CoV-2-induced reprogramming of the host metabolism. Glycerophosphorylcholine was identified as a potential marker of disease severity. Taken together, this study describes the metabolome-wide changes due to hospitalisation and COVID-19 disease progression. Moreover, we propose a wide range of novel potential biomarkers for monitoring COVID-19 disease course, both dependent and independent of the severity.
Collapse
Affiliation(s)
- Tim Hensen
- School of Medicine, University of Galway, H91 TK33 Galway, Ireland;
- School of Microbiology, University of Galway, H91 TK33 Galway, Ireland
- Ryan Institute, University of Galway, H91 TK33 Galway, Ireland
- APC Microbiome Ireland, T12 K8AF Cork, Ireland; (L.O.); (W.C.A.)
| | - Daniel Fässler
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Liam O’Mahony
- APC Microbiome Ireland, T12 K8AF Cork, Ireland; (L.O.); (W.C.A.)
- Department of Medicine and School of Microbiology, University College Cork, T12 K8AF Cork, Ireland
| | - Werner C. Albrich
- APC Microbiome Ireland, T12 K8AF Cork, Ireland; (L.O.); (W.C.A.)
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Beatrice Barda
- Fondazione Epatocentro Ticino, Via Soldino 5, 6900 Lugano, Switzerland; (B.B.); (C.G.)
| | - Christian Garzoni
- Fondazione Epatocentro Ticino, Via Soldino 5, 6900 Lugano, Switzerland; (B.B.); (C.G.)
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, 6900 Lugano, Switzerland
| | - Gian-Reto Kleger
- Division of Intensive Care, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland;
| | - Urs Pietsch
- Department of Anesthesia, Intensive Care, Emergency and Pain Medicine, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland;
| | - Noémie Suh
- Division of Intensive Care, Geneva University Hospitals, The Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
| | - Johannes Hertel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany;
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Ines Thiele
- School of Medicine, University of Galway, H91 TK33 Galway, Ireland;
- School of Microbiology, University of Galway, H91 TK33 Galway, Ireland
- Ryan Institute, University of Galway, H91 TK33 Galway, Ireland
- APC Microbiome Ireland, T12 K8AF Cork, Ireland; (L.O.); (W.C.A.)
| |
Collapse
|
113
|
Huang D, Yang Y, Song W, Jiang C, Zhang Y, Zhang A, Lin Z, Ke X. Untargeted metabonomic analysis of a cerebral stroke model in rats: a study based on UPLC-MS/MS. Front Neurosci 2023; 17:1084813. [PMID: 37614341 PMCID: PMC10442664 DOI: 10.3389/fnins.2023.1084813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/31/2022] [Accepted: 07/18/2023] [Indexed: 08/25/2023] Open
Abstract
Introduction Brain tissue damage caused by ischemic stroke can trigger changes in the body's metabolic response, and understanding the changes in the metabolic response of the gut after stroke can contribute to research on poststroke brain function recovery. Despite the increase in international research on poststroke metabolic mechanisms and the availability of powerful research tools in recent years, there is still an urgent need for poststroke metabolic studies. Metabolomic examination of feces from a cerebral ischemia-reperfusion rat model can provide new insights into poststroke metabolism and identify key metabolic pathways, which will help reveal diagnostic and therapeutic targets as well as inspire pathophysiological studies after stroke. Methods We randomly divided 16 healthy adult pathogen-free male Sprague-Dawley (SD) rats into the normal group and the study group, which received middle cerebral artery occlusion/reperfusion (MCAO/R). Ultra-performance liquid chromatography-tandem mass spectrometry (UPLCMS/MS) was used to determine the identities and concentrations of metabolites across all groups, and filtered high-quality data were analyzed for differential screening and differential metabolite functional analysis. Results After 1 and 14 days of modeling, compared to the normal group, rats in the study group showed significant neurological deficits (p < 0.001) and significantly increased infarct volume (day 1: p < 0.001; day 14: p = 0.001). Mass spectra identified 1,044 and 635 differential metabolites in rat feces in positive and negative ion modes, respectively, which differed significantly between the normal and study groups. The metabolites with increased levels identified in the study group were involved in tryptophan metabolism (p = 0.036678, p < 0.05), arachidonic acid metabolism (p = 0.15695), cysteine and methionine metabolism (p = 0.24705), and pyrimidine metabolism (p = 0.3413), whereas the metabolites with decreased levels were involved in arginine and proline metabolism (p = 0.15695) and starch and sucrose metabolism (p = 0.52256). Discussion We determined that UPLC-MS/MS could be employed for untargeted metabolomics research. Moreover, tryptophan metabolic pathways may have been disordered in the study group. Alterations in the tryptophan metabolome may provide additional theoretical and data support for elucidating stroke pathogenesis and selecting pathways for intervention.
Collapse
Affiliation(s)
- Dunbing Huang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yihan Yang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wei Song
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cai Jiang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Second Rehabilitation Department, Fujian Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, China
- Fujian Key Laboratory of Geriatrics Diseases, Fujian Provincial Hospital, Fuzhou, China
- Department of Complementary Medicine, University of Johannesburg, Johannesburg, South Africa
| | - Yuhao Zhang
- Department of Rehabilitation Medicine, Nanjing Lishui District Hospital of Traditional Chinese medicine, Nanjing, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhonghua Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Second Rehabilitation Department, Fujian Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, China
- Fujian Key Laboratory of Geriatrics Diseases, Fujian Provincial Hospital, Fuzhou, China
- Department of Complementary Medicine, University of Johannesburg, Johannesburg, South Africa
| | - Xiaohua Ke
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
114
|
Amini Khiabani S, Asgharzadeh M, Samadi Kafil H. Chronic kidney disease and gut microbiota. Heliyon 2023; 9:e18991. [PMID: 37609403 PMCID: PMC10440536 DOI: 10.1016/j.heliyon.2023.e18991] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/17/2023] [Revised: 07/25/2023] [Accepted: 08/04/2023] [Indexed: 08/24/2023] Open
Abstract
Chronic kidney disease (CKD) refers to a range of various pathophysiological processes correlated with abnormal renal function and a progressive loss in GFR. Just as dysbiosis and altered pathology of the gut are accompanied with hypertension, which is a significant CKD risk factor. Gut dysbiosis in CKD patients is associated with an elevated levels of uremic toxins, which in turn increases the CKD progression. According to research results, the gut-kidney axis has a role in the formation of kidney stones, also in IgAN. A number of researchers have categorized the gut microbiota as enterotypes, and others, skeptical of theory of enterotypes, have suggested biomarkers to describe taxa that related to lifestyle, nutrition, and disease status. Metabolome-microbiome studies have been used to investigate the interactions of host-gut microbiota in terms of the involvement of metabolites in these interactions and are yielded promising results. The correlation between gut microbiota and CKD requires further multi-omic researches. Also, with regard to systems biology, studies on the communication network of proteins and transporters such as SLC and ABC, can help us achieve a deeper understanding of the gut-liver-kidney axis communication and can thus provide promising new horizons in the treatment of CKD patients. Probiotic-based treatment is an approach to reduce uremic poisoning, which is accomplished by swallowing microbes those can catalyze URS in the gut. If further comprehensive studies are carried out, we will know about the probiotics impact in slowing the renal failure progression and reducing inflammatory markers.
Collapse
Affiliation(s)
- Siamak Amini Khiabani
- Research center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Asgharzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
115
|
Gao W, Yang Y, Shi L. Seasonal dietary shifts alter the gut microbiota of a frugivorous lizard Teratoscincus roborowskii (Squamata, Sphaerodactylidae). Ecol Evol 2023; 13:e10363. [PMID: 37546566 PMCID: PMC10396791 DOI: 10.1002/ece3.10363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/11/2023] [Revised: 07/03/2023] [Accepted: 07/14/2023] [Indexed: 08/08/2023] Open
Abstract
Seasonal dietary shifts in animals are important strategies for ecological adaptation. An increasing number of studies have shown that seasonal dietary shifts can influence or even determine the composition of gut microbiota. The Turpan wonder gecko, Teratoscincus roborowskii, lives in extreme desert environments and has a flexible dietary shift to fruit-eating in warm seasons. However, the effect of such shifts on the gut microbiota is poorly understood. In this study, 16S rRNA sequencing and LC-MS metabolomics were used to examine changes in the gut microbiota composition and metabolic patterns of T. roborowskii. The results demonstrated that the gut microbes of T. roborowskii underwent significant seasonal changes, and the abundance of phylum level in autumn was significantly higher than spring, but meanwhile, the diversity was lower. At the family level, the abundance and diversity of the gut microbiota were both higher in autumn. Firmicutes, Bacteroidetes, and Proteobacteria were the dominant gut microbes of T. roborowskii. Verrucomicrobia and Proteobacteria exhibited dynamic ebb and flow patterns between spring and autumn. Metabolomic profiling also revealed differences mainly related to the formation of secondary bile acids. The pantothenate and CoA biosynthesis, and lysine degradation pathways identified by KEGG enrichment symbolize the exuberant metabolic capacity of T. roborowskii. Furthermore, strong correlations were detected between metabolite types and bacteria, and this correlation may be an important adaptation of T. roborowskii to cope with dietary shifts and improve energy acquisition. Our study provides a theoretical basis for exploring the adaptive evolution of the special frugivorous behavior of T. roborowskii, which is an important progress in the study of gut microbes in desert lizards.
Collapse
Affiliation(s)
- Wei‐Zhen Gao
- College of Life SciencesXinjiang Agricultural UniversityUrumqiChina
| | - Yi Yang
- College of Life SciencesXinjiang Agricultural UniversityUrumqiChina
| | - Lei Shi
- College of Life SciencesXinjiang Agricultural UniversityUrumqiChina
| |
Collapse
|
116
|
Vich Vila A, Hu S, Andreu-Sánchez S, Collij V, Jansen BH, Augustijn HE, Bolte LA, Ruigrok RAAA, Abu-Ali G, Giallourakis C, Schneider J, Parkinson J, Al-Garawi A, Zhernakova A, Gacesa R, Fu J, Weersma RK. Faecal metabolome and its determinants in inflammatory bowel disease. Gut 2023; 72:1472-1485. [PMID: 36958817 PMCID: PMC10359577 DOI: 10.1136/gutjnl-2022-328048] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/14/2022] [Accepted: 03/05/2023] [Indexed: 03/25/2023]
Abstract
OBJECTIVE Inflammatory bowel disease (IBD) is a multifactorial immune-mediated inflammatory disease of the intestine, comprising Crohn's disease and ulcerative colitis. By characterising metabolites in faeces, combined with faecal metagenomics, host genetics and clinical characteristics, we aimed to unravel metabolic alterations in IBD. DESIGN We measured 1684 different faecal metabolites and 8 short-chain and branched-chain fatty acids in stool samples of 424 patients with IBD and 255 non-IBD controls. Regression analyses were used to compare concentrations of metabolites between cases and controls and determine the relationship between metabolites and each participant's lifestyle, clinical characteristics and gut microbiota composition. Moreover, genome-wide association analysis was conducted on faecal metabolite levels. RESULTS We identified over 300 molecules that were differentially abundant in the faeces of patients with IBD. The ratio between a sphingolipid and L-urobilin could discriminate between IBD and non-IBD samples (AUC=0.85). We found changes in the bile acid pool in patients with dysbiotic microbial communities and a strong association between faecal metabolome and gut microbiota. For example, the abundance of Ruminococcus gnavus was positively associated with tryptamine levels. In addition, we found 158 associations between metabolites and dietary patterns, and polymorphisms near NAT2 strongly associated with coffee metabolism. CONCLUSION In this large-scale analysis, we identified alterations in the metabolome of patients with IBD that are independent of commonly overlooked confounders such as diet and surgical history. Considering the influence of the microbiome on faecal metabolites, our results pave the way for future interventions targeting intestinal inflammation.
Collapse
Affiliation(s)
- Arnau Vich Vila
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
| | - Shixian Hu
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre, Groningen, The Netherlands
| | - Valerie Collij
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
| | - Bernadien H Jansen
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
| | - Hannah E Augustijn
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
| | - Laura A Bolte
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
| | - Renate A A A Ruigrok
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
| | - Galeb Abu-Ali
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical, Cambridge, Massachusetts, USA
| | - Cosmas Giallourakis
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical, Cambridge, Massachusetts, USA
| | - Jessica Schneider
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical, Cambridge, Massachusetts, USA
| | - John Parkinson
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical, Cambridge, Massachusetts, USA
| | - Amal Al-Garawi
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical, Cambridge, Massachusetts, USA
| | | | - Ranko Gacesa
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Pediatrics, University Medical Centre, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre, Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Genetics, University Medical Centre, Groningen, The Netherlands
| |
Collapse
|
117
|
Zhou Y, Zhou K, Lin X, Wei Y, Ma B, Lu S, Xie G, Zhang Z, Liang J. Association of gut microbiota, plasma and fecal metabolite profiles with intellectual development in school-age children. Transl Pediatr 2023; 12:1292-1304. [PMID: 37575906 PMCID: PMC10416130 DOI: 10.21037/tp-22-610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/22/2022] [Accepted: 04/18/2023] [Indexed: 08/15/2023] Open
Abstract
Background Little is known about how the gut microbiota and metabolic profiles are related to cognitive outcomes in young children until now. It was hypothesized that the gut microbiota, the plasma and fecal metabolites significantly correlated with intelligence quotient (IQ) in school-age children in current study. Methods This cross-sectional study enrolled 452 children aged 6-9 years old. IQ was measured using the Wechsler Intelligence Scale for Children-Fourth Edition. Fecal microbiota, plasma and fecal metabolites were analyzed using 16S rRNA amplicon sequencing and targeted metabolomic technologies, respectively. Results Restricted maximum likelihood (REML) analyses showed that microbiota composition and fecal metabolites were associated with neither subscale nor full-scale IQ (P: 0.059-0.500). However, plasma metabolites were significantly correlated with the processing speed (P=0.008). In multiple regression analysis after adjusting for confounders and multiple test correction, benzoic acid, azelaic acid, adipic acid, suberic acid and malonic acid selected by the multivariate methods with unbiased variable selection were positively associated with processing speed index (PSI) [Pfalse discovery rate (FDR): 0.006-0.024], whereas pyruvic acid was negatively associated with the PSI and full-scale IQ (PFDR: 0.014-0.030). Conclusions In normal school-age children, certain plasma metabolites concentrations but not the gut microbiota composition nor fecal metabolites are correlated with intelligence.
Collapse
Affiliation(s)
- Yingyu Zhou
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Kejun Zhou
- Human Metabolomics Institute, Inc., Shenzhen, China
| | - Xiaoping Lin
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuanhuan Wei
- Department of Clinical Nutrition, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Bingjie Ma
- Department of Child Health Care, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Shaomin Lu
- Department of Child Health Care, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Guoxiang Xie
- Human Metabolomics Institute, Inc., Shenzhen, China
| | - Zheqing Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jingjing Liang
- Department of Child Health Care, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| |
Collapse
|
118
|
Wang YZ, Chen YY, Wu XZ, Bai PR, An N, Liu XL, Zhu QF, Feng YQ. Uncovering the Carboxylated Metabolome in Gut Microbiota-Host Co-metabolism: A Chemical Derivatization-Molecular Networking Approach. Anal Chem 2023. [PMID: 37471289 DOI: 10.1021/acs.analchem.3c02353] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 07/22/2023]
Abstract
Gut microbiota-host co-metabolites serve as essential mediators of communication between the host and gut microbiota. They provide nutrient sources for host cells and regulate gut microenvironment, which are associated with a variety of diseases. Analysis of gut microbiota-host co-metabolites is of great significance to explore the host-gut microbiota interaction. In this study, we integrated chemical derivatization, liquid chromatography-mass spectrometry, and molecular networking (MN) to establish a novel CD-MN strategy for the analysis of carboxylated metabolites in gut microbial-host co-metabolism. Using this strategy, 261 carboxylated metabolites from mouse feces were detected, which grouped to various classes including fatty acids, bile acids, N-acyl amino acids, benzoheterocyclic acids, aromatic acids, and other unknown small-scale molecular clusters in MN. Based on the interpretation of the bile acid cluster, a novel type of phenylacetylated conjugates of host bile acids was identified, which were mediated by gut microbiota and exhibited a strong binding ability to Farnesoid X receptor and Takeda G protein-coupled receptor 5. Our proposed strategy offers a promising platform for uncovering carboxylated metabolites in gut microbial-host co-metabolism.
Collapse
Affiliation(s)
- Yan-Zhen Wang
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Yao-Yu Chen
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xin-Ze Wu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Pei-Rong Bai
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Na An
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xia-Lei Liu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Quan-Fei Zhu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Yu-Qi Feng
- Department of Chemistry, Wuhan University, Wuhan 430072, China
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China
| |
Collapse
|
119
|
Hintikka JE, Ahtiainen JP, Permi P, Jalkanen S, Lehtonen M, Pekkala S. Aerobic exercise training and gut microbiome-associated metabolic shifts in women with overweight: a multi-omic study. Sci Rep 2023; 13:11228. [PMID: 37433843 DOI: 10.1038/s41598-023-38357-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/06/2023] [Accepted: 07/06/2023] [Indexed: 07/13/2023] Open
Abstract
Physical activity is essential in weight management, improves overall health, and mitigates obesity-related risk markers. Besides inducing changes in systemic metabolism, habitual exercise may improve gut's microbial diversity and increase the abundance of beneficial taxa in a correlated fashion. Since there is a lack of integrative omics studies on exercise and overweight populations, we studied the metabolomes and gut microbiota associated with programmed exercise in obese individuals. We measured the serum and fecal metabolites of 17 adult women with overweight during a 6-week endurance exercise program. Further, we integrated the exercise-responsive metabolites with variations in the gut microbiome and cardiorespiratory parameters. We found clear correlation with several serum and fecal metabolites, and metabolic pathways, during the exercise period in comparison to the control period, indicating increased lipid oxidation and oxidative stress. Especially, exercise caused co-occurring increase in levels of serum lyso-phosphatidylcholine moieties and fecal glycerophosphocholine. This signature was associated with several microbial metagenome pathways and the abundance of Akkermansia. The study demonstrates that, in the absence of body composition changes, aerobic exercise can induce metabolic shifts that provide substrates for beneficial gut microbiota in overweight individuals.
Collapse
Affiliation(s)
- Jukka E Hintikka
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Juha P Ahtiainen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Perttu Permi
- Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
- Department of Chemistry, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Sirpa Jalkanen
- MediCity and InFLAMES Flagship, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marko Lehtonen
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
120
|
Nordin E, Hellström PM, Dicksved J, Pelve E, Landberg R, Brunius C. Effects of FODMAPs and Gluten on Gut Microbiota and Their Association with the Metabolome in Irritable Bowel Syndrome: A Double-Blind, Randomized, Cross-Over Intervention Study. Nutrients 2023; 15:3045. [PMID: 37447371 DOI: 10.3390/nu15133045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND A mechanistic understanding of the effects of dietary treatment in irritable bowel syndrome (IBS) is lacking. Our aim was therefore to investigate how fermentable oligo- di-, monosaccharides, and polyols (FODMAPs) and gluten affected gut microbiota and circulating metabolite profiles, as well as to investigate potential links between gut microbiota, metabolites, and IBS symptoms. METHODS We used data from a double-blind, randomized, crossover study with week-long provocations of FODMAPs, gluten, and placebo in participants with IBS. To study the effects of the provocations on fecal microbiota, fecal and plasma short-chain fatty acids, the untargeted plasma metabolome, and IBS symptoms, we used Random Forest, linear mixed model and Spearman correlation analysis. RESULTS FODMAPs increased fecal saccharolytic bacteria, plasma phenolic-derived metabolites, 3-indolepropionate, and decreased isobutyrate and bile acids. Gluten decreased fecal isovalerate and altered carnitine derivatives, CoA, and fatty acids in plasma. For FODMAPs, modest correlations were observed between microbiota and phenolic-derived metabolites and 3-indolepropionate, previously associated with improved metabolic health, and reduced inflammation. Correlations between molecular data and IBS symptoms were weak. CONCLUSIONS FODMAPs, but not gluten, altered microbiota composition and correlated with phenolic-derived metabolites and 3-indolepropionate, with only weak associations with IBS symptoms. Thus, the minor effect of FODMAPs on IBS symptoms must be weighed against the effect on microbiota and metabolites related to positive health factors.
Collapse
Affiliation(s)
- Elise Nordin
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| | - Per M Hellström
- Department of Medical Sciences, Gastroenterology/Hepatology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Johan Dicksved
- Department of Animal Nutrition and Management, Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden
| | - Erik Pelve
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden
| | - Rikard Landberg
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| | - Carl Brunius
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| |
Collapse
|
121
|
Zhu G, Zhao J, Zhang H, Wang G, Chen W. Gut Microbiota and its Metabolites: Bridge of Dietary Nutrients and Alzheimer's Disease. Adv Nutr 2023; 14:819-839. [PMID: 37075947 PMCID: PMC10334159 DOI: 10.1016/j.advnut.2023.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/16/2023] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive impairment and neuroinflammation. Recent research has revealed the crucial role of gut microbiota and microbial metabolites in modulating AD. However, the mechanisms by which the microbiome and microbial metabolites affect brain function remain poorly understood. Here, we review the literature on changes in the diversity and composition of the gut microbiome in patients with AD and in animal models of AD. We also discuss the latest progress in understanding the pathways by which the gut microbiota and microbial metabolites from the host or diet regulate AD. By understanding the effects of dietary components on brain function, microbiota composition, and microbial metabolites, we examine the potential for manipulation of the gut microbiota through dietary intervention to delay the progression of AD. Although it is challenging to translate our understanding of microbiome-based approaches to dietary guidelines or clinical therapies, these findings provide an attractive target for promoting brain function.
Collapse
Affiliation(s)
- Guangsu Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; College of Food Science and Technology, Henan University of Technology, Zhengzhou, Henan, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China; National Engineering Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China; National Engineering Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
122
|
Huang Y, Jiao J, Yao D, Guo F, Li Y. Altered fecal microbiome and metabolome profiles in rat models of short bowel syndrome. Front Microbiol 2023; 14:1185463. [PMID: 37362931 PMCID: PMC10289890 DOI: 10.3389/fmicb.2023.1185463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/13/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction Short bowel syndrome (SBS) is featured by impaired nutrients and fluids absorption due to massive small intestine resection. Gut dysbiosis has been implicated in SBS, this study aimed to characterize the metagenomic and metabolomic profiles of SBS and identify potential therapeutic targets. Methods Fecal samples from SBS and Sham rats (n = 8 per group) were collected for high-throughput metagenomic sequencing. Fecal metabolomics was measured by untargeted liquid chromatography-mass spectrometry. Results We found that the species-level α-diversity significantly decreased in SBS rats, accompanied by altered microbiome compositions. The beneficial anaerobes from Firmicutes and Bacteroidetes were depleted while microorganisms from Lactobacillus, Escherichia, Enterococcus, and Streptococcus were enriched in faces from SBS rats. LEfSe analysis identified 17 microbial species and 38 KEGG modules that were remarkably distinct between SBS and Sham rats. In total, 1,577 metabolites with known chemical identity were detected from all samples, among them, 276 metabolites were down-regulated and 224 metabolites were up-regulated in SBS group. The typical signatures of SBS fecal metabolome comprised reduced short-chain fatty acids and products of amino acid metabolism (indole derivatives and p-cresol), as well as altered bile acid spectrum. We revealed 215 robust associations between representative differentially abundant microbial species and metabolites, the species with the same changing trend tended to have a similar correlation with some certain metabolites. Conclusion The fecal microbiome and metabolome significantly altered in SBS. Our findings may lay the foundation for developing new strategies to facilitate intestinal adaptation in SBS patients.
Collapse
Affiliation(s)
- Yuhua Huang
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jian Jiao
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Danhua Yao
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Feilong Guo
- Department of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Yousheng Li
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
123
|
Li J, Feng S, Wang Z, He J, Zhang Z, Zou H, Wu Z, Liu X, Wei H, Tao S. Limosilactobacillus mucosae-derived extracellular vesicles modulates macrophage phenotype and orchestrates gut homeostasis in a diarrheal piglet model. NPJ Biofilms Microbiomes 2023; 9:33. [PMID: 37280255 DOI: 10.1038/s41522-023-00403-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/03/2023] [Accepted: 05/22/2023] [Indexed: 06/08/2023] Open
Abstract
The diarrheal disease causes high mortality, especially in children and young animals. The gut microbiome is strongly associated with diarrheal disease, and some specific strains of bacteria have demonstrated antidiarrheal effects. However, the antidiarrheal mechanisms of probiotic strains have not been elucidated. Here, we used neonatal piglets as a translational model and found that gut microbiota dysbiosis observed in diarrheal piglets was mainly characterized by a deficiency of Lactobacillus, an abundance of Escherichia coli, and enriched lipopolysaccharide biosynthesis. Limosilactobacillus mucosae and Limosilactobacillus reuteri were a signature bacterium that differentiated healthy and diarrheal piglets. Germ-free (GF) mice transplanted with fecal microbiota from diarrheal piglets reproduced diarrheal disease symptoms. Administration of Limosilactobacillus mucosae but not Limosilactobacillus reuteri alleviated diarrheal disease symptoms induced by fecal microbiota of diarrheal piglets and by ETEC K88 challenge. Notably, Limosilactobacillus mucosae-derived extracellular vesicles alleviated diarrheal disease symptoms caused by ETEC K88 by regulating macrophage phenotypes. Macrophage elimination experiments demonstrated that the extracellular vesicles alleviated diarrheal disease symptoms in a macrophage-dependent manner. Our findings provide insights into the pathogenesis of diarrheal disease from the perspective of intestinal microbiota and the development of probiotic-based antidiarrheal therapeutic strategies.
Collapse
Affiliation(s)
- Jingjing Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shuaifei Feng
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhenyu Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Jinhui He
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zeyue Zhang
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huicong Zou
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhifeng Wu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiangdong Liu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Hong Wei
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
124
|
Anderson BG, Raskind A, Hissong R, Dougherty MK, McGill SK, Gulati A, Theriot CM, Kennedy RT, Evans CR. Offline Two-dimensional Liquid Chromatography-Mass Spectrometry for Deep Annotation of the Fecal Metabolome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.543178. [PMID: 37333153 PMCID: PMC10274728 DOI: 10.1101/2023.05.31.543178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/20/2023]
Abstract
Compound identification is an essential task in the workflow of untargeted metabolomics since the interpretation of the data in a biological context depends on the correct assignment of chemical identities to the features it contains. Current techniques fall short of identifying all or even most observable features in untargeted metabolomics data, even after rigorous data cleaning approaches to remove degenerate features are applied. Hence, new strategies are required to annotate the metabolome more deeply and accurately. The human fecal metabolome, which is the focus of substantial biomedical interest, is a more complex, more variable, yet lesser-investigated sample matrix compared to widely studied sample types like human plasma. This manuscript describes a novel experimental strategy using multidimensional chromatography to facilitate compound identification in untargeted metabolomics. Pooled fecal metabolite extract samples were fractionated using offline semi-preparative liquid chromatography. The resulting fractions were analyzed by an orthogonal LC-MS/MS method, and the data were searched against commercial, public, and local spectral libraries. Multidimensional chromatography yielded more than a 3-fold improvement in identified compounds compared to the typical single-dimensional LC-MS/MS approach and successfully identified several rare and novel compounds, including atypical conjugated bile acid species. Most features identified by the new approach could be matched to features that were detectable but not identifiable in the original single-dimension LC-MS data. Overall, our approach represents a powerful strategy for deeper annotation of the metabolome that can be implemented with commercially-available instrumentation, and should apply to any dataset requiring deeper annotation of the metabolome.
Collapse
|
125
|
Amin N, Liu J, Bonnechere B, MahmoudianDehkordi S, Arnold M, Batra R, Chiou YJ, Fernandes M, Ikram MA, Kraaij R, Krumsiek J, Newby D, Nho K, Radjabzadeh D, Saykin AJ, Shi L, Sproviero W, Winchester L, Yang Y, Nevado-Holgado AJ, Kastenmüller G, Kaddurah-Daouk R, van Duijn CM. Interplay of Metabolome and Gut Microbiome in Individuals With Major Depressive Disorder vs Control Individuals. JAMA Psychiatry 2023; 80:597-609. [PMID: 37074710 PMCID: PMC10116384 DOI: 10.1001/jamapsychiatry.2023.0685] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/01/2022] [Accepted: 02/07/2023] [Indexed: 04/20/2023]
Abstract
Importance Metabolomics reflect the net effect of genetic and environmental influences and thus provide a comprehensive approach to evaluating the pathogenesis of complex diseases, such as depression. Objective To identify the metabolic signatures of major depressive disorder (MDD), elucidate the direction of associations using mendelian randomization, and evaluate the interplay of the human gut microbiome and metabolome in the development of MDD. Design, Setting and Participants This cohort study used data from participants in the UK Biobank cohort (n = 500 000; aged 37 to 73 years; recruited from 2006 to 2010) whose blood was profiled for metabolomics. Replication was sought in the PREDICT and BBMRI-NL studies. Publicly available summary statistics from a 2019 genome-wide association study of depression were used for the mendelian randomization (individuals with MDD = 59 851; control individuals = 113 154). Summary statistics for the metabolites were obtained from OpenGWAS in MRbase (n = 118 000). To evaluate the interplay of the metabolome and the gut microbiome in the pathogenesis of depression, metabolic signatures of the gut microbiome were obtained from a 2019 study performed in Dutch cohorts. Data were analyzed from March to December 2021. Main Outcomes and Measures Outcomes were lifetime and recurrent MDD, with 249 metabolites profiled with nuclear magnetic resonance spectroscopy with the Nightingale platform. Results The study included 6811 individuals with lifetime MDD compared with 51 446 control individuals and 4370 individuals with recurrent MDD compared with 62 508 control individuals. Individuals with lifetime MDD were younger (median [IQR] age, 56 [49-62] years vs 58 [51-64] years) and more often female (4447 [65%] vs 2364 [35%]) than control individuals. Metabolic signatures of MDD consisted of 124 metabolites spanning the energy and lipid metabolism pathways. Novel findings included 49 metabolites, including those involved in the tricarboxylic acid cycle (ie, citrate and pyruvate). Citrate was significantly decreased (β [SE], -0.07 [0.02]; FDR = 4 × 10-04) and pyruvate was significantly increased (β [SE], 0.04 [0.02]; FDR = 0.02) in individuals with MDD. Changes observed in these metabolites, particularly lipoproteins, were consistent with the differential composition of gut microbiota belonging to the order Clostridiales and the phyla Proteobacteria/Pseudomonadota and Bacteroidetes/Bacteroidota. Mendelian randomization suggested that fatty acids and intermediate and very large density lipoproteins changed in association with the disease process but high-density lipoproteins and the metabolites in the tricarboxylic acid cycle did not. Conclusions and Relevance The study findings showed that energy metabolism was disturbed in individuals with MDD and that the interplay of the gut microbiome and blood metabolome may play a role in lipid metabolism in individuals with MDD.
Collapse
Affiliation(s)
- Najaf Amin
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Jun Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Bruno Bonnechere
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- REVAL Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, Hasselt, Belgium
- Technology-Supported and Data-Driven Rehabilitation, Data Sciences Institute, Hasselt University, Hasselt, Belgium
| | | | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Richa Batra
- Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Yu-Jie Chiou
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Marco Fernandes
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jan Krumsiek
- Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Danielle Newby
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis
| | - Djawad Radjabzadeh
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Andrew J. Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - William Sproviero
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Laura Winchester
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Yang Yang
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | | | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina
| | - Cornelia M. van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
126
|
Correia GD, Marchesi JR, MacIntyre DA. Moving beyond DNA: towards functional analysis of the vaginal microbiome by non-sequencing-based methods. Curr Opin Microbiol 2023; 73:102292. [PMID: 36931094 DOI: 10.1016/j.mib.2023.102292] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 03/17/2023]
Abstract
Over the last two decades, sequencing-based methods have revolutionised our understanding of niche-specific microbial complexity. In the lower female reproductive tract, these approaches have enabled identification of bacterial compositional structures associated with health and disease. Application of metagenomics and metatranscriptomics strategies have provided insight into the putative function of these communities but it is increasingly clear that direct measures of microbial and host cell function are required to understand the contribution of microbe-host interactions to pathophysiology. Here we explore and discuss current methods and approaches, many of which rely upon mass-spectrometry, being used to capture functional insight into the vaginal mucosal interface. In addition to improving mechanistic understanding, these methods offer innovative solutions for the development of diagnostic and therapeutic strategies designed to improve women's health.
Collapse
Affiliation(s)
- Gonçalo Ds Correia
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK; March of Dimes Prematurity Research Centre at Imperial College London, London, UK
| | - Julian R Marchesi
- March of Dimes Prematurity Research Centre at Imperial College London, London, UK; Centre for Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, Imperial College London, London W2 1NY, UK
| | - David A MacIntyre
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK; March of Dimes Prematurity Research Centre at Imperial College London, London, UK.
| |
Collapse
|
127
|
Wang J, Dong P, Zheng S, Mai Y, Ding J, Pan P, Tang L, Wan Y, Liang H. Advances in gut microbiome in metabonomics perspective: based on bibliometrics methods and visualization analysis. Front Cell Infect Microbiol 2023; 13:1196967. [PMID: 37325519 PMCID: PMC10266355 DOI: 10.3389/fcimb.2023.1196967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Background and aims Gastrointestinal microbial metabolomics is closely related to the state of the organism and has significant interaction with the pathogenesis of many diseases. Based on the publications in Web of Science Core Collection(WoSCC) from 2004 to 2022, this study conducted a bibliometric analysis of this field, aiming to understand its development trend and frontier, and provide basic information and potential points for in-depth exploration of this field. Methods All articles on gastrointestinal flora and metabolism published from 2004 to 2022 were collected and identified in WoCSS. CiteSpace v.6.1 and VOSviewer v.1.6.15.0 were used to calculate bibliometric indicators, including number of publications and citations, study categories, countries/institutions, authors/co-cited authors, journals/co-cited journals, co-cited references, and keywords. A map was drawn to visualize the data based on the analysis results for a more intuitive view. Results There were 3811 articles in WoSCC that met our criteria. Analysis results show that the number of publications and citations in this field are increasing year by year. China is the country with the highest number of publications and USA owns the highest total link strength and citations. Chinese Acad Sci rank first for the number of institutional publications and total link strength. Journal of Proteome Research has the most publications. Nicholson, Jeremy K. is one of the most important scholars in this field. The most cited reference is "Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease". Burst detection indicates that Urine, spectroscopy, metabonomic and gut microflora are long-standing hot topics in this field, while autism spectrum disorder and omics are likely to be at the forefront of research. The study of related metabolic small molecules and the application of gastrointestinal microbiome metabolomics in various diseases are currently emerging research directions and frontier in this field. Conclusion This study is the first to make a bibliometric analysis of the studies related to gastrointestinal microbial metabolomics and reveal the development trends and current research hotspots in this field. This can contribute to the development of the field by providing relevant scholars with valuable and effective information about the current state of the field.
Collapse
Affiliation(s)
- Jieyan Wang
- Department of Urology, The People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzen, China
| | - Peng Dong
- College of Anesthesiology, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shuqian Zheng
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yiyin Mai
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jianan Ding
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Pinfei Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Liugang Tang
- Tendon and Injury Department, Sichuan Provincial Orthopedics Hospital, Chengdu, China
| | - Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou, China
| | - Hui Liang
- Department of Urology, The People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzen, China
| |
Collapse
|
128
|
Kaur A, Lin W, Dovhalyuk V, Driutti L, Di Martino ML, Vujasinovic M, Löhr JM, Sellin ME, Globisch D. Chemoselective bicyclobutane-based mass spectrometric detection of biological thiols uncovers human and bacterial metabolites. Chem Sci 2023; 14:5291-5301. [PMID: 37234898 PMCID: PMC10207876 DOI: 10.1039/d3sc00224a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2023] [Accepted: 04/05/2023] [Indexed: 05/28/2023] Open
Abstract
Sulfur is an essential element of life. Thiol-containing metabolites in all organisms are involved in the regulation of diverse biological processes. Especially, the microbiome produces bioactive metabolites or biological intermediates of this compound class. The analysis of thiol-containing metabolites is challenging due to the lack of specific tools, making these compounds difficult to investigate selectively. We have now developed a new methodology comprising bicyclobutane for chemoselective and irreversible capturing of this metabolite class. We utilized this new chemical biology tool immobilized onto magnetic beads for the investigation of human plasma, fecal samples, and bacterial cultures. Our mass spectrometric investigation detected a broad range of human, dietary and bacterial thiol-containing metabolites and we even captured the reactive sulfur species cysteine persulfide in both fecal and bacterial samples. The described comprehensive methodology represents a new mass spectrometric strategy for the discovery of bioactive thiol-containing metabolites in humans and the microbiome.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University 75124 Uppsala Sweden
| | - Weifeng Lin
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University 75124 Uppsala Sweden
| | - Vladyslav Dovhalyuk
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University 75124 Uppsala Sweden
| | - Léna Driutti
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University 75124 Uppsala Sweden
| | - Maria Letizia Di Martino
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University 75123 Uppsala Sweden
| | - Miroslav Vujasinovic
- Department for Digestive Diseases, Karolinska University Hospital Stockholm Sweden
| | - J-Matthias Löhr
- Department for Digestive Diseases, Karolinska University Hospital Stockholm Sweden
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute Stockholm Sweden
| | - Mikael E Sellin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University 75123 Uppsala Sweden
| | - Daniel Globisch
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University 75124 Uppsala Sweden
| |
Collapse
|
129
|
Wang AJ, Song D, Hong YM, Liu NN. Multi-omics insights into the interplay between gut microbiota and colorectal cancer in the "microworld" age. Mol Omics 2023; 19:283-296. [PMID: 36916422 DOI: 10.1039/d2mo00288d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/18/2023]
Abstract
Colorectal cancer (CRC) is a multifactorial heterogeneous disease largely due to both genetic predisposition and environmental factors including the gut microbiota, a dynamic microbial ecosystem inhabiting the gastrointestinal tract. Elucidation of the molecular mechanisms by which the gut microbiota interacts with the host may contribute to the pathogenesis, diagnosis, and promotion of CRC. However, deciphering the influence of genetic variants and interactions with the gut microbial ecosystem is rather challenging. Despite recent advancements in single omics analysis, the application of multi-omics approaches to integrate multiple layers of information in the microbiome and host to introduce effective prevention, diagnosis, and treatment strategies is still in its infancy. Here, we integrate host- and microbe-based multi-omics studies, respectively, to provide a strategy to explore potential causal relationships between gut microbiota and colorectal cancer. Specifically, we summarize the recent multi-omics studies such as metagenomics combined with metabolomics and metagenomics combined with genomics. Meanwhile, the sample size and sample types commonly used in multi-omics research, as well as the methods of data analysis, were also generalized. We highlight multiple layers of information from multi-omics that need to be verified by different types of models. Together, this review provides new insights into the clinical diagnosis and treatment of colorectal cancer patients.
Collapse
Affiliation(s)
- An-Jun Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Dingka Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Yue-Mei Hong
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Ning-Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| |
Collapse
|
130
|
Radford-Smith DE, Anthony DC. Prebiotic and Probiotic Modulation of the Microbiota-Gut-Brain Axis in Depression. Nutrients 2023; 15:nu15081880. [PMID: 37111100 PMCID: PMC10146605 DOI: 10.3390/nu15081880] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/26/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Emerging evidence demonstrates that alterations to the gut microbiota can affect mood, suggesting that the microbiota-gut-brain (MGB) axis contributes to the pathogenesis of depression. Many of these pathways overlap with the way in which the gut microbiota are thought to contribute to metabolic disease progression and obesity. In rodents, prebiotics and probiotics have been shown to modulate the composition and function of the gut microbiota. Together with germ-free rodent models, probiotics have provided compelling evidence for a causal relationship between microbes, microbial metabolites, and altered neurochemical signalling and inflammatory pathways in the brain. In humans, probiotic supplementation has demonstrated modest antidepressant effects in individuals with depressive symptoms, though more studies in clinically relevant populations are needed. This review critically discusses the role of the MGB axis in depression pathophysiology, integrating preclinical and clinical evidence, as well as the putative routes of communication between the microbiota-gut interface and the brain. A critical overview of the current approaches to investigating microbiome changes in depression is provided. To effectively translate preclinical breakthroughs in MGB axis research into novel therapies, rigorous placebo-controlled trials alongside a mechanistic and biochemical understanding of prebiotic and probiotic action are required from future research.
Collapse
Affiliation(s)
- Daniel E Radford-Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX3 7JX, UK
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
131
|
Li M, Zhang Z, Yu B, Jia S, Cui B. Lycium barbarum Oligosaccharides Alleviate Hepatic Steatosis by Modulating Gut Microbiota in C57BL/6J Mice Fed a High-Fat Diet. Foods 2023; 12:foods12081617. [PMID: 37107413 PMCID: PMC10138177 DOI: 10.3390/foods12081617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/21/2023] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
High-fat diets (HFD) can promote the development of hepatic steatosis by altering the structure and composition of gut flora. In this study, the potential therapeutic mechanism of Lycium barbarum oligosaccharide (LBO) against hepatic steatosis was investigated by analyzing the changes in the intestinal flora and metabolites in mice. Mice on an HFD were administered LBO by gavage once daily for a continuous period of eight weeks. Compared with the HFD group, the levels of triglyceride (TG), alanine aminotransferase (ALT) in the serum, and hepatic TG were significantly reduced in the LBO group, and liver lipid accumulation was obviously improved. In addition, LBO could regulate the HFD-induced alteration of intestinal flora. The HFD increased the proportion of Barnesiellaceae, Barnesiella, and CHKCI001. LBO increased the proportion of Dubosiella, Eubacterium, and Lactobacillus. LBO also altered the fecal metabolic profile. Significantly different metabolites between LBO and the HFD, such as taurochenodeoxycholate, taurocholate, fluvastatin, and kynurenic acid, were related to the cholesterol metabolism, bile acid metabolism, and tryptophan metabolic pathways. In light of the above, LBO can alleviate HFD-induced NAFLD by modulating the components of the intestinal flora and fecal metabolites.
Collapse
Affiliation(s)
- Mengjie Li
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
- School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Daxue Road, Changqing District, Jinan 250353, China
| | - Zheng Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
- School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Daxue Road, Changqing District, Jinan 250353, China
| | - Bin Yu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
- School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Daxue Road, Changqing District, Jinan 250353, China
| | - Siqiang Jia
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
- School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Daxue Road, Changqing District, Jinan 250353, China
| | - Bo Cui
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
- School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Daxue Road, Changqing District, Jinan 250353, China
| |
Collapse
|
132
|
Yan Z, Xu Q, Yao Y, Ayala J, Hou R, Wang H. Fecal Metabolomics Reveals the Foraging Strategies of Giant Pandas for Different Parts of Bamboo. Animals (Basel) 2023; 13:ani13081278. [PMID: 37106841 PMCID: PMC10135075 DOI: 10.3390/ani13081278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/15/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Climate change-induced food shortages pose major threats to wildlife conservation, and the exclusive reliance of giant pandas on bamboo makes them particularly vulnerable. The aim of this study was to provide insight into the reasons for the foraging strategies of giant pandas to selectively forage for different bamboo parts (bamboo shoot, culm, and leaf) during different seasons. This study used a metabolomic approach to analyze the fecal metabolites of giant pandas and conducted a correlation analysis with their gut microbiota. The results indicate that the fecal metabolites of giant pandas differ significantly depending on the bamboo parts they forage on, with higher sugar content observed when they consume bamboo culm with high fiber content. By functional annotation, culm group metabolites were enriched in the galactose metabolic pathway, while shoot group metabolites were enriched in the phenylalanine, tyrosine and tryptophan biosynthesis pathways. Moreover, Streptococcus showed a significant positive correlation with glucose and acetic acid content. Therefore, the foraging strategy of giant pandas is based on the ability to utilize the nutrient content of different bamboo parts. Captive feeding and habitat construction should enrich bamboo species to allow them to express their natural foraging strategies and improve their welfare and reproductive status.
Collapse
Affiliation(s)
- Zheng Yan
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
- Sichuan Academy of Giant Panda, Chengdu 610081, China
| | - Qin Xu
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
- Sichuan Academy of Giant Panda, Chengdu 610081, China
| | - Ying Yao
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
- Sichuan Academy of Giant Panda, Chengdu 610081, China
| | - James Ayala
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
- Sichuan Academy of Giant Panda, Chengdu 610081, China
| | - Rong Hou
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
- Sichuan Academy of Giant Panda, Chengdu 610081, China
| | - Hairui Wang
- Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu 610081, China
- Sichuan Academy of Giant Panda, Chengdu 610081, China
| |
Collapse
|
133
|
Jiang W, Chen J, Gong L, Liu F, Zhao H, Yan Z, Li Y, Zhang J, Xiao M, Mu J. Microbiota-derived short-chain fatty acids may participate in post-stroke depression by regulating host's lipid metabolism. J Psychiatr Res 2023; 161:426-434. [PMID: 37031497 DOI: 10.1016/j.jpsychires.2023.03.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/25/2022] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND Post-stroke depression (PSD) is a common mental disorder of stroke survivors, its pathogenesis remains elusive. Previous studies suggested a role of the microbiota-gut-brain (MGB) axis in stroke and depression. In this study, we characterized microbial composition and function, and gut-brain metabolic signatures, in PSD rats. We aim to explore how disordered gut microbes participate in the pathogenesis of PSD through the MGB axis. MATERIALS AND METHODS 16S rRNA gene sequence and fecal metabolome analysis were performed to identify the gut microbiome and their functional metabolites in PSD rats. Then, the lipid metabolic signatures in the prefrontal cortex (PFC) of PSD were conducted by liquid chromatography mass spectrometry. Finally, the potential pathway between gut and brain in the onset of PSD were explored. RESULTS Compared to control and stroke rats, there were 10 genera (most of them belonged to phylum Firmicutes) significantly changed and 3 short chain fatty acids (SCFAs: butyric acid, acetic acid and pentanoic acid) significantly decreased in PSD rats. Meanwhile, altered gut microbial in PSD rats was significantly associated with these SCFAs. Compared with control and stroke rats, 57 lipid metabolites in the PFC of PSD rats were significantly changed. In addition, the altered SCFAs in PSD rats were also significantly correlated with most of disordered lipid metabolites in PFC. CONCLUSIONS Our findings suggest that the SCFAs may be a bridge of gut-brain communication. The Firmicutes-SCFAs-lipid metabolism might be a potential pathway to further investigate the MGB axis and pathogenesis of PSD.
Collapse
Affiliation(s)
- Wenxia Jiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Jianjun Chen
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Lei Gong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Fang Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Huan Zhao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Zhiwen Yan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Yingli Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Jie Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Mi Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China
| | - Jun Mu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, No.1 Yixueyuan Road, Chongqing, China.
| |
Collapse
|
134
|
Munir SS, Sert Kuniyoshi FH, Singh P, Covassin N. Is the Gut Microbiome Implicated in the Excess Risk of Hypertension Associated with Obstructive Sleep Apnea? A Contemporary Review. Antioxidants (Basel) 2023; 12:antiox12040866. [PMID: 37107242 PMCID: PMC10135363 DOI: 10.3390/antiox12040866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a highly prevalent sleep disorder and an established risk factor for cardiovascular diseases, including hypertension. The pathogenesis of elevated blood pressure (BP) in OSA is multifactorial, including sympathetic overdrive, vascular aberrations, oxidative stress, inflammation, and metabolic dysregulation. Among the mechanisms potentially involved in OSA-induced hypertension, the role of the gut microbiome is gaining increasing attention. Perturbations in the diversity, composition, and function of the gut microbiota have been causally linked to numerous disorders, and robust evidence has identified gut dysbiosis as a determinant of BP elevation in various populations. In this brief review, we summarize the current body of literature on the implications of altered gut microbiota for hypertension risk in OSA. Data from both preclinical models of OSA and patient populations are presented, and potential mechanistic pathways are highlighted, along with therapeutic considerations. Available evidence suggests that gut dysbiosis may promote the development of hypertension in OSA and may thus be a target for interventions aimed at attenuating the adverse consequences of OSA in relation to cardiovascular risk.
Collapse
Affiliation(s)
- Sanah S. Munir
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, MN 55905, USA
| | - Fatima H. Sert Kuniyoshi
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, MN 55905, USA
- ResMed Science Center, San Diego, CA 92123, USA
| | - Prachi Singh
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Naima Covassin
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, MN 55905, USA
| |
Collapse
|
135
|
Murali A, Giri V, Zickgraf FM, Ternes P, Cameron HJ, Sperber S, Haake V, Driemert P, Kamp H, Funk-Weyer D, Sturla SJ, Rietjens IMCM, van Ravenzwaay B. Connecting Gut Microbial Diversity with Plasma Metabolome and Fecal Bile Acid Changes Induced by the Antibiotics Tobramycin and Colistin Sulfate. Chem Res Toxicol 2023; 36:598-616. [PMID: 36972423 DOI: 10.1021/acs.chemrestox.2c00316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 03/29/2023]
Abstract
The diversity of microbial species in the gut has a strong influence on health and development of the host. Further, there are indications that the variation in expression of gut bacterial metabolic enzymes is less diverse than the taxonomic profile, underlying the importance of microbiome functionality, particularly from a toxicological perspective. To address these relationships, the gut bacterial composition of Wistar rats was altered by a 28 day oral treatment with the antibiotics tobramycin or colistin sulfate. On the basis of 16S marker gene sequencing data, tobramycin was found to cause a strong reduction in the diversity and relative abundance of the microbiome, whereas colistin sulfate had only a marginal impact. Associated plasma and fecal metabolomes were characterized by targeted mass spectrometry-based profiling. The fecal metabolome of tobramycin-treated animals had a high number of significant alterations in metabolite levels compared to controls, particularly in amino acids, lipids, bile acids (BAs), carbohydrates, and energy metabolites. The accumulation of primary BAs and significant reduction of secondary BAs in the feces indicated that the microbial alterations induced by tobramycin inhibit bacterial deconjugation reactions. The plasma metabolome showed less, but still many alterations in the same metabolite groups, including reductions in indole derivatives and hippuric acid, and furthermore, despite marginal effects of colistin sulfate treatment, there were nonetheless systemic alterations also in BAs. Aside from these treatment-based differences, we also uncovered interindividual differences particularly centering on the loss of Verrucomicrobiaceae in the microbiome, but with no apparent associated metabolite alterations. Finally, by comparing the data set from this study with metabolome alterations in the MetaMapTox database, key metabolite alterations were identified as plasma biomarkers indicative of altered gut microbiomes resulting from a wide activity spectrum of antibiotics.
Collapse
Affiliation(s)
| | - Varun Giri
- BASF SE, Ludwigshafen am Rhein 67056, Rheinland-Pfalz, Germany
| | | | - Philipp Ternes
- Metanomics (BASF Metabolome Solutions) GmbH, Tegeler Weg 33, Berlin 10589, Germany
| | - Hunter James Cameron
- BASF Corporation Computational Biology (RTP), Research Triangle Park, 3500 Paramount Parkway, Morrisvile, North Carolina 27560, United States
| | - Saskia Sperber
- BASF SE, Ludwigshafen am Rhein 67056, Rheinland-Pfalz, Germany
| | - Volker Haake
- Metanomics (BASF Metabolome Solutions) GmbH, Tegeler Weg 33, Berlin 10589, Germany
| | - Peter Driemert
- Metanomics (BASF Metabolome Solutions) GmbH, Tegeler Weg 33, Berlin 10589, Germany
| | - Hennicke Kamp
- Metanomics (BASF Metabolome Solutions) GmbH, Tegeler Weg 33, Berlin 10589, Germany
| | | | - Shana J Sturla
- Department of Health Sciences and Technology, ETH Zürich, Schmelzbergstrasse 9, Zurich CH 8092, Switzerland
| | | | | |
Collapse
|
136
|
Li Y, Li J, Cheng R, Liu H, Zhao Y, Liu Y, Chen Y, Sun Z, Zhai Z, Wu M, Yan Y, Sun Y, Zhang Z. Alteration of the gut microbiome and correlated metabolism in a rat model of long-term depression. Front Cell Infect Microbiol 2023; 13:1116277. [PMID: 37051300 PMCID: PMC10084793 DOI: 10.3389/fcimb.2023.1116277] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/05/2022] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
ObjectiveThis study aims to investigate the composition and function of the gut microbiome in long-term depression using an 8-week chronic unpredictable mild stress (CUMS) rat model.Materials and methodsAnimals were sacrificed after either 4 weeks or 8 weeks under CUMS to mimic long-term depression in humans. The gut microbiome was analyzed to identify potential depression-related gut microbes, and the fecal metabolome was analyzed to detect their functional metabolites. The correlations between altered gut microbes and metabolites in the long-term depression rats were explored. The crucial metabolic pathways related to long-term depression were uncovered through enrichment analysis based on these gut microbes and metabolites.ResultsThe microbial composition of long-term depression (8-week CUMS) showed decreased species richness indices and different profiles compared with the control group and the 4-week CUMS group, characterized by disturbance of Alistipes indistinctus, Bacteroides ovatus, and Alistipes senegalensis at the species level. Additionally, long-term depression was associated with disturbances in fecal metabolomics. D-pinitol was the only increased metabolite in the 8-week CUMS group among the top 10 differential metabolites, while the top 3 decreased metabolites in the long-term depression rats included indoxyl sulfate, trimethylaminen-oxide, and 3 alpha,7 alpha-dihydroxy-12-oxocholanoic acid. The disordered fecal metabolomics in the long-term depression rats mainly involved the biosynthesis of pantothenate, CoA, valine, leucine and isoleucine.ConclusionOur findings suggest that the gut microbiome may participate in the long-term development of depression, and the mechanism may be related to the regulation of gut metabolism.
Collapse
Affiliation(s)
- Yubo Li
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yubo Li, ; Yuxiu Sun, ; Zhiguo Zhang,
| | - Junling Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ran Cheng
- Department of Gynaecology and Obstetrics, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Haixia Liu
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yukun Zhao
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanjun Liu
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanjing Chen
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhibo Sun
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiguang Zhai
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meng Wu
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yupeng Yan
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxiu Sun
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yubo Li, ; Yuxiu Sun, ; Zhiguo Zhang,
| | - Zhiguo Zhang
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yubo Li, ; Yuxiu Sun, ; Zhiguo Zhang,
| |
Collapse
|
137
|
Yu X, Dong J, Xiang S, Li X, Qin Y, Zhu S, Zheng J, Yan Y, Mi J. Association of intestinal microbiota and its metabolite markers with excess weight in Chinese children and adolescents. Pediatr Obes 2023; 18:e13019. [PMID: 36878714 DOI: 10.1111/ijpo.13019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/29/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023]
Abstract
OBJECTIVE The objective of this study was to identify the intestinal microbiota and faecal metabolic biomarkers associated with excess weight in Chinese children and adolescents. METHODS This cross-sectional study included 163 children aged 6-14 years (including 72 with normal-weight and 91 with overweight/obesity from three Chinese boarding schools). We used 16S rRNA high-throughput sequencing to analyse the diversity and composition of intestinal microbiota. Of these participants, we selected 10 children with normal-weight and 10 with obesity (matched 1:1 for school, sex and age) and measured faecal metabolites using ultra-performance liquid chromatography coupled with tandem mass spectrometry. RESULTS Alpha diversity was significantly elevated in children with normal-weight compared to overweight/obese. Principle coordinate analysis and permutational multivariate analysis of variance revealed a significant difference in intestinal microbial community structure between the normal-weight and overweight/obese groups. The two groups differed significantly in the relative abundances of Megamonas, Bifidobacterium and Alistipes. In faecal metabolomics analysis, we identified 14 differential metabolites and 2 main differential metabolic pathways associated with obesity. CONCLUSION This study identified intestinal microbiota and metabolic markers associated with excess weight in Chinese children.
Collapse
Affiliation(s)
- Xiaoran Yu
- Center for Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jie Dong
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, China
| | - Shiting Xiang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, China
| | - Xun Li
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, China
| | | | - Shaolun Zhu
- The Middle School of Fengshu, Taoyuan, China
| | - Jie Zheng
- The Primary School of Qinglin, Taoyuan, China
| | - Yinkun Yan
- Center for Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jie Mi
- Center for Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
138
|
Li Z, Dong Y, Zhang Y, Zheng M, Jiang Z, Zhu Y, Deng S, Li Q, Ni H. Lactobacillus-fermentation enhances nutritional value and improves the inhibition on pancreatic lipase and oral pathogens of edible red seaweed Bangia fusco-purpurea. Lebensm Wiss Technol 2023. [DOI: 10.1016/j.lwt.2023.114643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 03/17/2023]
|
139
|
Valles-Colomer M, Menni C, Berry SE, Valdes AM, Spector TD, Segata N. Cardiometabolic health, diet and the gut microbiome: a meta-omics perspective. Nat Med 2023; 29:551-561. [PMID: 36932240 PMCID: PMC11258867 DOI: 10.1038/s41591-023-02260-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2022] [Accepted: 02/16/2023] [Indexed: 03/19/2023]
Abstract
Cardiometabolic diseases have become a leading cause of morbidity and mortality globally. They have been tightly linked to microbiome taxonomic and functional composition, with diet possibly mediating some of the associations described. Both the microbiome and diet are modifiable, which opens the way for novel therapeutic strategies. High-throughput omics techniques applied on microbiome samples (meta-omics) hold the unprecedented potential to shed light on the intricate links between diet, the microbiome, the metabolome and cardiometabolic health, with a top-down approach. However, effective integration of complementary meta-omic techniques is an open challenge and their application on large cohorts is still limited. Here we review meta-omics techniques and discuss their potential in this context, highlighting recent large-scale efforts and the novel insights they provided. Finally, we look to the next decade of meta-omics research and discuss various translational and clinical pathways to improving cardiometabolic health.
Collapse
Affiliation(s)
- Mireia Valles-Colomer
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Cristina Menni
- Department of Twin Research, King's College London, London, UK
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | - Ana M Valdes
- School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham National Institute for Health Research Biomedical Research Centre, Nottingham, UK
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.
- European Institute of Oncology, Scientific Institute for Research, Hospitalization and Healthcare, Milan, Italy.
| |
Collapse
|
140
|
Wei J, Dai W, Pan X, Zhong Y, Xu N, Ye P, Wang J, Li J, Yang F, Luo J, Luo M. Identifying the Novel Gut Microbial Metabolite Contributing to Metabolic Syndrome in Children Based on Integrative Analyses of Microbiome-Metabolome Signatures. Microbiol Spectr 2023; 11:e0377122. [PMID: 36794949 PMCID: PMC10101147 DOI: 10.1128/spectrum.03771-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/15/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
The pathogenesis of gut microbiota and their metabolites in the development of metabolic syndrome (MS) remains unclear. This study aimed to evaluate the signatures of gut microbiota and metabolites as well as their functions in obese children with MS. A case-control study was conducted based on 23 MS children and 31 obese controls. The gut microbiome and metabolome were measured using 16S rRNA gene amplicon sequencing and liquid chromatography-mass spectrometry. An integrative analysis was conducted, combining the results of the gut microbiome and metabolome with extensive clinical indicators. The biological functions of the candidate microbial metabolites were validated in vitro. We identified 9 microbiota and 26 metabolites that were significantly different from the MS and the control group. The clinical indicators of MS were correlated with the altered microbiota Lachnoclostridium, Dialister, and Bacteroides, as well as with the altered metabolites all-trans-13,14-dihydroretinol, DL-dipalmitoylphosphatidylcholine (DPPC), LPC 24: 1, PC (14:1e/10:0), and 4-phenyl-3-buten-2-one, etc. The association network analysis further identified three MS-linked metabolites, including all-trans-13,14-dihydroretinol, DPPC, and 4-phenyl-3-buten-2-one, that were significantly correlated with the altered microbiota. Bio-functional validation showed that all-trans-13, 14-dihydroretinol could significantly upregulate the expression of lipid synthesis genes and inflammatory genes. This study identified a new biomarker that may contribute to MS development. These findings provided new insights regarding the development of efficient therapeutic strategies for MS. IMPORTANCE Metabolic syndrome (MS) has become a health concern worldwide. Gut microbiota and metabolites play an important role in human health. We first endeavored to comprehensively analyze the microbiome and metabolome signatures in obese children and found the novel microbial metabolites in MS. We further validated the biological functions of the metabolites in vitro and illustrated the effects of the microbial metabolites on lipid synthesis and inflammation. The microbial metabolite all-trans-13, 14-dihydroretinol may be a new biomarker in the pathogenesis of MS, especially in obese children. These findings were not available in previous studies, and they provide new insights regarding the management of metabolic syndrome.
Collapse
Affiliation(s)
- Jia Wei
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Wen Dai
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Xiongfeng Pan
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Yan Zhong
- Institute of Children Health, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Ningan Xu
- Institute of Children Health, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Ping Ye
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Jie Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Jina Li
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Fei Yang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang, Hunan, China
| | - Jiayou Luo
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Miyang Luo
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| |
Collapse
|
141
|
Xu M, Lan R, Qiao L, Lin X, Hu D, Zhang S, Yang J, Zhou J, Ren Z, Li X, Liu G, Liu L, Xu J. Bacteroides vulgatus Ameliorates Lipid Metabolic Disorders and Modulates Gut Microbial Composition in Hyperlipidemic Rats. Microbiol Spectr 2023; 11:e0251722. [PMID: 36625637 PMCID: PMC9927244 DOI: 10.1128/spectrum.02517-22] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/11/2023] Open
Abstract
Hyperlipidemia is a risk factor and key indicator for cardiovascular diseases, and the gut microbiota is highly associated with hyperlipidemia. Bacteroides vulgatus is a prevalent mutualist across human populations and confers multiple health benefits such as immunoregulation, antiobesity, and coronary artery disease intervention. However, its role in antihyperlipidemia has not been systematically characterized. This study sought to identify the effect of B. vulgatus Bv46 on hyperlipidemia. Hyperlipidemic rats were modeled by feeding them a high-fat diet for 6 weeks. The effect of B. vulgatus Bv46 supplementation was evaluated by measuring anthropometric parameters, lipid and inflammation markers, and the liver pathology. Multi-omics was used to explore the underlying mechanisms. The ability of B. vulgatus Bv46 to produce bile salt hydrolase was confirmed by gene annotation and in vitro experiments. Oral administration of B. vulgatus Bv46 in hyperlipidemic rats significantly reduced the body weight gain, food efficiency, and liver index, improved the serum lipid profile, lowered the levels of serum inflammatory cytokines, promoted the loss of fecal bile acids (BAs), and extended the fecal pool of short-chain fatty acids (SCFAs), especially propionate and butyrate. B. vulgatus Bv46 induced compositional shifts of the gut microbial community of hyperlipidemic rats, characterized by a lower ratio of Firmicutes to Bacteroidetes with an increase of genera Bacteroides and Parabacteroides. After intervention, serum metabolite profiling exhibited an adaptation in amino acids and glycerophospholipid metabolism. Transcriptomics further detected altered biological processes, including primary bile acid biosynthesis and fatty acid metabolic process. Taken together, the findings suggest that B. vulgatus Bv46 could be a promising candidate for interventions against hyperlipidemia. IMPORTANCE As a core microbe of the human gut ecosystem, Bacteroides vulgatus has been linked to multiple aspects of metabolic disorders in a collection of associative studies, which, while indicative, warrants more direct experimental evidence to verify. In this study, we experimentally demonstrated that oral administration of B. vulgatus Bv46 ameliorated the serum lipid profile and systemic inflammation of high-fat diet-induced hyperlipidemic rats in a microbiome-regulated manner, which appears to be associated with changes of bile acid metabolism, short-chain fatty acid biosynthesis, and serum metabolomic profile. This finding supports the causal contribution of B. vulgatus in host metabolism and helps to form the basis of novel therapies for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Mingchao Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Lei Qiao
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoying Lin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dalong Hu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Suping Zhang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing Yang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| | - Juan Zhou
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhihong Ren
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xianping Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guoxing Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liyun Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguo Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Public Health, Nankai University, Tianjin, China
| |
Collapse
|
142
|
Pharmacometabolomics for the Study of Lipid-Lowering Therapies: Opportunities and Challenges. Int J Mol Sci 2023; 24:ijms24043291. [PMID: 36834701 PMCID: PMC9960554 DOI: 10.3390/ijms24043291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/21/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Lipid-lowering therapies are widely used to prevent the development of atherosclerotic cardiovascular disease (ASCVD) and related mortality worldwide. "Omics" technologies have been successfully applied in recent decades to investigate the mechanisms of action of these drugs, their pleiotropic effects, and their side effects, aiming to identify novel targets for future personalized medicine with an improvement of the efficacy and safety associated with the treatment. Pharmacometabolomics is a branch of metabolomics that is focused on the study of drug effects on metabolic pathways that are implicated in the variation of response to the treatment considering also the influences from a specific disease, environment, and concomitant pharmacological therapies. In this review, we summarized the most significant metabolomic studies on the effects of lipid-lowering therapies, including the most commonly used statins and fibrates to novel drugs or nutraceutical approaches. The integration of pharmacometabolomics data with the information obtained from the other "omics" approaches could help in the comprehension of the biological mechanisms underlying the use of lipid-lowering drugs in view of defining a precision medicine to improve the efficacy and reduce the side effects associated with the treatment.
Collapse
|
143
|
Neveu V, Nicolas G, Amara A, Salek RM, Scalbert A. The human microbial exposome: expanding the Exposome-Explorer database with gut microbial metabolites. Sci Rep 2023; 13:1946. [PMID: 36732606 PMCID: PMC9894932 DOI: 10.1038/s41598-022-26366-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2022] [Accepted: 12/13/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolites produced by the gut microbiota play an important role in the cross-talk with the human host. Many microbial metabolites are biologically active and can pass the gut barrier and make it into the systemic circulation, where they form the gut microbial exposome, i.e. the totality of gut microbial metabolites in body fluids or tissues of the host. A major difficulty faced when studying the microbial exposome and its role in health and diseases is to differentiate metabolites solely or partially derived from microbial metabolism from those produced by the host or coming from the diet. Our objective was to collect data from the scientific literature and build a database on gut microbial metabolites and on evidence of their microbial origin. Three types of evidence on the microbial origin of the gut microbial exposome were defined: (1) metabolites are produced in vitro by human faecal bacteria; (2) metabolites show reduced concentrations in humans or experimental animals upon treatment with antibiotics; (3) metabolites show reduced concentrations in germ-free animals when compared with conventional animals. Data was manually collected from peer-reviewed publications and inserted in the Exposome-Explorer database. Furthermore, to explore the chemical space of the microbial exposome and predict metabolites uniquely formed by the microbiota, genome-scale metabolic models (GSMMs) of gut bacterial strains and humans were compared. A total of 1848 records on one or more types of evidence on the gut microbial origin of 457 metabolites was collected in Exposome-Explorer. Data on their known precursors and concentrations in human blood, urine and faeces was also collected. About 66% of the predicted gut microbial metabolites (n = 1543) were found to be unique microbial metabolites not found in the human GSMM, neither in the list of 457 metabolites curated in Exposome-Explorer, and can be targets for new experimental studies. This new data on the gut microbial exposome, freely available in Exposome-Explorer ( http://exposome-explorer.iarc.fr/ ), will help researchers to identify poorly studied microbial metabolites to be considered in future studies on the gut microbiota, and study their functionalities and role in health and diseases.
Collapse
Affiliation(s)
- Vanessa Neveu
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), 25 avenue Tony Garnier, CS 90627, 69366, Lyon Cedex 07, France
| | - Geneviève Nicolas
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), 25 avenue Tony Garnier, CS 90627, 69366, Lyon Cedex 07, France
| | - Adam Amara
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), 25 avenue Tony Garnier, CS 90627, 69366, Lyon Cedex 07, France
| | - Reza M Salek
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), 25 avenue Tony Garnier, CS 90627, 69366, Lyon Cedex 07, France
| | - Augustin Scalbert
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), 25 avenue Tony Garnier, CS 90627, 69366, Lyon Cedex 07, France.
| |
Collapse
|
144
|
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: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution 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
|
145
|
Lee-Sarwar KA, Chen YC, Lasky-Su J, Kelly RS, Zeiger RS, O’Connor GT, Bacharier LB, Jia X, Beigelman A, Gold DR, Laranjo N, Bunyavanich S, Weiss ST, Litonjua AA, Brennan PJ. Early-life fecal metabolomics of food allergy. Allergy 2023; 78:512-521. [PMID: 36448508 PMCID: PMC10590492 DOI: 10.1111/all.15602] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/04/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Intestinal microenvironmental perturbations may increase food allergy risk. We hypothesize that children with clinical food allergy, those with food sensitization, and healthy children can be differentiated by intestinal metabolites in the first years of life. METHODS In this ancillary analysis of the Vitamin D Antenatal Asthma Reduction Trial (VDAART), we performed untargeted metabolomic profiling in 824 stool samples collected at ages 3-6 months, 1 year and 3 years. Subjects included 23 with clinical food allergy at age 3 and/or 6 years, 151 with food sensitization but no clinical food allergy, and 220 controls. We identified modules of correlated, functionally related metabolites and sought associations of metabolite modules and individual metabolites with food allergy/sensitization using regression models. RESULTS Several modules of functionally related intestinal metabolites were reduced among subjects with food allergy, including bile acids at ages 3-6 months and 1 year, amino acids at age 3-6 months, steroid hormones at 1 year, and sphingolipids at age 3 years. One module primarily containing diacylglycerols was increased in those with food allergy at age 3-6 months. Fecal caffeine metabolites at age 3-6 months, likely derived from breast milk, were increased in those with food allergy and/or sensitization (beta = 5.9, 95% CI 1.0-10.8, p = .02) and were inversely correlated with fecal bile acids and bilirubin metabolites, though maternal plasma caffeine levels were not associated with food allergy and/or sensitization. CONCLUSIONS Several classes of bioactive fecal metabolites are associated with food allergy and/or sensitization including bile acids, steroid hormones, sphingolipids, and caffeine metabolites.
Collapse
Affiliation(s)
- Kathleen A. Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Yih-Chieh Chen
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Rachel S. Kelly
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Robert S. Zeiger
- Department of Clinical Science Kaiser Permanente Bernard J. Tyson School of Medicine; Pasadena, CA, USA
| | - George T. O’Connor
- Pulmonary Center and Department of Medicine, Boston University School of Medicine; Boston, MA, USA
| | - Leonard B. Bacharier
- Department of Pediatric Allergy, Immunology, and Pulmonary, Vanderbilt Children’s Hospital, Vanderbilt University Medical Center; Nashville, TN, USA
| | - Xiaojiong Jia
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Avraham Beigelman
- Schneider Children’s Medical Center of Israel, Tel Aviv University; Tel Aviv, Israel; Division of Pediatric Allergy, Immunology & Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine; St. Louis, MO, USA
| | - Diane R. Gold
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Nancy Laranjo
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Supinda Bunyavanich
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai; New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai; New York, NY, USA
| | - Scott T. Weiss
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children’s Hospital at Strong, University of Rochester Medical Center; Rochester, NY, USA
| | - Patrick J. Brennan
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| |
Collapse
|
146
|
Nagata N, Takeuchi T, Masuoka H, Aoki R, Ishikane M, Iwamoto N, Sugiyama M, Suda W, Nakanishi Y, Terada-Hirashima J, Kimura M, Nishijima T, Inooka H, Miyoshi-Akiyama T, Kojima Y, Shimokawa C, Hisaeda H, Zhang F, Yeoh YK, Ng SC, Uemura N, Itoi T, Mizokami M, Kawai T, Sugiyama H, Ohmagari N, Ohno H. Human Gut Microbiota and Its Metabolites Impact Immune Responses in COVID-19 and Its Complications. Gastroenterology 2023; 164:272-288. [PMID: 36155191 PMCID: PMC9499989 DOI: 10.1053/j.gastro.2022.09.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/25/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS We investigate interrelationships between gut microbes, metabolites, and cytokines that characterize COVID-19 and its complications, and we validate the results with follow-up, the Japanese 4D (Disease, Drug, Diet, Daily Life) microbiome cohort, and non-Japanese data sets. METHODS We performed shotgun metagenomic sequencing and metabolomics on stools and cytokine measurements on plasma from 112 hospitalized patients with SARS-CoV-2 infection and 112 non-COVID-19 control individuals matched by important confounders. RESULTS Multiple correlations were found between COVID-19-related microbes (eg, oral microbes and short-chain fatty acid producers) and gut metabolites (eg, branched-chain and aromatic amino acids, short-chain fatty acids, carbohydrates, neurotransmitters, and vitamin B6). Both were also linked to inflammatory cytokine dynamics (eg, interferon γ, interferon λ3, interleukin 6, CXCL-9, and CXCL-10). Such interrelationships were detected highly in severe disease and pneumonia; moderately in the high D-dimer level, kidney dysfunction, and liver dysfunction groups; but rarely in the diarrhea group. We confirmed concordances of altered metabolites (eg, branched-chain amino acids, spermidine, putrescine, and vitamin B6) in COVID-19 with their corresponding microbial functional genes. Results in microbial and metabolomic alterations with severe disease from the cross-sectional data set were partly concordant with those from the follow-up data set. Microbial signatures for COVID-19 were distinct from diabetes, inflammatory bowel disease, and proton-pump inhibitors but overlapping for rheumatoid arthritis. Random forest classifier models using microbiomes can highly predict COVID-19 and severe disease. The microbial signatures for COVID-19 showed moderate concordance between Hong Kong and Japan. CONCLUSIONS Multiomics analysis revealed multiple gut microbe-metabolite-cytokine interrelationships in COVID-19 and COVID-19related complications but few in gastrointestinal complications, suggesting microbiota-mediated immune responses distinct between the organ sites. Our results underscore the existence of a gut-lung axis in COVID-19.
Collapse
Affiliation(s)
- Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan; Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Tadashi Takeuchi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroaki Masuoka
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Ryo Aoki
- Mechanism-based Research Laboratory, Ezaki Glico Co, Ltd, Osaka, Japan
| | - Masahiro Ishikane
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Noriko Iwamoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masaya Sugiyama
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa, Japan,Department of Viral Pathogenesis and Controls, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Wataru Suda
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yumiko Nakanishi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Junko Terada-Hirashima
- Division of Respiratory Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Moto Kimura
- Department of Clinical Research Strategic Planning Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Hiroshi Inooka
- Mechanism-based Research Laboratory, Ezaki Glico Co, Ltd, Osaka, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasushi Kojima
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Chikako Shimokawa
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Fen Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China,Microbiota I-Center, Hong Kong, China,Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yun Kit Yeoh
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China,Microbiota I-Center, Hong Kong, China,Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China,Microbiota I-Center, Hong Kong, China,Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Naomi Uemura
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan,Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Kohnodai Hospital, Tokyo, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Masashi Mizokami
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Takashi Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Haruhito Sugiyama
- Division of Respiratory Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| |
Collapse
|
147
|
Zhang X, Adebayo AS, Wang D, Raza Y, Tomlinson M, Dooley H, Bowyer RC, Small KS, Steves CJ, Spector TD, Duncan EL, Visconti A, Falchi M. PPI-Induced Changes in Plasma Metabolite Levels Influence Total Hip Bone Mineral Density in a UK Cohort. J Bone Miner Res 2023; 38:326-334. [PMID: 36458982 PMCID: PMC10108201 DOI: 10.1002/jbmr.4754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/22/2022] [Revised: 11/08/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022]
Abstract
Proton pump inhibitors (PPIs) are among the most used drugs in the UK. PPI use has been associated with decreased bone mineral density (BMD) and increased fracture risk, although these results have been inconsistent. We hypothesized that PPI could modulate BMD by altering gut and/or host systemic metabolic environments. Using data from more than 5000 British male and female individuals, we confirmed that PPI use is associated with decreased lumbar spine and total hip BMD. This effect was not mediated through the gut microbiome. We suggest here that PPI use may influence total hip BMD, both directly and indirectly, via plasma metabolites involved in the sex hormone pathway. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Adewale S. Adebayo
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
- Present address:
NIHR Leicester Biomedical Research Centre, Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Dongmeng Wang
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Yasrab Raza
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Max Tomlinson
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Hannah Dooley
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Ruth C.E. Bowyer
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Kerrin S. Small
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Claire J. Steves
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Tim D. Spector
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Emma L. Duncan
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Alessia Visconti
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| | - Mario Falchi
- Department of Twins Research & Genetics EpidemiologyKing's College LondonLondonUK
| |
Collapse
|
148
|
Safari-Alighiarloo N, Emami Z, Rezaei-Tavirani M, Alaei-Shahmiri F, Razavi S. Gut Microbiota and Their Associated Metabolites in Diabetes: A Cross Talk Between Host and Microbes-A Review. Metab Syndr Relat Disord 2023; 21:3-15. [PMID: 36301254 DOI: 10.1089/met.2022.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022] Open
Abstract
Dysbiosis of the gut microbiota's composition and function is important in developing insulin resistance and diabetes. Diabetes has also been linked to changes in the circulating and fecal metabolites. Evidence suggests the associations between the gut microbiota and the aberrant diabetes-related metabolome. Metabolites play a crucial role in the host-microbiota interactions. Researchers have used a combination of metagenomic and metabolomic approaches to investigate the relationships between gut microbial dysbiosis and metabolic abnormalities in diabetes. We summarized current discoveries on the associations between the gut microbiota and metabolites in type 1 diabetes, type 2 diabetes, and gestational diabetes mellitus in the scoping review. According to research, the gut microbiota changes might involve in the development of diabetes through modulating the host's metabolic pathways such as immunity, energy metabolism, lipid metabolism, and amino acid metabolism. These results add to our understanding of the interplay between the host and gut microbiota metabolism.
Collapse
Affiliation(s)
- Nahid Safari-Alighiarloo
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Emami
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Alaei-Shahmiri
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Razavi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
149
|
Chavan AR, Singh AK, Gupta RK, Nakhate SP, Poddar BJ, Gujar VV, Purohit HJ, Khardenavis AA. Recent trends in the biotechnology of functional non-digestible oligosaccharides with prebiotic potential. Biotechnol Genet Eng Rev 2023:1-46. [PMID: 36714949 DOI: 10.1080/02648725.2022.2152627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/20/2022] [Accepted: 11/13/2022] [Indexed: 01/31/2023]
Abstract
Prebiotics as a part of dietary nutrition can play a crucial role in structuring the composition and metabolic function of intestinal microbiota and can thus help in managing a clinical scenario by preventing diseases and/or improving health. Among the different prebiotics, non-digestible carbohydrates are molecules that selectively enrich a typical class of bacteria with probiotic potential. This review summarizes the current knowledge about the different aspects of prebiotics, such as its production, characterization and purification by various techniques, and its link to novel product development at an industrial scale for wide-scale use in diverse range of health management applications. Furthermore, the path to effective valorization of agricultural residues in prebiotic production has been elucidated. This review also discusses the recent developments in application of genomic tools in the area of prebiotics for providing new insights into the taxonomic characterization of gut microorganisms, and exploring their functional metabolic pathways for enzyme synthesis. However, the information regarding the cumulative effect of prebiotics with beneficial bacteria, their colonization and its direct influence through altered metabolic profile is still getting established. The future of this area lies in the designing of clinical condition specific functional foods taking into consideration the host genotypes, thus facilitating the creation of balanced and required metabolome and enabling to maintain the healthy status of the host.
Collapse
Affiliation(s)
- Atul Rajkumar Chavan
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ashish Kumar Singh
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rakesh Kumar Gupta
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Suraj Prabhakarrao Nakhate
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Bhagyashri Jagdishprasad Poddar
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vaibhav Vilasrao Gujar
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
- JoVE, Mumbai, India
| | - Hemant J Purohit
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
| | - Anshuman Arun Khardenavis
- Environmental Biotechnology and Genomics Division, CSIR-National Environmental Engineering Research Institute, Nagpur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
150
|
Morrison AG, Sarkar S, Umar S, Lee STM, Thomas SM. The Contribution of the Human Oral Microbiome to Oral Disease: A Review. Microorganisms 2023; 11:318. [PMID: 36838283 PMCID: PMC9962706 DOI: 10.3390/microorganisms11020318] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/28/2023] Open
Abstract
The oral microbiome is an emerging field that has been a topic of discussion since the development of next generation sequencing and the implementation of the human microbiome project. This article reviews the current literature surrounding the oral microbiome, briefly highlighting most recent methods of microbiome characterization including cutting edge omics, databases for the microbiome, and areas with current gaps in knowledge. This article also describes reports on microorganisms contained in the oral microbiome which include viruses, archaea, fungi, and bacteria, and provides an in-depth analysis of their significant roles in tissue homeostasis. Finally, we detail key bacteria involved in oral disease, including oral cancer, and the current research surrounding their role in stimulation of inflammatory cytokines, the role of gingival crevicular fluid in periodontal disease, the creation of a network of interactions between microorganisms, the influence of the planktonic microbiome and cospecies biofilms, and the implications of antibiotic resistance. This paper provides a comprehensive literature analysis while also identifying gaps in knowledge to enable future studies to be conducted.
Collapse
Affiliation(s)
- Austin Gregory Morrison
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soumyadev Sarkar
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Shahid Umar
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sonny T. M. Lee
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
- 1717 Claflin Road, 136 Ackert Hall, Manhattan, KS 66506, USA
| | - Sufi Mary Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Departments of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Departments of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- 3901 Rainbow Blvd., 4031 Wahl Hall East, MS 3040, Kansas City, KS 66160, USA
| |
Collapse
|