351
|
Mojsak P, Rey-Stolle F, Parfieniuk E, Kretowski A, Ciborowski M. The role of gut microbiota (GM) and GM-related metabolites in diabetes and obesity. A review of analytical methods used to measure GM-related metabolites in fecal samples with a focus on metabolites' derivatization step. J Pharm Biomed Anal 2020; 191:113617. [PMID: 32971497 DOI: 10.1016/j.jpba.2020.113617] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022]
Abstract
Disruption of gut microbiota (GM) composition is increasingly related to the pathogenesis of various metabolic diseases. Additionally, GM is responsible for the production and transformation of metabolites involved in the development of metabolic disorders, such as obesity and type 2 diabetes mellitus (T2DM). The current state of knowledge regarding the composition of GM and GM-related metabolites in relation to the progress and development of obesity and T2DM is presented in this review. To understand the relationships between GM-related metabolites and the development of metabolic disorders, their accurate qualitative and quantitative measurement in biological samples is needed. Feces represent a valuable biological matrix which composition may reflect the health status of the lower gastrointestinal tract and the whole organism. Mass spectrometry (MS), mainly in combination with gas chromatography (GC) or liquid chromatography (LC), is commonly used to measure fecal metabolites. However, profiling metabolites in such a complex matrix as feces is challenging from both analytical chemistry and biochemistry standpoints. Chemical derivatization is one of the most effective methods used to overcome these problems. In this review, we provide a comprehensive summary of the derivatization methods of GM-related metabolites prior to GC-MS or LC-MS analysis, which have been published in the last five years (2015-2020). Additionally, analytical methods used for the analysis of GM-related metabolites without the derivatization step are also presented.
Collapse
Affiliation(s)
- Patrycja Mojsak
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Fernanda Rey-Stolle
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, Spain
| | - Ewa Parfieniuk
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Adam Kretowski
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Michal Ciborowski
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
352
|
Hu X, Xie Y, Xiao Y, Zeng W, Gong Z, Du J. Longitudinal analysis of fecal microbiome and metabolome during renal fibrotic progression in a unilateral ureteral obstruction animal model. Eur J Pharmacol 2020; 886:173555. [PMID: 32937112 DOI: 10.1016/j.ejphar.2020.173555] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 02/02/2023]
Abstract
Renal fibrosis is a major pathological process in the progression of various chronic kidney diseases to end-stage renal disease (ESRD). Growing evidence has suggested that gut microbiota dysbiosis is closely related to ESRD. However, the interplay between altered fecal microbiome and metabolome during the renal fibrotic process remains unclear. Herein, an integrated approach of 16S ribosomal DNA sequencing combined with an ultra-high performance liquid chromatography-mass spectrometry-based metabolomics platform was applied to investigate the dynamic changes of fecal microbiota and metabolites throughout renal fibrosis progression in a mouse model of unilateral ureteral obstruction (UUO). The composition of gut microbiota changed markedly before and after UUO surgery. UUO mice showed a decrease in short-chain fatty acids-producing genera, including Bacteroides, Prevotellaceae_UCG-001, Roseburia, and Lachnospiraceae_NK4A136_group, as well as an increase in the genera Parasutterella and Alistipes, which changed dynamically over time. Additionally, 41 differential metabolites, mainly involved in 12 metabolic pathways, including inositol phosphate metabolism, primary bile acid biosynthesis, biosynthesis of unsaturated fatty acids, taurine and hypotaurine metabolism, purine metabolism, were identified in the UUO mice before and after surgery. Four fecal metabolites, myo-inositol, dodecanoic acid, N-acetylputrescine, and anthranilic acid, were positively associated with the progression of renal fibrosis. Moreover, by using multi-omics analyses, we found the alteration in UUO-related gut microbiota was correlated with a change in fecal metabolites. Therefore, our results provide insights into disturbances of the microbiome-metabolome interface in the progression of UUO-related renal fibrosis.
Collapse
Affiliation(s)
- Xiaofang Hu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; The Hunan Institute of Pharmacy Practice and Clinical Research, Changsha, 410008, Hunan, China
| | - Yuhong Xie
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; The Hunan Institute of Pharmacy Practice and Clinical Research, Changsha, 410008, Hunan, China
| | - Yi Xiao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; The Hunan Institute of Pharmacy Practice and Clinical Research, Changsha, 410008, Hunan, China
| | - Wenjing Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; The Hunan Institute of Pharmacy Practice and Clinical Research, Changsha, 410008, Hunan, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; The Hunan Institute of Pharmacy Practice and Clinical Research, Changsha, 410008, Hunan, China.
| | - Jie Du
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; The Hunan Institute of Pharmacy Practice and Clinical Research, Changsha, 410008, Hunan, China.
| |
Collapse
|
353
|
Bailey MJ, Naik NN, Wild LE, Patterson WB, Alderete TL. Exposure to air pollutants and the gut microbiota: a potential link between exposure, obesity, and type 2 diabetes. Gut Microbes 2020; 11:1188-1202. [PMID: 32347153 PMCID: PMC7524284 DOI: 10.1080/19490976.2020.1749754] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Work has shown that increased exposure to air pollutants independently contributes to obesity and type 2 diabetes risk, yet the exact mechanisms underlying these associations have not been fully characterized. The current review summarizes recent findings regarding the impact of inhaled and ingested air pollutants on the gut microbiota. Animal and human studies provide evidence that air pollutants, such as particulate matter, nitrogen oxides, and ozone, have the potential to alter the gut microbiota. Further, studies suggest that such exposure-induced alterations to the gut microbiota may contribute to increased risk for obesity and type 2 diabetes through inflammatory pathways. Future work is needed to fully understand the complex interactions between air pollution, the gut microbiome, and human health. Additionally, advanced sequencing methods for gut microbiome research present unique opportunities to study the underlying pathways that link increased air pollution exposure with obesity and type 2 diabetes risk.
Collapse
Affiliation(s)
- Maximillian J. Bailey
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Noopur N. Naik
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Laura E. Wild
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - William B. Patterson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Tanya L. Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA,CONTACT Tanya L. Alderete University of Colorado Boulder, Department of Integrative Physiology, Ramaley Biology Building, 1800 Colorado Avenue, N379, Boulder, CO80309
| |
Collapse
|
354
|
Identification of new biomarkers of pyridoxine-dependent epilepsy by GC/MS-based urine metabolomics. Anal Biochem 2020; 604:113739. [DOI: 10.1016/j.ab.2020.113739] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/15/2022]
|
355
|
Tang Q, Cang S, Jiao J, Rong W, Xu H, Bi K, Li Q, Liu R. Integrated study of metabolomics and gut metabolic activity from ulcerative colitis to colorectal cancer: The combined action of disordered gut microbiota and linoleic acid metabolic pathway might fuel cancer. J Chromatogr A 2020; 1629:461503. [PMID: 32858455 DOI: 10.1016/j.chroma.2020.461503] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is one of the most serious complications of ulcerative colitis (UC). Altered gut microbiota is implicated in the development of CRC and metabolic perturbations are often associated with changes in the gut microbiome composition. Given the links between gut microbiome and the metabolic profiles in the body, an approach involving ultra-high-performance liquid chromatography combined with electrospray ionization quadrupole time-of-flight tandem mass spectrometry (UHPLC-Q-TOF-MS/MS) metabolomics and 16S rDNA sequencing technology was applied to trace the development UC into CRC in rats. The study identified 11 differential metabolites related to both UC and CRC, which mainly referred to the linoleic acid metabolism. Among these, linoleic acid and 12‑hydroxy‑8,10-octadecadienoic acid could serve as key biomarkers for the development of UC into CRC. Besides, a significant change was observed in the microflora structure during the development from UC to CRC; this mainly involved a gradual increase in Escherichia-Shigella and a gradual decrease in Lactobacillus. In addition, Pearson's correlation analysis revealed strong correlations between intestinal microflora-related metabolites and specific intestinal microflora, which indicated both of them can promote the transition of UC to CRC. The results of the present study provided positive support for the involvement of intestinal microflora and host metabolism in the pathophysiological mechanism that is responsible for the development of UC into CRC. This information can help understand the risk for CRC that accompanies a diagnosis of UC and also provide different means of targeting these differential metabolites and intestinal microbiota to avoid UC-induced CRC.
Collapse
Affiliation(s)
- Qi Tang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Song Cang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Jiao Jiao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Weiwei Rong
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Huarong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Qing Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Ran Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
356
|
Han S, Chen ZJ, Zhou D, Zheng P, Zhang JH, Jia G. [Effects of titanium dioxide nanoparticles on fecal metabolome in rats after oral administration for 90 days]. JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2020; 52:457-463. [PMID: 32541978 DOI: 10.19723/j.issn.1671-167x.2020.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To explore the effects and related mechanisms of oral exposure titanium dioxide nanoparticles (TiO2 NPs) for 90 days on the intestinal and the gut microbiota of rats, through fecal metabolomics. METHODS Twelve 4-week-old clean-grade Sprague Dawley (SD) rats were randomly de-vided into 2 groups by body weight, treated with TiO2 NPs at dose of 0 or 50 mg/kg body weight everyday respectively for 90 days. The solution of each infection was freshly prepared and shocked fully by ultrasonic. Characterization of the particle size, crystal form, purity, and specific surface area of TiO2 NPs was conducted. And the fresh feces of the rats were collected on the 90th day. After lyophilized and hydrophilic phase extraction, ultra performance liquid chromatography-Q-exactive orbitrap-high-resolution mass spectrometry system (UPLC-QEMS) was utilized for non-targeted determination of fecal meta-bolites. The metabolites were identified and labeled through Compound Discoverer 3.0 software, and used for subsequent metabolomics analysis. Bioinformatics analysis was carried out including unsupervised principal component analysis and supervised orthogonal projection to latent structure discriminant analysis for the differential metabolites between the two groups. The differential metabolites were followed-up for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. RESULTS Compared with the control group, the body weight of the rats was significantly reduced (P<0.05) in the treatment group. A total of 22 metabolites in fecal metabolomics showed significant changes. Among them, xanthine, 1-methyladenine, 3-hydroxypyridine, methionine sulfoxide, pyridoxine, 1,5-isoquinolinediol, N-acetylornithine, N-acetyl-D-galactosamine, L-citrulline, L-methionine, leucine, DL-tryptophan, L-ornithine, 4-methyl-5-thiazoleethanol, and L-glutamic acid totaled 15 metabolites increased significantly. N-acetylhistamine, D-pipecolinic acid, imidazolelactic acid, L-valine, 2,3,4,6-tetramethylpyrazine, caprolactam, and histamine totaled 7 metabolites decreased significantly. N-acetylhistamine, L-valine and methionine sulfoxide were changed more than 16 times. Analysis of KEGG pathway revealed that the two metabolic pathways arginine biosynthesis and aminoacyl-tRNA biosynthesis were significantly changed (false discover rate < 0.05, pathway impact > 0.1). CONCLUSION Oral exposure to TiO2 NPs for 90 days could disrupt the metabolism of the intestine and gut microbiota, causing significant changes in metabolites and metabolic pathways which were related to inflammatory response, oxidative stress, glucose homeostasis, blood system and amino acid homeostasis in rat feces. It is suggested that the toxic effect of TiO2 NPs on rats may be closely related to intestinal and gut microbiota metabolism.
Collapse
Affiliation(s)
- S Han
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing 100191, China
| | - Z J Chen
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing 100191, China
| | - D Zhou
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing 100191, China
| | - P Zheng
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing 100191, China
| | - J H Zhang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing 100191, China
| | - G Jia
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing 100191, China
| |
Collapse
|
357
|
Jing N, Liu X, Jin M, Yang X, Hu X, Li C, Zhao K. Fubrick tea attenuates high-fat diet induced fat deposition and metabolic disorder by regulating gut microbiota and caffeine metabolism. Food Funct 2020; 11:6971-6986. [PMID: 32697259 DOI: 10.1039/d0fo01282c] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fubrick tea aqueous extract (FTEs) has been reported to improve lipid metabolism and gut microbiota communities in mice and humans. However, it is still unclear how FTEs prevents obesity through gut microbiota, and whether some other regulatory mechanisms are involved in the process. Here, we found that FTEs supplementation effectively alleviated the body weight gain, visceral fat accumulation, dyslipidemia, and impaired glucose tolerance induced by a high-fat diet (HFD), and fecal microbiota transplantation (FMT) from FTEs-treated mice showed similar protective effects as FTEs supplementation in mice fed with a HFD. The results confirmed that gut microbiota played key roles in attenuating HFD-induced fat deposition and metabolic disorder. In particular, FTEs reversed HFD-induced gut microbiota dysbiosis via increasing the relative abundances of Bacteroides, Adlercreutzia, Alistipes, Parabacteroides, and norank_f_Lachnospiraceae, and reducing that of Staphylococcus. Interestingly, FTEs could still alleviate HFD-induced lipid accumulation in mice treated with antibiotics, which had increased relative abundances of Bacteroidetes, Bacteroides, and Bacteroides_uniformis sp. In addition, supplementation with FTEs also modified the serum metabolome, especially the "caffeine metabolism" pathway. Furthermore, FTEs supplementation increased the concentrations of caffeine, theophylline, and theobromine in serum, which were positively correlated with an abundance of norank_f_Lachnospiraceae. Overall, FTEs exerts beneficial effects against obesity induced by HFD, and the underlying mechanism is partially related to the reprogramming of intestinal microbiota, while the metabolism of caffeine in FTEs also played an important role in the process. This study provides a theoretical basis for the further study of the anti-obesity effects of FTEs and the consideration of gut microbiota as a potential target for the treatment of obesity induced by a HFD.
Collapse
Affiliation(s)
- Nana Jing
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, PR China.
| | | | | | | | | | | | | |
Collapse
|
358
|
Fraisier-Vannier O, Chervin J, Cabanac G, Puech V, Fournier S, Durand V, Amiel A, André O, Benamar OA, Dumas B, Tsugawa H, Marti G. MS-CleanR: A Feature-Filtering Workflow for Untargeted LC-MS Based Metabolomics. Anal Chem 2020; 92:9971-9981. [PMID: 32589017 DOI: 10.1021/acs.analchem.0c01594] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Untargeted metabolomics using liquid chromatography-mass spectrometry (LC-MS) is currently the gold-standard technique to determine the full chemical diversity in biological samples. However, this approach still has many limitations; notably, the difficulty of accurately estimating the number of unique metabolites profiled among the thousands of MS ion signals arising from chromatograms. Here, we describe a new workflow, MS-CleanR, based on the MS-DIAL/MS-FINDER suite, which tackles feature degeneracy and improves annotation rates. We show that implementation of MS-CleanR reduces the number of signals by nearly 80% while retaining 95% of unique metabolite features. Moreover, the annotation results from MS-FINDER can be ranked according to the database chosen by the user, which enhance identification accuracy. Application of MS-CleanR to the analysis of Arabidopsis thaliana grown in three different conditions fostered class separation resulting from multivariate data analysis and led to annotation of 75% of the final features. The full workflow was applied to metabolomic profiles from three strains of the leguminous plant Medicago truncatula that have different susceptibilities to the oomycete pathogen Aphanomyces euteiches. A group of glycosylated triterpenoids overrepresented in resistant lines were identified as candidate compounds conferring pathogen resistance. MS-CleanR is implemented through a Shiny interface for intuitive use by end-users (available at https://github.com/eMetaboHUB/MS-CleanR).
Collapse
Affiliation(s)
- Ophélie Fraisier-Vannier
- Pharma Dev, Université de Toulouse, IRD, UPS, 31400 Toulouse, France.,Institut de Recherche en Informatique de Toulouse, Université de Toulouse, UPS, Toulouse 31400, France
| | - Justine Chervin
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,Metatoul-AgromiX Platform, MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, LRSV, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France
| | - Guillaume Cabanac
- Institut de Recherche en Informatique de Toulouse, Université de Toulouse, UPS, Toulouse 31400, France
| | - Virginie Puech
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,Metatoul-AgromiX Platform, MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, LRSV, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France
| | - Sylvie Fournier
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,Metatoul-AgromiX Platform, MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, LRSV, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France
| | - Virginie Durand
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France
| | - Aurélien Amiel
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,De Sangosse, Bonnel, 47480 Pont-Du-Casse, France
| | - Olivier André
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,De Sangosse, Bonnel, 47480 Pont-Du-Casse, France
| | - Omar Abdelaziz Benamar
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,De Sangosse, Bonnel, 47480 Pont-Du-Casse, France
| | - Bernard Dumas
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France
| | - Hiroshi Tsugawa
- RIKEN Center for Sustainable Resource Science, Yokohama 230-0045, Japan.,RIKEN Center for Integrative Medical Science, Yokohama 230-0045, Japan
| | - Guillaume Marti
- Pharma Dev, Université de Toulouse, IRD, UPS, 31400 Toulouse, France.,Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,Metatoul-AgromiX Platform, MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, LRSV, Université de Toulouse, CNRS, UPS, 31400 Toulouse, France.,Institut de Recherche en Informatique de Toulouse, Université de Toulouse, UPS, Toulouse 31400, France
| |
Collapse
|
359
|
Oleskin AV, Shenderov BA. Probiotics and Psychobiotics: the Role of Microbial Neurochemicals. Probiotics Antimicrob Proteins 2020; 11:1071-1085. [PMID: 31493127 DOI: 10.1007/s12602-019-09583-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In light of recent data, microorganisms should be construed as organisms that are capable of communication and collective behaviors. Microbial communication signals are involved both in interactions among microbial cells within microbial social systems, including the human body-inhabiting microconsortium, and the dialog between the microbiota and the host organism. The microbiota inhabits various niches of the host organism, especially the gastrointestinal (GI) tract. Microorganisms release diverse signal molecules and, in addition, specifically respond to host signals. This enables them to constantly interact with the nervous system including the brain and the immune system of the host organism. Evolutionarily conserved signals that are involved in the communication between microbiota and the host include neuroactive substances (neurochemicals) such as peptides, amino acids, biogenic amines, short-chain fatty acids, and gaseous substances. This ongoing dialog may either stabilize the host's physical and mental health state or, alternatively, cause serious health problems. Attempts are made to correct imbalances in the brain-gut-microbiota axis with probiotics including their subgroup called psychobiotics that release neuroactive substances directly influencing the human brain, psyche, and behavior. A number of recent review works address the microbiota-host system and its communication signals. Some of the publications focus on the involvement of neurochemicals in the bidirectional communication within the host-microbiota system. However, this work concentrates on the impact of bacterial cell components, metabolites, and signal molecules as promising alternatives to the currently widespread probiotics that have both advantages and disadvantages. Such biologically active agents of microbial origin are referred to as postbiotics or, alternatively, metabiotics (the term preferred in this work).
Collapse
Affiliation(s)
- Alexander V Oleskin
- General Ecology Department, School of Biology, Moscow State University, Vorobiev Hills, Moscow, Russia, 119991.
| | - Boris A Shenderov
- Laboratory of Bacteriology and Parasitology, Centre for Strategic Planning, Russian Ministry of Health; Moscow, Research Laboratory for Design & Implementation of Personalized Nutrition-Related Products & Diets, K. G. Razumovsky University of Technology & Management, Moscow, Russia
| |
Collapse
|
360
|
Consumption of Stilbenes and Flavonoids is Linked to Reduced Risk of Obesity Independently of Fiber Intake. Nutrients 2020; 12:nu12061871. [PMID: 32585900 PMCID: PMC7353284 DOI: 10.3390/nu12061871] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Polyphenol consumption is implicated in gut microbiome composition and improved metabolic outcomes, but it is unclear whether the effect is independent of dietary fiber. Methods: We investigated the links between (poly)phenol intake, gut microbiome composition (16s RNA) and obesity independently of fiber intake in UK women (n = 1810) and in a small group of UK men (n = 64). Results: (Poly)phenol intakes correlated with microbiome alpha diversity (Shannon Index) after adjusting for confounders and fiber intake. Moreover, flavonoid intake was significantly correlated with the abundance of Veillonella, (a genus known to improve physical performance), and stilbene intake with that of butyrate-producing bacteria (Lachnospira and Faecalibacterium). Stilbene and flavonoid intake also correlated with lower odds of prevalent obesity (Stilbenes: Odds Ratio (95% Confidence Interval) (OR(95%CI)) = 0.80 (0.73, 0.87), p = 4.90 × 10−7; Flavonoids: OR(95%CI) = 0.77 (0.65, 0.91), p = 0.002). Formal mediation analyses revealed that gut microbiome mediates ~11% of the total effect of flavonoid and stilbene intake on prevalent obesity. Conclusions: Our findings highlight the importance of (poly)phenol consumption for optimal human health.
Collapse
|
361
|
Berry SE, Valdes AM, Drew DA, Asnicar F, Mazidi M, Wolf J, Capdevila J, Hadjigeorgiou G, Davies R, Al Khatib H, Bonnett C, Ganesh S, Bakker E, Hart D, Mangino M, Merino J, Linenberg I, Wyatt P, Ordovas JM, Gardner CD, Delahanty LM, Chan AT, Segata N, Franks PW, Spector TD. Human postprandial responses to food and potential for precision nutrition. Nat Med 2020; 26:964-973. [PMID: 32528151 PMCID: PMC8265154 DOI: 10.1038/s41591-020-0934-0] [Citation(s) in RCA: 420] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 05/11/2020] [Indexed: 12/18/2022]
Abstract
Metabolic responses to food influence risk of cardiometabolic disease, but large-scale high-resolution studies are lacking. We recruited n = 1,002 twins and unrelated healthy adults in the United Kingdom to the PREDICT 1 study and assessed postprandial metabolic responses in a clinical setting and at home. We observed large inter-individual variability (as measured by the population coefficient of variation (s.d./mean, %)) in postprandial responses of blood triglyceride (103%), glucose (68%) and insulin (59%) following identical meals. Person-specific factors, such as gut microbiome, had a greater influence (7.1% of variance) than did meal macronutrients (3.6%) for postprandial lipemia, but not for postprandial glycemia (6.0% and 15.4%, respectively); genetic variants had a modest impact on predictions (9.5% for glucose, 0.8% for triglyceride, 0.2% for C-peptide). Findings were independently validated in a US cohort (n = 100 people). We developed a machine-learning model that predicted both triglyceride (r = 0.47) and glycemic (r = 0.77) responses to food intake. These findings may be informative for developing personalized diet strategies. The ClinicalTrials.gov registration identifier is NCT03479866.
Collapse
Affiliation(s)
- Sarah E Berry
- Department of Nutrition, King's College London, London, UK
| | - Ana M Valdes
- School of Medicine, University of Nottingham, Nottingham, UK.
- Nottingham NIHR Biomedical Research Centre, Nottingham, UK.
| | - David A Drew
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Mohsen Mazidi
- Department of Twins Research & Genetic Epidemiology, King's College London, London, UK
| | | | | | | | | | - Haya Al Khatib
- Department of Nutrition, King's College London, London, UK
- Zoe Global Ltd, London, UK
| | | | | | | | - Deborah Hart
- Department of Twins Research & Genetic Epidemiology, King's College London, London, UK
| | - Massimo Mangino
- Department of Twins Research & Genetic Epidemiology, King's College London, London, UK
| | - Jordi Merino
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | | | | | - Jose M Ordovas
- JM-USDA-HNRCA at Tufts University, Boston, MA, USA
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | | | - Linda M Delahanty
- Diabetes Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
| | - Paul W Franks
- Department of Twins Research & Genetic Epidemiology, King's College London, London, UK
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Tim D Spector
- Department of Twins Research & Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
362
|
Abstract
Metabolomics has been identified as a means of functionally assessing the net biological activity of a particular microbial community. Considering the oral microbiome, such an approach remains largely underused. While the current knowledge of the oral microbiome is constantly expanding, there are several deficits in knowledge particularly relating to their interactions with their host. This work uses nuclear magnetic resonance spectroscopy to investigate metabolic differences between oral microbial metabolism of endogenous (i.e., salivary protein) and exogenous (i.e., dietary carbohydrates) substrates. It also investigated whether microbial generation of different metabolites may be associated with host taste perception. This work found that in the absence of exogenous substrate, oral bacteria readily catabolize salivary protein and generate metabolic profiles similar to those seen in vivo. Important metabolites such as acetate, butyrate, and propionate are generated at relatively high concentrations. Higher concentrations of metabolites were generated by tongue biofilm compared to planktonic salivary bacteria. Thus, as has been postulated, metabolite production in proximity to taste receptors could reach relatively high concentrations. In the presence of 0.25 M exogenous sucrose, increased catabolism was observed with increased concentrations of a range of metabolites relating to glycolysis (lactate, pyruvate, succinate). Additional pyruvate-derived molecules such as acetoin and alanine were also increased. Furthermore, there was evidence that individual taste sensitivity to sucrose was related to differences in the metabolic fate of sucrose in the mouth. High-sensitivity perceivers appeared more inclined toward continual citric acid cycle activity postsucrose, whereas low-sensitivity perceivers had a more efficient conversion of pyruvate to lactate. This work collectively indicates that the oral microbiome exists in a complex balance with the host, with fluctuating metabolic activity depending on nutrient availability. There is preliminary evidence of an association between host behavior (sweet taste perception) and oral catabolism of sugar.
Collapse
Affiliation(s)
- A Gardner
- Salivary Research, Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King's College London, London, UK
- Department of Restorative Dentistry, Dental Hospital and School, University of Dundee, Dundee, UK
| | - P W So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - G H Carpenter
- Salivary Research, Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
363
|
Tolstikov V, Moser AJ, Sarangarajan R, Narain NR, Kiebish MA. Current Status of Metabolomic Biomarker Discovery: Impact of Study Design and Demographic Characteristics. Metabolites 2020; 10:metabo10060224. [PMID: 32485899 PMCID: PMC7345110 DOI: 10.3390/metabo10060224] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
Widespread application of omic technologies is evolving our understanding of population health and holds promise in providing precise guidance for selection of therapeutic interventions based on patient biology. The opportunity to use hundreds of analytes for diagnostic assessment of human health compared to the current use of 10–20 analytes will provide greater accuracy in deconstructing the complexity of human biology in disease states. Conventional biochemical measurements like cholesterol, creatinine, and urea nitrogen are currently used to assess health status; however, metabolomics captures a comprehensive set of analytes characterizing the human phenotype and its complex metabolic processes in real-time. Unlike conventional clinical analytes, metabolomic profiles are dramatically influenced by demographic and environmental factors that affect the range of normal values and increase the risk of false biomarker discovery. This review addresses the challenges and opportunities created by the evolving field of clinical metabolomics and highlights features of study design and bioinformatics necessary to maximize the utility of metabolomics data across demographic groups.
Collapse
Affiliation(s)
- Vladimir Tolstikov
- BERG, Precision Medicine Division, Framingham, MA 01701, USA; (V.T.); (R.S.); (N.R.N.)
| | - A. James Moser
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA;
| | | | - Niven R. Narain
- BERG, Precision Medicine Division, Framingham, MA 01701, USA; (V.T.); (R.S.); (N.R.N.)
| | - Michael A. Kiebish
- BERG, Precision Medicine Division, Framingham, MA 01701, USA; (V.T.); (R.S.); (N.R.N.)
- Correspondence: ; Tel.: +1-617-588-2245
| |
Collapse
|
364
|
Sudhakaran M, Doseff AI. The Targeted Impact of Flavones on Obesity-Induced Inflammation and the Potential Synergistic Role in Cancer and the Gut Microbiota. Molecules 2020; 25:E2477. [PMID: 32471061 PMCID: PMC7321129 DOI: 10.3390/molecules25112477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is an inflammatory disease that is approaching pandemic levels, affecting nearly 30% of the world's total population. Obesity increases the risk of diabetes, cardiovascular disorders, and cancer, consequentially impacting the quality of life and imposing a serious socioeconomic burden. Hence, reducing obesity and related life-threatening conditions has become a paramount health challenge. The chronic systemic inflammation characteristic of obesity promotes adipose tissue remodeling and metabolic changes. Macrophages, the major culprits in obesity-induced inflammation, contribute to sustaining a dysregulated immune function, which creates a vicious adipocyte-macrophage crosstalk, leading to insulin resistance and metabolic disorders. Therefore, targeting regulatory inflammatory pathways has attracted great attention to overcome obesity and its related conditions. However, the lack of clinical efficacy and the undesirable side-effects of available therapeutic options for obesity provide compelling reasons for the need to identify additional approaches for the prevention and treatment of obesity-induced inflammation. Plant-based active metabolites or nutraceuticals and diets with an increased content of these compounds are emerging as subjects of intense scientific investigation, due to their ability to ameliorate inflammatory conditions and offer safe and cost-effective opportunities to improve health. Flavones are a class of flavonoids with anti-obesogenic, anti-inflammatory and anti-carcinogenic properties. Preclinical studies have laid foundations by establishing the potential role of flavones in suppressing adipogenesis, inducing browning, modulating immune responses in the adipose tissues, and hindering obesity-induced inflammation. Nonetheless, the understanding of the molecular mechanisms responsible for the anti-obesogenic activity of flavones remains scarce and requires further investigations. This review recapitulates the molecular aspects of obesity-induced inflammation and the crosstalk between adipocytes and macrophages, while focusing on the current evidence on the health benefits of flavones against obesity and chronic inflammation, which has been positively correlated with an enhanced cancer incidence. We conclude the review by highlighting the areas of research warranting a deeper investigation, with an emphasis on flavones and their potential impact on the crosstalk between adipocytes, the immune system, the gut microbiome, and their role in the regulation of obesity.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Andrea I. Doseff
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
365
|
Hardikar S, Albrechtsen RD, Achaintre D, Lin T, Pauleck S, Playdon M, Holowatyj AN, Gigic B, Schrotz-King P, Boehm J, Habermann N, Brezina S, Gsur A, van Roekel EH, Weijenberg MP, Keski-Rahkonen P, Scalbert A, Ose J, Ulrich CM. Impact of Pre-blood Collection Factors on Plasma Metabolomic Profiles. Metabolites 2020; 10:E213. [PMID: 32455751 PMCID: PMC7281389 DOI: 10.3390/metabo10050213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/30/2022] Open
Abstract
Demographic, lifestyle and biospecimen-related factors at the time of blood collection can influence metabolite levels in epidemiological studies. Identifying the major influences on metabolite concentrations is critical to designing appropriate sample collection protocols and considering covariate adjustment in metabolomics analyses. We examined the association of age, sex, and other short-term pre-blood collection factors (time of day, season, fasting duration, physical activity, NSAID use, smoking and alcohol consumption in the days prior to collection) with 133 targeted plasma metabolites (acylcarnitines, amino acids, biogenic amines, sphingolipids, glycerophospholipids, and hexoses) among 108 individuals that reported exposures within 48 h before collection. The differences in mean metabolite concentrations were assessed between groups based on pre-collection factors using two-sided t-tests and ANOVA with FDR correction. Percent differences in metabolite concentrations were negligible across season, time of day of collection, fasting status or lifestyle behaviors at the time of collection, including physical activity or the use of tobacco, alcohol or NSAIDs. The metabolites differed in concentration between the age and sex categories for 21.8% and 14.3% metabolites, respectively. In conclusion, extrinsic factors in the short period prior to collection were not meaningfully associated with concentrations of selected endogenous metabolites in a cross-sectional sample, though metabolite concentrations differed by age and sex. Larger studies with more coverage of the human metabolome are warranted.
Collapse
Affiliation(s)
- Sheetal Hardikar
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84108, USA
- Cancer Prevention, Population Health Sciences, Fred Hutchinson Cancer Research Institute, Seattle, WA 19024, USA
| | - Richard D. Albrechtsen
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
| | - David Achaintre
- International Agency for Research on Cancer, 69372 Lyon, France; (D.A.); (P.K.-R.); (A.S.)
| | - Tengda Lin
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84108, USA
| | - Svenja Pauleck
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
| | - Mary Playdon
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84108, USA
| | - Andreana N. Holowatyj
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84108, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA
| | - Biljana Gigic
- Department of Surgery, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Petra Schrotz-King
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (P.S.-K.); (N.H.)
| | - Juergen Boehm
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
| | - Nina Habermann
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (P.S.-K.); (N.H.)
- Genome Biology, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Stefanie Brezina
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (S.B.); (A.G.)
| | - Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (S.B.); (A.G.)
| | - Eline H. van Roekel
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, 6211 LK Maastricht, The Netherlands; (E.H.v.R.); (M.P.W.)
| | - Matty P. Weijenberg
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, 6211 LK Maastricht, The Netherlands; (E.H.v.R.); (M.P.W.)
| | - Pekka Keski-Rahkonen
- International Agency for Research on Cancer, 69372 Lyon, France; (D.A.); (P.K.-R.); (A.S.)
| | - Augustin Scalbert
- International Agency for Research on Cancer, 69372 Lyon, France; (D.A.); (P.K.-R.); (A.S.)
| | - Jennifer Ose
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84108, USA
| | - Cornelia M. Ulrich
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA; (R.D.A.); (T.L.); (S.P.); (M.P.); (A.N.H.); (J.B.); (J.O.); (C.M.U.)
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84108, USA
| |
Collapse
|
366
|
Relationship between dementia and gut microbiome-associated metabolites: a cross-sectional study in Japan. Sci Rep 2020; 10:8088. [PMID: 32424166 PMCID: PMC7235213 DOI: 10.1038/s41598-020-65196-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 04/30/2020] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of the gut microbiome is associated with dementia. However, the relationship between microbiome-associated metabolites and dementia has yet to be identified. Outpatients visiting a memory clinic in Japan enrolled in this cross-sectional study; 107 subjects were eligible for the study, 25 of which had dementia. We collected demographics, activities of daily living, risk factors, cognitive function, and brain imaging data. The gut microbiome was assessed using terminal restriction fragment length polymorphism analysis. Concentrations of faecal metabolite were measured. We used multivariable logistic regression analyses to identify whether metabolites were independently related to dementia. The concentrations of metabolites were significantly different between subjects with and those without dementia. Every 1 standard deviation increment in faecal ammonia concentration was associated with around a 1.6-fold risk for the presence of dementia. A higher faecal lactic acid concentration was related to a lower risk of dementia, by around 60%. A combination of higher faecal ammonia and lactic acid concentrations was indicative of the presence of dementia, and had a similar predictive value as traditional biomarkers of dementia. Thus, faecal ammonia and lactic acid are related to dementia, independently of the other risk factors for dementia and dysregulation of the gut microbiome.
Collapse
|
367
|
Mohr AE, Jäger R, Carpenter KC, Kerksick CM, Purpura M, Townsend JR, West NP, Black K, Gleeson M, Pyne DB, Wells SD, Arent SM, Kreider RB, Campbell BI, Bannock L, Scheiman J, Wissent CJ, Pane M, Kalman DS, Pugh JN, Ortega-Santos CP, Ter Haar JA, Arciero PJ, Antonio J. The athletic gut microbiota. J Int Soc Sports Nutr 2020; 17:24. [PMID: 32398103 PMCID: PMC7218537 DOI: 10.1186/s12970-020-00353-w] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
The microorganisms in the gastrointestinal tract play a significant role in nutrient uptake, vitamin synthesis, energy harvest, inflammatory modulation, and host immune response, collectively contributing to human health. Important factors such as age, birth method, antibiotic use, and diet have been established as formative factors that shape the gut microbiota. Yet, less described is the role that exercise plays, particularly how associated factors and stressors, such as sport/exercise-specific diet, environment, and their interactions, may influence the gut microbiota. In particular, high-level athletes offer remarkable physiology and metabolism (including muscular strength/power, aerobic capacity, energy expenditure, and heat production) compared to sedentary individuals, and provide unique insight in gut microbiota research. In addition, the gut microbiota with its ability to harvest energy, modulate the immune system, and influence gastrointestinal health, likely plays an important role in athlete health, wellbeing, and sports performance. Therefore, understanding the mechanisms in which the gut microbiota could play in the role of influencing athletic performance is of considerable interest to athletes who work to improve their results in competition as well as reduce recovery time during training. Ultimately this research is expected to extend beyond athletics as understanding optimal fitness has applications for overall health and wellness in larger communities. Therefore, the purpose of this narrative review is to summarize current knowledge of the athletic gut microbiota and the factors that shape it. Exercise, associated dietary factors, and the athletic classification promote a more “health-associated” gut microbiota. Such features include a higher abundance of health-promoting bacterial species, increased microbial diversity, functional metabolic capacity, and microbial-associated metabolites, stimulation of bacterial abundance that can modulate mucosal immunity, and improved gastrointestinal barrier function.
Collapse
Affiliation(s)
- Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA.
| | - Ralf Jäger
- Increnovo LLC, Milwaukee, WI, 53202, USA
| | | | - Chad M Kerksick
- Exercise and Performance Nutrition Laboratory, School of Health Sciences, Lindenwood University, St. Charles, MO, USA
| | | | - Jeremy R Townsend
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, TN, 37204, USA
| | - Nicholas P West
- School of Medical Research and Menzies Health Institute of QLD, Griffith Health, Griffith University, Southport, Australia
| | - Katherine Black
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Michael Gleeson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - David B Pyne
- Research Institute for Sport and Exercise, University of Canberra, Canberra, ACT 2617, Australia
| | | | - Shawn M Arent
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Richard B Kreider
- Exercise & Sport Nutrition Lab, Human Clinical Research Facility, Department of Health & Kinesiology, Texas A&M University, College Station, TX, 77843-4253, USA
| | - Bill I Campbell
- Performance & Physique Enhancement Laboratory, University of South Florida, Tampa, FL, USA
| | | | | | - Craig J Wissent
- Jamieson Wellness Inc., 4025 Rhodes Drive, Windsor, Ontario, N8W 5B5, Canada
| | - Marco Pane
- Bioloab Research, Via E. Mattei 3, 28100, Novara, Italy
| | - Douglas S Kalman
- Scientific Affairs, Nutrasource Diagnostics, Inc. Guelph, Guelph, Ontario, Canada
| | - Jamie N Pugh
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom St Campus, Liverpool, L3 3AF, UK
| | | | | | - Paul J Arciero
- Health and Human Physiological Sciences Department, Skidmore College, Saratoga Springs, NY, USA
| | - Jose Antonio
- Exercise and Sport Science, Nova Southeastern University, Davie, FL, USA
| |
Collapse
|
368
|
Sata Y, Marques FZ, Kaye DM. The Emerging Role of Gut Dysbiosis in Cardio-metabolic Risk Factors for Heart Failure. Curr Hypertens Rep 2020; 22:38. [DOI: 10.1007/s11906-020-01046-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
369
|
Administration of Bifidobacterium bifidum CGMCC 15068 modulates gut microbiota and metabolome in azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced colitis-associated colon cancer (CAC) in mice. Appl Microbiol Biotechnol 2020; 104:5915-5928. [PMID: 32367312 DOI: 10.1007/s00253-020-10621-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/31/2022]
Abstract
The gut microbiota plays an important role in colorectal cancer (CRC), and the use of probiotics might be a promising intervention method. The aim of our study was to investigate the beneficial effect of Bifidobacterium bifidum CGMCC 15068 on an azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced colitis-associated CRC (CAC) mouse model. CAC was induced by an intra-peritoneal injection of AOM (10 mg/kg) and three 7-day cycles of 2% DSS in drinking water with a 14-day recovery period between two consecutive DSS administrations. B. bifidum CGMCC 15068 (3 × 109 CFU/mL) was gavaged once daily during the recovery period. Then, the faecal microbial composition and metabolome were profiled using the 16S rRNA sequencing technology and gas chromatography-mass spectrometry (GC-MS), respectively. The administration of B. bifidum CGMCC 15068 attenuated tumourigenesis in the CAC mouse model. In addition, B. bifidum CGMCC 15068 pre-treatment increased the relative abundance of Akkermansia, Desulfovibrionaceae, Romboutsia, Turicibacter, Verrucomicrobiaceae, Ruminococcaceae_UCG_013, Lachnospiraceae_UCG_004, and Lactobacillus. Meanwhile, B. bifidum CGMCC 15068 altered metabolites involved in the citrate cycle (TCA cycle), glycolysis, butyrate metabolism, fatty acid biosynthesis, and galactose metabolism. Several significant correlations were identified between the differentially abundant microbes and metabolites. These findings supported the beneficial role of B. bifidum CGMCC 15068 in intestinal health by modulating dysbiosis and the gut metabolic profile. The manipulation of the gut microbial composition using probiotics might be a promising prevention strategy for CRC. Long-term and large-scale clinical trials are warranted for the potential clinical applications of this strategy in the future.
Collapse
|
370
|
Lee-Sarwar KA, Lasky-Su J, Kelly RS, Litonjua AA, Weiss ST. Metabolome-Microbiome Crosstalk and Human Disease. Metabolites 2020; 10:E181. [PMID: 32369899 PMCID: PMC7281736 DOI: 10.3390/metabo10050181] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/23/2020] [Accepted: 04/29/2020] [Indexed: 12/25/2022] Open
Abstract
In this review, we discuss the growing literature demonstrating robust and pervasive associations between the microbiome and metabolome. We focus on the gut microbiome, which harbors the taxonomically most diverse and the largest collection of microorganisms in the human body. Methods for integrative analysis of these "omics" are under active investigation and we discuss the advances and challenges in the combined use of metabolomics and microbiome data. Findings from large consortia, including the Human Microbiome Project and Metagenomics of the Human Intestinal Tract (MetaHIT) and others demonstrate the impact of microbiome-metabolome interactions on human health. Mechanisms whereby the microbes residing in the human body interact with metabolites to impact disease risk are beginning to be elucidated, and discoveries in this area will likely be harnessed to develop preventive and treatment strategies for complex diseases.
Collapse
Affiliation(s)
- Kathleen A. Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (J.L.-S.); (R.S.K.); (S.T.W.)
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (J.L.-S.); (R.S.K.); (S.T.W.)
| | - Rachel S. Kelly
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (J.L.-S.); (R.S.K.); (S.T.W.)
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children’s Hospital at Strong, University of Rochester Medical Center, Rochester, NY 14612, USA;
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (J.L.-S.); (R.S.K.); (S.T.W.)
| |
Collapse
|
371
|
Boland ML, Laker RC, Mather K, Nawrocki A, Oldham S, Boland BB, Lewis H, Conway J, Naylor J, Guionaud S, Feigh M, Veidal SS, Lantier L, McGuinness OP, Grimsby J, Rondinone CM, Jermutus L, Larsen MR, Trevaskis JL, Rhodes CJ. Resolution of NASH and hepatic fibrosis by the GLP-1R/GcgR dual-agonist Cotadutide via modulating mitochondrial function and lipogenesis. Nat Metab 2020; 2:413-431. [PMID: 32478287 PMCID: PMC7258337 DOI: 10.1038/s42255-020-0209-6] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-alcoholic fatty liver disease and steatohepatitis are highly associated with obesity and type 2 diabetes mellitus. Cotadutide, a GLP-1R/GcgR agonist, was shown to reduce blood glycemia, body weight and hepatic steatosis in patients with T2DM. Here, we demonstrate that the effects of Cotadutide to reduce body weight, food intake and improve glucose control are predominantly mediated through the GLP-1 signaling, while, its action on the liver to reduce lipid content, drive glycogen flux and improve mitochondrial turnover and function are directly mediated through Gcg signaling. This was confirmed by the identification of phosphorylation sites on key lipogenic and glucose metabolism enzymes in liver of mice treated with Cotadutide. Complementary metabolomic and transcriptomic analyses implicated lipogenic, fibrotic and inflammatory pathways, which are consistent with a unique therapeutic contribution of GcgR agonism by Cotadutide in vivo. Significantly, Cotadutide also alleviated fibrosis to a greater extent than Liraglutide or Obeticholic acid (OCA), despite adjusting dose to achieve similar weight loss in 2 preclinical mouse models of NASH. Thus Cotadutide, via direct hepatic (GcgR) and extra-hepatic (GLP-1R) effects, exerts multi-factorial improvement in liver function and is a promising therapeutic option for the treatment of steatohepatitis.
Collapse
Affiliation(s)
- Michelle L Boland
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Rhianna C Laker
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Karly Mather
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Arkadiusz Nawrocki
- Department of Biochemistry and Molecular Biology, PR group, University of Southern Denmark, Odense, Denmark
| | - Stephanie Oldham
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Brandon B Boland
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Hilary Lewis
- Research and Early Development, Oncology, AstraZeneca, Cambridge, UK
| | - James Conway
- Translational Sciences, AstraZeneca, Gaithersburg, MD, USA
| | - Jacqueline Naylor
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | | | - Louise Lantier
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Owen P McGuinness
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Joseph Grimsby
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Cristina M Rondinone
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Lutz Jermutus
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, PR group, University of Southern Denmark, Odense, Denmark
| | - James L Trevaskis
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
- Gilead Sciences, Foster City, CA, USA
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| |
Collapse
|
372
|
Palmnäs M, Brunius C, Shi L, Rostgaard-Hansen A, Torres NE, González-Domínguez R, Zamora-Ros R, Ye YL, Halkjær J, Tjønneland A, Riccardi G, Giacco R, Costabile G, Vetrani C, Nielsen J, Andres-Lacueva C, Landberg R. Perspective: Metabotyping-A Potential Personalized Nutrition Strategy for Precision Prevention of Cardiometabolic Disease. Adv Nutr 2020; 11:524-532. [PMID: 31782487 PMCID: PMC7231594 DOI: 10.1093/advances/nmz121] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/26/2019] [Accepted: 10/14/2019] [Indexed: 12/22/2022] Open
Abstract
Diet is an important, modifiable lifestyle factor of cardiometabolic disease risk, and an improved diet can delay or even prevent the onset of disease. Recent evidence suggests that individuals could benefit from diets adapted to their genotype and phenotype: that is, personalized nutrition. A novel strategy is to tailor diets for groups of individuals according to their metabolic phenotypes (metabotypes). Randomized controlled trials evaluating metabotype-specific responses and nonresponses are urgently needed to bridge the current gap of knowledge with regard to the efficacy of personalized strategies in nutrition. In this Perspective, we discuss the concept of metabotyping, review the current literature on metabotyping in the context of cardiometabolic disease prevention, and suggest potential strategies for metabotype-based nutritional advice for future work. We also discuss potential determinants of metabotypes, including gut microbiota, and highlight the use of metabolomics to define effective markers for cardiometabolic disease-related metabotypes. Moreover, we hypothesize that people at high risk for cardiometabolic diseases have distinct metabotypes and that individuals grouped into specific metabotypes may respond differently to the same diet, which is being tested in a project of the Joint Programming Initiative: A Healthy Diet for a Healthy Life.
Collapse
Affiliation(s)
- Marie Palmnäs
- Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Carl Brunius
- Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Lin Shi
- Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Agneta Rostgaard-Hansen
- Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
- Diet, Genes, and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Núria Estanyol Torres
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences, and Gastronomy, Institute for Research on Nutrition and Food Safety, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Centro de Investigacion Biomedica en Red (CIBER) of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Barcelona, Spain
| | - Raúl González-Domínguez
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences, and Gastronomy, Institute for Research on Nutrition and Food Safety, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Centro de Investigacion Biomedica en Red (CIBER) of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Barcelona, Spain
| | - Raul Zamora-Ros
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences, and Gastronomy, Institute for Research on Nutrition and Food Safety, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Prgramme, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de LLobregat, Barcelona, Spain
| | - Ye Lingqun Ye
- Department of Biology and Biological Engineering, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
| | - Jytte Halkjær
- Diet, Genes, and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anne Tjønneland
- Diet, Genes, and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Gabriele Riccardi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rosalba Giacco
- Institute of Food Science, Italian National Research Council, Avellino, Italy
| | - Giuseppina Costabile
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Claudia Vetrani
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences, and Gastronomy, Institute for Research on Nutrition and Food Safety, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Centro de Investigacion Biomedica en Red (CIBER) of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Barcelona, Spain
| | - Rikard Landberg
- Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
373
|
Troll M, Brandmaier S, Reitmeier S, Adam J, Sharma S, Sommer A, Bind MA, Neuhaus K, Clavel T, Adamski J, Haller D, Peters A, Grallert H. Investigation of Adiposity Measures and Operational Taxonomic unit (OTU) Data Transformation Procedures in Stool Samples from a German Cohort Study Using Machine Learning Algorithms. Microorganisms 2020; 8:E547. [PMID: 32290101 PMCID: PMC7232268 DOI: 10.3390/microorganisms8040547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 01/14/2023] Open
Abstract
The analysis of the gut microbiome with respect to health care prevention and diagnostic purposes is increasingly the focus of current research. We analyzed around 2000 stool samples from the KORA (Cooperative Health Research in the Region of Augsburg) cohort using high-throughput 16S rRNA gene amplicon sequencing representing a total microbial diversity of 2089 operational taxonomic units (OTUs). We evaluated the combination of three different components to assess the reflection of obesity related to microbiota profiles: (i) four prediction methods (i.e., partial least squares (PLS), support vector machine regression (SVMReg), random forest (RF), and M5Rules); (ii) five OTU data transformation approaches (i.e., no transformation, relative abundance without and with log-transformation, as well as centered and isometric log-ratio transformations); and (iii) predictions from nine measurements of obesity (i.e., body mass index, three measures of body shape, and five measures of body composition). Our results showed a substantial impact of all three components. The applications of SVMReg and PLS in combination with logarithmic data transformations resulted in considerably predictive models for waist circumference-related endpoints. These combinations were at best able to explain almost 40% of the variance in obesity measurements based on stool microbiota data (i.e., OTUs) only. A reduced loss in predictive performance was seen after sex-stratification in waist-height ratio compared to other waist-related measurements. Moreover, our analysis showed that the contribution of OTUs less prevalent and abundant is minor concerning the predictive power of our models.
Collapse
Affiliation(s)
- Martina Troll
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (S.B.); (J.A.); (S.S.)
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.S.); (A.P.)
| | - Stefan Brandmaier
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (S.B.); (J.A.); (S.S.)
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.S.); (A.P.)
| | - Sandra Reitmeier
- ZIEL Institute for Food & Health, Technical University of Munich, 85354 Freising-Weihenstephan, Germany; (S.R.); (K.N.); (T.C.); (D.H.)
- Chair of Nutrition and Immunology, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
| | - Jonathan Adam
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (S.B.); (J.A.); (S.S.)
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.S.); (A.P.)
| | - Sapna Sharma
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (S.B.); (J.A.); (S.S.)
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.S.); (A.P.)
| | - Alice Sommer
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.S.); (A.P.)
- Department of Statistics, Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138-2901, USA;
| | - Marie-Abèle Bind
- Department of Statistics, Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138-2901, USA;
| | - Klaus Neuhaus
- ZIEL Institute for Food & Health, Technical University of Munich, 85354 Freising-Weihenstephan, Germany; (S.R.); (K.N.); (T.C.); (D.H.)
| | - Thomas Clavel
- ZIEL Institute for Food & Health, Technical University of Munich, 85354 Freising-Weihenstephan, Germany; (S.R.); (K.N.); (T.C.); (D.H.)
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, 52074 Aachen, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, 85764 Neuherberg, Germany;
- Chair of Experimental Genetics, Technical University of Munich, 85350 Freising-Weihenstephan, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Dirk Haller
- ZIEL Institute for Food & Health, Technical University of Munich, 85354 Freising-Weihenstephan, Germany; (S.R.); (K.N.); (T.C.); (D.H.)
- Chair of Nutrition and Immunology, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.S.); (A.P.)
- Chair of Epidemiology, Faculty of Medicine, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (S.B.); (J.A.); (S.S.)
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.S.); (A.P.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|
374
|
Noerman S, Kolehmainen M, Hanhineva K. Profiling of Endogenous and Gut Microbial Metabolites to Indicate Metabotype-Specific Dietary Responses: A Systematic Review. Adv Nutr 2020; 11:1237-1254. [PMID: 32271864 PMCID: PMC7490160 DOI: 10.1093/advances/nmaa031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/21/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
Upon dietary exposure, the endogenous metabolism responds to the diet-derived nutrients and bioactive compounds, such as phytochemicals. However, the responses vary remarkably due to the interplay with other dietary components, lifestyle exposures, and intrinsic factors, which lead to differences in endogenous regulatory metabolism. These physiological processes are evidenced as a signature profile composed of various metabolites constituting metabolic phenotypes, or metabotypes. The metabolic profiling of biological samples following dietary intake hence would provide information about diet-that is, as the intake biomarkers and the ongoing physiological reactions triggered by this intake-thereby enable evaluation of the metabolic basis required to distinguish the different metabotypes. The capacity of nontargeted metabolomics to also encompass the unprecedented metabolite species has enabled the profiling of multiple metabolites and the corresponding metabotypes with a single analysis, decoding the complex interplay between diet, other relevant factors, and health. In this systematic review, we screened 345 articles published in English in January 2007-July 2018, which applied the metabolomics approach to profile the changes of endogenous metabolites in the blood related to dietary interventions, either derived by metabolism of gut microbiota or the human host. We excluded all the compounds that were directly derived from diet, and also the dietary interventions focusing on supplementation with individual compounds. After the removal of less relevant studies and assessment of eligibility, 49 articles were included in this review. First, we mention the contribution of individual factors, either modifiable or nonmodifiable factors, in shaping metabolic profile. Then, how different aspects of the diet would affect the metabolic profiles are disentangled. Next, the classes of endogenous metabolites altered following included dietary interventions are listed. We also discuss the current challenges in the field, along with future research opportunities.
Collapse
Affiliation(s)
- Stefania Noerman
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland,Address correspondence to SN (e-mail: )
| | - Marjukka Kolehmainen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Kati Hanhineva
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland,Address correspondence to KH ()
| |
Collapse
|
375
|
Cheng K, Brunius C, Fristedt R, Landberg R. An LC-QToF MS based method for untargeted metabolomics of human fecal samples. Metabolomics 2020; 16:46. [PMID: 32246267 PMCID: PMC7125068 DOI: 10.1007/s11306-020-01669-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/23/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Consensus in sample preparation for untargeted human fecal metabolomics is lacking. OBJECTIVES To obtain sample preparation with broad metabolite coverage for high-throughput LC-MS. METHODS Extraction solvent, solvent ratio and fresh frozen-vs-lyophilized samples were evaluated by metabolite feature quality. RESULTS Methanol at 5 mL per g wet feces provided a wide metabolite coverage with optimal balance between signal intensity and saturation for both fresh frozen and lyophilized samples. Lyophilization did not affect SCFA and is recommended because of convenience in normalizing to dry matter. CONCLUSION The suggested sample preparation is simple, efficient and suitable for large-scale human fecal metabolomics.
Collapse
Affiliation(s)
- Ken Cheng
- Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden.
| | - Carl Brunius
- Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Rikard Fristedt
- Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden.
| |
Collapse
|
376
|
Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2020; 17:223-237. [PMID: 32076145 DOI: 10.1038/s41575-019-0258-z] [Citation(s) in RCA: 1048] [Impact Index Per Article: 209.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2019] [Indexed: 02/06/2023]
Abstract
A key role of the gut microbiota in the establishment and maintenance of health, as well as in the pathogenesis of disease, has been identified over the past two decades. One of the primary modes by which the gut microbiota interacts with the host is by means of metabolites, which are small molecules that are produced as intermediate or end products of microbial metabolism. These metabolites can derive from bacterial metabolism of dietary substrates, modification of host molecules, such as bile acids, or directly from bacteria. Signals from microbial metabolites influence immune maturation, immune homeostasis, host energy metabolism and maintenance of mucosal integrity. Alterations in the composition and function of the microbiota have been described in many studies on IBD. Alterations have also been described in the metabolite profiles of patients with IBD. Furthermore, specific classes of metabolites, notably bile acids, short-chain fatty acids and tryptophan metabolites, have been implicated in the pathogenesis of IBD. This Review aims to define the key classes of microbial-derived metabolites that are altered in IBD, describe the pathophysiological basis of these associations and identify future targets for precision therapeutic modulation.
Collapse
|
377
|
Nag A, Kurushima Y, Bowyer RCE, Wells PM, Weiss S, Pietzner M, Kocher T, Raffler J, Völker U, Mangino M, Spector TD, Milburn MV, Kastenmüller G, Mohney RP, Suhre K, Menni C, Steves CJ. Genome-wide scan identifies novel genetic loci regulating salivary metabolite levels. Hum Mol Genet 2020; 29:864-875. [PMID: 31960908 PMCID: PMC7104674 DOI: 10.1093/hmg/ddz308] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/05/2019] [Accepted: 12/11/2019] [Indexed: 12/26/2022] Open
Abstract
Saliva, as a biofluid, is inexpensive and non-invasive to obtain, and provides a vital tool to investigate oral health and its interaction with systemic health conditions. There is growing interest in salivary biomarkers for systemic diseases, notably cardiovascular disease. Whereas hundreds of genetic loci have been shown to be involved in the regulation of blood metabolites, leading to significant insights into the pathogenesis of complex human diseases, little is known about the impact of host genetics on salivary metabolites. Here we report the first genome-wide association study exploring 476 salivary metabolites in 1419 subjects from the TwinsUK cohort (discovery phase), followed by replication in the Study of Health in Pomerania (SHIP-2) cohort. A total of 14 distinct locus-metabolite associations were identified in the discovery phase, most of which were replicated in SHIP-2. While only a limited number of the loci that are known to regulate blood metabolites were also associated with salivary metabolites in our study, we identified several novel saliva-specific locus-metabolite associations, including associations for the AGMAT (with the metabolites 4-guanidinobutanoate and beta-guanidinopropanoate), ATP13A5 (with the metabolite creatinine) and DPYS (with the metabolites 3-ureidopropionate and 3-ureidoisobutyrate) loci. Our study suggests that there may be regulatory pathways of particular relevance to the salivary metabolome. In addition, some of our findings may have clinical significance, such as the utility of the pyrimidine (uracil) degradation metabolites in predicting 5-fluorouracil toxicity and the role of the agmatine pathway metabolites as biomarkers of oral health.
Collapse
Affiliation(s)
- Abhishek Nag
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Yuko Kurushima
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Ruth C E Bowyer
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Philippa M Wells
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Stefan Weiss
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald & University of Greifswald, 17489 Greifswald, Germany
| | - Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald & University of Greifswald, 17489 Greifswald, Germany
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Michael V Milburn
- Discovery and Translational Sciences, Metabolon, Inc., Morrisville, NC 27560, USA
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Robert P Mohney
- Discovery and Translational Sciences, Metabolon, Inc., Morrisville, NC 27560, USA
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| |
Collapse
|
378
|
Petersen C, Bell R, Klag KA, Lee SH, Soto R, Ghazaryan A, Buhrke K, Ekiz HA, Ost KS, Boudina S, O'Connell RM, Cox JE, Villanueva CJ, Stephens WZ, Round JL. T cell-mediated regulation of the microbiota protects against obesity. Science 2020; 365:365/6451/eaat9351. [PMID: 31346040 DOI: 10.1126/science.aat9351] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 02/22/2019] [Accepted: 06/13/2019] [Indexed: 12/16/2022]
Abstract
The microbiota influences obesity, yet organisms that protect from disease remain unknown. During studies interrogating host-microbiota interactions, we observed the development of age-associated metabolic syndrome (MetS). Expansion of Desulfovibrio and loss of Clostridia were key features associated with obesity in this model and are present in humans with MetS. T cell-dependent events were required to prevent disease, and replacement of Clostridia rescued obesity. Inappropriate immunoglobulin A targeting of Clostridia and increased Desulfovibrio antagonized the colonization of beneficial Clostridia. Transcriptional and metabolic analysis revealed enhanced lipid absorption in the obese host. Colonization of germ-free mice with Clostridia, but not Desulfovibrio, down-regulated genes that control lipid absorption and reduced adiposity. Thus, immune control of the microbiota maintains beneficial microbial populations that constrain lipid metabolism to prevent MetS.
Collapse
Affiliation(s)
- Charisse Petersen
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Rickesha Bell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Kendra A Klag
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Soh-Hyun Lee
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Raymond Soto
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Arevik Ghazaryan
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Kaitlin Buhrke
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - H Atakan Ekiz
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Kyla S Ost
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - James E Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Claudio J Villanueva
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - W Zac Stephens
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
379
|
Nontargeted fecal metabolomics: an emerging tool to probe the role of the gut microbiome in host health. Bioanalysis 2020; 12:351-353. [PMID: 32209031 DOI: 10.4155/bio-2020-0010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
380
|
Gong S, Ye T, Wang M, Wang M, Li Y, Ma L, Yang Y, Wang Y, Zhao X, Liu L, Yang M, Chen H, Qian J. Traditional Chinese Medicine Formula Kang Shuai Lao Pian Improves Obesity, Gut Dysbiosis, and Fecal Metabolic Disorders in High-Fat Diet-Fed Mice. Front Pharmacol 2020; 11:297. [PMID: 32269525 PMCID: PMC7109517 DOI: 10.3389/fphar.2020.00297] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/27/2020] [Indexed: 12/19/2022] Open
Abstract
High-fat diet (HFD)-induced obesity is a risk factor for many metabolic disorders including cardiovascular diseases, diabetes, and fatty liver disease. Although there are accumulating evidences supporting the assumption that regulating gut microbiota as well as its metabolic status is able to mitigate obesity, the inner relationship between the obesity-related gut microbiota and the relevant metabolites are not well defined. In current study, we applied a traditional herbal formula Kang Shuai Lao Pian (KSLP) to HFD-fed mice and evaluated its effect against obesity. Emphases were addressed on identifying profiles of gut microbiota and fecal metabolites with the aid of 16S rRNA gene sequencing and non-target fecal metabolomics techniques. We showed that KSLP could improve HFD-induced obesity, glucose tolerance disorder, as well as gut dysbiosis. In the gut, KSLP corrected the increased abundance of Firmicutes and Proteobacteria, increased ratio of Firmicutes/Bacteroidetes, and decreased abundance of Bacteroidetes caused by HFD. KSLP also reversed HFD-induced significant changes in the abundance of certain genus including Intestinimonas, Oscillibacter, Christensenellaceae_R-7_group, Ruminococcaceae_UCG-010, and Aliihoeflea. Pearson correlation analysis indicated that except for Ruminococcaceae_UCG-010, other four genera had positive correlations with obesity. In addition, 22 key fecal metabolites responding to KSLP treatment were identified. Pearson correlation analysis showed that those metabolites are intimately related to KSLP effective genera of Intestinimonas, Oscillibacter, and Christensenellaceae_R-7_group. Our results indicate that KSLP is a promising traditional Chinese medicine (TCM) applicable for individuals with HFD habit. Intestinimonas, Oscillibacter, and Christensenellaceae_R-7_group might be responsible for the regulatory effect of KSLP. Linking of obesity phenotypes with gut microbiota as well as fecal metabolites is therefore a powerful research strategy to reveal the mechanism of obesity and the targets of intervention.
Collapse
Affiliation(s)
- Shuqing Gong
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Tingting Ye
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meixia Wang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, China.,NMPA Key Laboratory for Testing and Risk Warning of Pharmaceutical Microbiology, Zhejiang Institute of Microbiology, Hangzhou, China
| | - Mengying Wang
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yufei Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lina Ma
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yulian Yang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoping Zhao
- College of Preclinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Liu
- Technical Center, Chiatai Qingchunbao Pharmaceutical Co. Ltd, Hangzhou, China
| | - Min Yang
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Huan Chen
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, China.,NMPA Key Laboratory for Testing and Risk Warning of Pharmaceutical Microbiology, Zhejiang Institute of Microbiology, Hangzhou, China
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
381
|
Important Considerations for Sample Collection in Metabolomics Studies with a Special Focus on Applications to Liver Functions. Metabolites 2020; 10:metabo10030104. [PMID: 32178364 PMCID: PMC7142637 DOI: 10.3390/metabo10030104] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/05/2020] [Accepted: 03/07/2020] [Indexed: 12/16/2022] Open
Abstract
Metabolomics has found numerous applications in the study of liver metabolism in health and disease. Metabolomics studies can be conducted in a variety of biological matrices ranging from easily accessible biofluids such as urine, blood or feces, to organs, tissues or even cells. Sample collection and storage are critical steps for which standard operating procedures must be followed. Inappropriate sample collection or storage can indeed result in high variability, interferences with instrumentation or degradation of metabolites. In this review, we will first highlight important general factors that should be considered when planning sample collection in the study design of metabolomic studies, such as nutritional status and circadian rhythm. Then, we will discuss in more detail the specific procedures that have been described for optimal pre-analytical handling of the most commonly used matrices (urine, blood, feces, tissues and cells).
Collapse
|
382
|
Shah RM, McKenzie EJ, Rosin MT, Jadhav SR, Gondalia SV, Rosendale D, Beale DJ. An Integrated Multi-Disciplinary Perspectivefor Addressing Challenges of the Human Gut Microbiome. Metabolites 2020; 10:E94. [PMID: 32155792 PMCID: PMC7143645 DOI: 10.3390/metabo10030094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
Our understanding of the human gut microbiome has grown exponentially. Advances in genome sequencing technologies and metagenomics analysis have enabled researchers to study microbial communities and their potential function within the context of a range of human gut related diseases and disorders. However, up until recently, much of this research has focused on characterizing the gut microbiological community structure and understanding its potential through system wide (meta) genomic and transcriptomic-based studies. Thus far, the functional output of these microbiomes, in terms of protein and metabolite expression, and within the broader context of host-gut microbiome interactions, has been limited. Furthermore, these studies highlight our need to address the issues of individual variation, and of samples as proxies. Here we provide a perspective review of the recent literature that focuses on the challenges of exploring the human gut microbiome, with a strong focus on an integrated perspective applied to these themes. In doing so, we contextualize the experimental and technical challenges of undertaking such studies and provide a framework for capitalizing on the breadth of insight such approaches afford. An integrated perspective of the human gut microbiome and the linkages to human health will pave the way forward for delivering against the objectives of precision medicine, which is targeted to specific individuals and addresses the issues and mechanisms in situ.
Collapse
Affiliation(s)
- Rohan M. Shah
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia;
- Land and Water, Commonwealth Scientific and Industrial Research Organization (CSIRO), Dutton Park, QLD 4102, Australia
| | - Elizabeth J. McKenzie
- Liggins Institute, The University of Auckland, Grafton, Auckland 1142, New Zealand; (E.J.M.); (M.T.R.)
| | - Magda T. Rosin
- Liggins Institute, The University of Auckland, Grafton, Auckland 1142, New Zealand; (E.J.M.); (M.T.R.)
| | - Snehal R. Jadhav
- Centre for Advanced Sensory Science, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC 3125, Australia;
| | - Shakuntla V. Gondalia
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia;
| | | | - David J. Beale
- Land and Water, Commonwealth Scientific and Industrial Research Organization (CSIRO), Dutton Park, QLD 4102, Australia
| |
Collapse
|
383
|
Abstract
AbstractKnowing the biological signals associated with appetite control is crucial for understanding the regulation of food intake. Biomarkers of appetite have been defined as physiological measures that relate to subjective appetite ratings, measured food intake, or both. Several metabolites including amino acids, lipids and glucose were proposed as key molecules associated with appetite control over 60 years ago, and along with bile acids are all among possible appetite biomarker candidates. Additional metabolites that have been associated with appetite include endocannabinoids, lactate, cortisol and β-hydroxybutyrate. However, although appetite is a complex integrative process, studies often investigated a limited number of markers in isolation. Metabolomics involves the study of small molecules or metabolites present in biological samples such as urine or blood, and may present a powerful approach to further the understanding of appetite control. Using multiple analytical techniques allows the characterisation of molecules, such as carbohydrates, lipids, amino acids, bile acids and fatty acids. Metabolomics has proven successful in identifying markers of consumption of certain foods and biomarkers implicated in several diseases. However, it has been underexploited in appetite control or obesity. The aim of the present narrative review is to: (1) provide an overview of existing metabolites that have been identified in human biofluids and associated with appetite control; and (2) discuss the potential of metabolomics to deepen understanding of appetite control in humans.
Collapse
|
384
|
Gray ID, Kross AR, Renfrew ME, Wood P. Precision Medicine in Lifestyle Medicine: The Way of the Future? Am J Lifestyle Med 2020; 14:169-186. [PMID: 32231483 PMCID: PMC7092395 DOI: 10.1177/1559827619834527] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/21/2018] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
Precision medicine has captured the imagination of the medical community with visions of therapies precisely targeted to the specific individual's genetic, biological, social, and environmental profile. However, in practice it has become synonymous with genomic medicine. As such its successes have been limited, with poor predictive or clinical value for the majority of people. It adds little to lifestyle medicine, other than in establishing why a healthy lifestyle is effective in combatting chronic disease. The challenge of lifestyle medicine remains getting people to actually adopt, sustain, and naturalize a healthy lifestyle, and this will require an approach that treats the patient as a person with individual needs and providing them with suitable types of support. The future of lifestyle medicine is holistic and person-centered rather than technological.
Collapse
Affiliation(s)
- Ian D. Gray
- Avondale College of Higher Education, Cooranbong,
New South Wales, Australia
| | - Andrea R. Kross
- Avondale College of Higher Education, Cooranbong,
New South Wales, Australia
| | - Melanie E. Renfrew
- Avondale College of Higher Education, Cooranbong,
New South Wales, Australia
| | - Paul Wood
- Avondale College of Higher Education, Cooranbong,
New South Wales, Australia
| |
Collapse
|
385
|
Abstract
Over the past decade, it has become exceedingly clear that the microbiome is a critical factor in human health and disease and thus should be investigated to develop innovative treatment strategies. The field of metagenomics has come a long way in leveraging the advances of next-generation sequencing technologies resulting in the capability to identify and quantify all microorganisms present in human specimens. However, the field of metagenomics is still in its infancy, specifically in regard to the limitations in computational analysis, statistical assessments, standardization, and validation due to vast variability in the cohorts themselves, experimental design, and bioinformatic workflows. This review summarizes the methods, technologies, computational tools, and model systems for characterizing and studying the microbiome. We also discuss important considerations investigators must make when interrogating the involvement of the microbiome in health and disease in order to establish robust results and mechanistic insights before moving into therapeutic design and intervention.
Collapse
|
386
|
Lee-Sarwar KA, Kelly RS, Lasky-Su J, Zeiger RS, O'Connor GT, Sandel MT, Bacharier LB, Beigelman A, Rifas-Shiman SL, Carey VJ, Harshfield BJ, Laranjo N, Gold DR, Weiss ST, Litonjua AA. Fecal short-chain fatty acids in pregnancy and offspring asthma and allergic outcomes. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2020; 8:1100-1102.e13. [PMID: 31494295 PMCID: PMC7056490 DOI: 10.1016/j.jaip.2019.08.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Kathleen A Lee-Sarwar
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass.
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Robert S Zeiger
- Departments of Allergy and Research and Evaluation, Kaiser Permanente Southern California, San Diego and Pasadena, Calif
| | - George T O'Connor
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Mass
| | - Megan T Sandel
- Department of Pediatrics, Boston Medical Center, Boston, Mass
| | - Leonard B Bacharier
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, Mo; St Louis Children's Hospital, St Louis, Mo
| | - Avraham Beigelman
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, Mo; St Louis Children's Hospital, St Louis, Mo
| | - Sheryl L Rifas-Shiman
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Mass
| | - Vincent J Carey
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Benjamin J Harshfield
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Nancy Laranjo
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Diane R Gold
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children's Hospital at Strong, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
387
|
Sharpton TJ, Combrink L, Arnold HK, Gaulke CA, Kent M. Harnessing the gut microbiome in the fight against anthelminthic drug resistance. Curr Opin Microbiol 2020; 53:26-34. [PMID: 32114334 DOI: 10.1016/j.mib.2020.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 01/08/2023]
Abstract
Intestinal helminth parasites present major challenges to the welfare of humans and threaten the global food supply. While the discovery of anthelminthic drugs empowered our ability to offset these harms to society, the alarming rise of anthelminthic drug resistance mitigates contemporary efforts to treat and control intestinal helminthic infections. Fortunately, emerging research points to potential opportunities to combat anthelminthic drug resistance by harnessing the gut microbiome as a resource for discovering novel therapeutics and informing responsible drug administration. In this review, we highlight research that demonstrates this potential and provide rationale to support increased investment in efforts to uncover and translationally utilize knowledge about how the gut microbiome mediates intestinal helminthic infection and its outcomes.
Collapse
Affiliation(s)
- Thomas J Sharpton
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA; Department of Statistics, Oregon State University, Corvallis, OR, 97331, USA.
| | - Leigh Combrink
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA; Department of Biomedical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Holly K Arnold
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA; Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, USA
| | | | - Michael Kent
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA; Department of Biomedical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
388
|
Integrated metabolome analysis reveals novel connections between maternal fecal metabolome and the neonatal blood metabolome in women with gestational diabetes mellitus. Sci Rep 2020; 10:3660. [PMID: 32107447 PMCID: PMC7046769 DOI: 10.1038/s41598-020-60540-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
Gestational Diabetes Mellitus (GDM), which is correlated with changes in the gut microbiota, is a risk factor for neonatal inborn errors of metabolism (IEMs). Maternal hyperglycemia exerts epigenetic effects on genes that encode IEM-associated enzymes, resulting in changes in the neonatal blood metabolome. However, the relationship between maternal gut microbiota and the neonatal blood metabolome remains poorly understood. This study aimed at understanding the connections between maternal gut microbiota and the neonatal blood metabolome in GDM. 1H-NMR-based untargeted metabolomics was performed on maternal fecal samples and targeted metabolomics on the matched neonatal dry blood spots from a cohort of 40 pregnant women, including 22 with GDM and 18 controls. Multi-omic association methods (including Co-Inertia Analysis and Procrustes Analysis) were applied to investigate the relationship between maternal fecal metabolome and the neonatal blood metabolome. Both maternal fecal metabolome and the matched neonatal blood metabolome could be separated along the vector of maternal hyperglycemia. A close relationship between the maternal and neonatal metabolomes was observed by multi-omic association approaches. Twelve out of thirty-two maternal fecal metabolites with altered abundances from 872 1H- NMR features (Bonferroni-adjusted P < 0.05) in women with GDM and the controls were identified, among which 8 metabolites contribute (P < 0.05 in a 999-step permutation test) to the close connection between maternal and the neonatal metabolomes in GDM. Four of these eight maternal fecal metabolites, including lysine, putrescine, guanidinoacetate, and hexadecanedioate, were negatively associated (Spearman rank correlation, coefficient value < −0.6, P < 0.05) with maternal hyperglycemia. Biotin metabolism was enriched (Bonferroni-adjusted P < 0.05 in the hypergeometric test) with the four-hyperglycemia associated fecal metabolites. The results of this study suggested that maternal fecal metabolites contribute to the connections between maternal fecal metabolome and the neonatal blood metabolome and may further affect the risk of IEMs.
Collapse
|
389
|
Bowers SJ, Vargas F, González A, He S, Jiang P, Dorrestein PC, Knight R, Wright KP, Lowry CA, Fleshner M, Vitaterna MH, Turek FW. Repeated sleep disruption in mice leads to persistent shifts in the fecal microbiome and metabolome. PLoS One 2020; 15:e0229001. [PMID: 32078624 PMCID: PMC7032712 DOI: 10.1371/journal.pone.0229001] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023] Open
Abstract
It has been established in recent years that the gut microbiome plays a role in health and disease, potentially via alterations in metabolites that influence host physiology. Although sleep disruption and gut dysbiosis have been associated with many of the same diseases, studies investigating the gut microbiome in the context of sleep disruption have yielded inconsistent results, and have not assessed the fecal metabolome. We exposed mice to five days of sleep disruption followed by four days of ad libitum recovery sleep, and assessed the fecal microbiome and fecal metabolome at multiple timepoints using 16S rRNA gene amplicons and untargeted LC-MS/MS mass spectrometry. We found global shifts in both the microbiome and metabolome in the sleep-disrupted group on the second day of recovery sleep, when most sleep parameters had recovered to baseline levels. We observed an increase in the Firmicutes:Bacteroidetes ratio, along with decreases in the genus Lactobacillus, phylum Actinobacteria, and genus Bifidobacterium in sleep-disrupted mice compared to control mice. The latter two taxa remained low at the fourth day post-sleep disruption. We also identified multiple classes of fecal metabolites that were differentially abundant in sleep-disrupted mice, some of which are physiologically relevant and commonly influenced by the microbiome. This included bile acids, and inference of microbial functional gene content suggested reduced levels of the microbial bile salt hydrolase gene in sleep-disrupted mice. Overall, this study adds to the evidence base linking disrupted sleep to the gut microbiome and expands it to the fecal metabolome, identifying sleep disruption-sensitive bacterial taxa and classes of metabolites that may serve as therapeutic targets to improve health after poor sleep.
Collapse
Affiliation(s)
- Samuel J. Bowers
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| | - Fernando Vargas
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Antonio González
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Shannon He
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Peng Jiang
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Pieter C. Dorrestein
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, United States of America
| | - Rob Knight
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, United States of America
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Kenneth P. Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
- Sleep and Chronobiology Laboratory, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Martha H. Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Fred W. Turek
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
390
|
Kim M, Vogtmann E, Ahlquist DA, Devens ME, Kisiel JB, Taylor WR, White BA, Hale VL, Sung J, Chia N, Sinha R, Chen J. Fecal Metabolomic Signatures in Colorectal Adenoma Patients Are Associated with Gut Microbiota and Early Events of Colorectal Cancer Pathogenesis. mBio 2020; 11:e03186-19. [PMID: 32071266 PMCID: PMC7029137 DOI: 10.1128/mbio.03186-19] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/10/2020] [Indexed: 12/16/2022] Open
Abstract
Colorectal adenomas are precancerous lesions of colorectal cancer (CRC) that offer a means of viewing the events key to early CRC development. A number of studies have investigated the changes and roles of gut microbiota in adenoma and carcinoma development, highlighting its impact on carcinogenesis. However, there has been less of a focus on the gut metabolome, which mediates interactions between the host and gut microbes. Here, we investigated metabolomic profiles of stool samples from patients with advanced adenoma (n = 102), matched controls (n = 102), and patients with CRC (n = 36). We found that several classes of bioactive lipids, including polyunsaturated fatty acids, secondary bile acids, and sphingolipids, were elevated in the adenoma patients compared to the controls. Most such metabolites showed directionally consistent changes in the CRC patients, suggesting that those changes may represent early events of carcinogenesis. We also examined gut microbiome-metabolome associations using gut microbiota profiles in these patients. We found remarkably strong overall associations between the microbiome and metabolome data and catalogued a list of robustly correlated pairs of bacterial taxa and metabolomic features which included signatures of adenoma. Our findings highlight the importance of gut metabolites, and potentially their interplay with gut microbes, in the early events of CRC pathogenesis.IMPORTANCE Colorectal adenomas are precursors of CRC. Recently, the gut microbiota, i.e., the collection of microbes residing in our gut, has been recognized as a key player in CRC development. There have been a number of gut microbiota profiling studies for colorectal adenoma and CRC; however, fewer studies have considered the gut metabolome, which serves as the chemical interface between the host and gut microbiota. Here, we conducted a gut metabolome profiling study of colorectal adenoma and CRC and analyzed the metabolomic profiles together with paired microbiota composition profiles. We found several chemical signatures of colorectal adenoma that were associated with some gut microbes and potentially indicative of future CRC. This study highlights potential early-driver metabolites in CRC pathogenesis and guides further targeted experiments and thus provides an important stepping stone toward developing better CRC prevention strategies.
Collapse
Affiliation(s)
- Minsuk Kim
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Surgical Research, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Emily Vogtmann
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - David A Ahlquist
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mary E Devens
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - John B Kisiel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - William R Taylor
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Bryan A White
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Vanessa L Hale
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Surgical Research, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Veterinary Preventive Medicine, The Ohio State University College of Veterinary Medicine, Columbus, Ohio, USA
| | - Jaeyun Sung
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Surgical Research, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Nicholas Chia
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Surgical Research, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Rashmi Sinha
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jun Chen
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
391
|
Mirzaei R, Mirzaei H, Alikhani MY, Sholeh M, Arabestani MR, Saidijam M, Karampoor S, Ahmadyousefi Y, Moghadam MS, Irajian GR, Hasanvand H, Yousefimashouf R. Bacterial biofilm in colorectal cancer: What is the real mechanism of action? Microb Pathog 2020; 142:104052. [PMID: 32045645 DOI: 10.1016/j.micpath.2020.104052] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
Human colorectal cancer is the third most common cancer around the world. Colorectal cancer has various risk factors, but current works have bolded a significant activity for the microbiota of the human colon in the development of this disease. Bacterial biofilm has been mediated to non-malignant pathologies like inflammatory bowel disease but has not been fully documented in the setting of colorectal cancer. The investigation has currently found that bacterial biofilm is mediated to colon cancer in the human and linked to the location of human cancer, with almost all right-sided adenomas of colon cancers possessing bacterial biofilm, whilst left-sided cancer is rarely biofilm positive. The profound comprehension of the changes in colorectal cancer can provide interesting novel concepts for anticancer treatments. In this review, we will summarize and examine the new knowledge about the links between colorectal cancer and bacterial biofilm.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Sholeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Shokri Moghadam
- Department of Microbiology, School of Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholam Reza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamze Hasanvand
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
392
|
Misra BB. The Connection and Disconnection Between Microbiome and Metabolome: A Critical Appraisal in Clinical Research. Biol Res Nurs 2020; 22:561-576. [PMID: 32013533 DOI: 10.1177/1099800420903083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Big data-driven omics research has led to a steep rise in investigations involving two of the most functional omes, the metabolome and microbiome. The former is touted as the closest to the phenotype, and the latter is implicated in general well-being and a plethora of human diseases. Although some research publications have integrated the concepts of the two domains, most focus their analyses on evidence solely originating from one or the other. With a growing interest in connecting the microbiome and metabolome in the context of disease, researchers must also appreciate the disconnect between the two domains. In the present review, drawing examples from the current literature, tools, and resources, I discuss the connections between the microbiome and metabolome and highlight challenges and opportunities in linking them together for the basic, translational, clinical, and nursing research communities.
Collapse
Affiliation(s)
- Biswapriya B Misra
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, 12279Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| |
Collapse
|
393
|
Botticelli A, Vernocchi P, Marini F, Quagliariello A, Cerbelli B, Reddel S, Del Chierico F, Di Pietro F, Giusti R, Tomassini A, Giampaoli O, Miccheli A, Zizzari IG, Nuti M, Putignani L, Marchetti P. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J Transl Med 2020; 18:49. [PMID: 32014010 PMCID: PMC6998840 DOI: 10.1186/s12967-020-02231-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Despite the efficacy of immune checkpoint inhibitors (ICIs) only the 20-30% of treated patients present long term benefits. The metabolic changes occurring in the gut microbiota metabolome are herein proposed as a factor potentially influencing the response to immunotherapy. METHODS The metabolomic profiling of gut microbiota was characterized in 11 patients affected by non-small cell lung cancer (NSCLC) treated with nivolumab in second-line treatment with anti-PD-1 nivolumab. The metabolomics analyses were performed by GC-MS/SPME and 1H-NMR in order to detect volatile and non-volatile metabolites. Metabolomic data were processed by statistical profiling and chemometric analyses. RESULTS Four out of 11 patients (36%) presented early progression, while the remaining 7 out of 11 (64%) presented disease progression after 12 months. 2-Pentanone (ketone) and tridecane (alkane) were significantly associated with early progression, and on the contrary short chain fatty acids (SCFAs) (i.e., propionate, butyrate), lysine and nicotinic acid were significantly associated with long-term beneficial effects. CONCLUSIONS Our preliminary data suggest a significant role of gut microbiota metabolic pathways in affecting response to immunotherapy. The metabolic approach could be a promising strategy to contribute to the personalized management of cancer patients by the identification of microbiota-linked "indicators" of early progressor and long responder patients.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,AOU Policlinico Umberto I, Rome, Italy
| | - Pamela Vernocchi
- Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Federico Marini
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-based Metabolomics Laboratory, Sapienza University of Rome, Rome, Italy
| | | | - Bruna Cerbelli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Sofia Reddel
- Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | | | | | - Alberta Tomassini
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-based Metabolomics Laboratory, Sapienza University of Rome, Rome, Italy
| | | | - Alfredo Miccheli
- NMR-based Metabolomics Laboratory, Sapienza University of Rome, Rome, Italy.,Department of Enviromental Biology, University of Rome, Rome, Italy
| | | | - Marianna Nuti
- Department of Experimental Medicine, University Sapienza, Rome, Italy
| | - Lorenza Putignani
- Unit of Parasitology and Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,AOU Policlinico Umberto I, Rome, Italy.,AOU Sant'Andrea Hospital, Rome, Italy
| |
Collapse
|
394
|
Yuan Z, Yan W, Wen C, Zheng J, Yang N, Sun C. Enterotype identification and its influence on regulating the duodenum metabolism in chickens. Poult Sci 2020; 99:1515-1527. [PMID: 32111319 PMCID: PMC7587748 DOI: 10.1016/j.psj.2019.10.078] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Abstract
Enterotypes are used to describe clusters of specific gut microbial community structures, but few reports exist on the identification of enterotypes in poultry. In addition, there is incomplete understanding on the role of the foregut microbiota in the digestion and absorption of nutrients in poultry. Thus, this study aimed to identify the duodenal enterotypes by examining microbial communities from 206 broilers using 16S rRNA high-throughput sequencing and explore the effects of enterotypes on phenotypic performance and nutrient metabolism with metabolomics. The duodenal microbial communities of the broiler population were partitioned into 3 enterotypes (ET1, ET2, and ET3), and significant differences were observed in α-diversity among the enterotypes (P < 0.01). At the genus level, the ET1 group was over-represented by Bacteroides (9.8%) and Escherichia–Shigella (8.9%), the ET2 group was over-represented by Ochrobactrum (19.4%) and Rhodococcus (14.7%), and the ET3 group was over-represented by Bacillus (23.4%) and Akkermansia (16.2%). The relative abundance of the dominant taxa of each enterotype was significantly higher than that in the other 2 enterotypes (P < 0.01). The results showed that Ochrobactrum and Rhodococcus were positively correlated with cellobiose, alpha-D-glucose, D-mannose, and D-allose (r = 0.429, 0.435, 0.482, and 0.562, respectively; all P < 0.05). Rhodococcus was also positively correlated with tridecanoic acid and glycerol 1-myristate (r = 0.655 and 0.489, respectively; all P < 0.01). In terms of phenotype, the triglyceride level in the ET2 group was significantly higher than that in the ET1 group (P < 0.05), and the subcutaneous fat thickness and abdominal fat weight in the ET2 group were the highest (P > 0.05). Taken together, these results confirmed the presence of enterotypes in broilers and found that the dominant microbes in broilers of the ET2 group might play a major role in the degradation and utilization of plant polysaccharides, which may have an impact on the serum triglyceride level and fat deposition in broilers. These findings lay a foundation for further studies on the gut microbial interactions with the metabolism in broilers and the regulation of the gut microbiota to promote growth and well-being in broilers.
Collapse
Affiliation(s)
- Zhongyang Yuan
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Wei Yan
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chaoliang Wen
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiangxia Zheng
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
395
|
Gardner A, Carpenter G, So PW. Salivary Metabolomics: From Diagnostic Biomarker Discovery to Investigating Biological Function. Metabolites 2020; 10:E47. [PMID: 31991929 PMCID: PMC7073850 DOI: 10.3390/metabo10020047] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/15/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolomic profiling of biofluids, e.g., urine, plasma, has generated vast and ever-increasing amounts of knowledge over the last few decades. Paradoxically, metabolomic analysis of saliva, the most readily-available human biofluid, has lagged. This review explores the history of saliva-based metabolomics and summarizes current knowledge of salivary metabolomics. Current applications of salivary metabolomics have largely focused on diagnostic biomarker discovery and the diagnostic value of the current literature base is explored. There is also a small, albeit promising, literature base concerning the use of salivary metabolomics in monitoring athletic performance. Functional roles of salivary metabolites remain largely unexplored. Areas of emerging knowledge include the role of oral host-microbiome interactions in shaping the salivary metabolite profile and the potential roles of salivary metabolites in oral physiology, e.g., in taste perception. Discussion of future research directions describes the need to begin acquiring a greater knowledge of the function of salivary metabolites, a current research direction in the field of the gut metabolome. The role of saliva as an easily obtainable, information-rich fluid that could complement other gastrointestinal fluids in the exploration of the gut metabolome is emphasized.
Collapse
Affiliation(s)
- Alexander Gardner
- Salivary Research, Centre for Host–Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.G.); (G.C.)
- Department of Restorative Dentistry, Dental Hospital and School, University of Dundee, Dundee DD1 4HR, UK
| | - Guy Carpenter
- Salivary Research, Centre for Host–Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.G.); (G.C.)
| | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| |
Collapse
|
396
|
Stacchiotti A, Favero G, Rodella LF. Impact of Melatonin on Skeletal Muscle and Exercise. Cells 2020; 9:cells9020288. [PMID: 31991655 PMCID: PMC7072499 DOI: 10.3390/cells9020288] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle disorders are dramatically increasing with human aging with enormous sanitary costs and impact on the quality of life. Preventive and therapeutic tools to limit onset and progression of muscle frailty include nutrition and physical training. Melatonin, the indole produced at nighttime in pineal and extra-pineal sites in mammalians, has recognized anti-aging, anti-inflammatory, and anti-oxidant properties. Mitochondria are the favorite target of melatonin, which maintains them efficiently, scavenging free radicals and reducing oxidative damage. Here, we discuss the most recent evidence of dietary melatonin efficacy in age-related skeletal muscle disorders in cellular, preclinical, and clinical studies. Furthermore, we analyze the emerging impact of melatonin on physical activity. Finally, we consider the newest evidence of the gut-muscle axis and the influence of exercise and probably melatonin on the microbiota. In our opinion, this review reinforces the relevance of melatonin as a safe nutraceutical that limits skeletal muscle frailty and prolongs physical performance.
Collapse
Affiliation(s)
- Alessandra Stacchiotti
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (L.F.R.)
- Interdepartmental University Center of Research “Adaptation and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Correspondence: ; Tel.: +39-030-3717478; Fax: +39-030-3717486
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (L.F.R.)
| | - Luigi Fabrizio Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (L.F.R.)
- Interdepartmental University Center of Research “Adaptation and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
397
|
Trošt K, Ahonen L, Suvitaival T, Christiansen N, Nielsen T, Thiele M, Jacobsen S, Krag A, Rossing P, Hansen T, Dragsted LO, Legido-Quigley C. Describing the fecal metabolome in cryogenically collected samples from healthy participants. Sci Rep 2020; 10:885. [PMID: 31965056 PMCID: PMC6972823 DOI: 10.1038/s41598-020-57888-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
The chemical composition of feces plays an important role in human metabolism. Metabolomics and lipidomics are valuable tools for screening the metabolite composition in feces. Here we set out to describe fecal metabolite composition in healthy participants in frozen stools. Frozen stool samples were collected from 10 healthy volunteers and cryogenically drilled in four areas along the specimen. Polar metabolites were analyzed using derivatization followed by two-dimensional gas chromatography and time of flight mass spectrometry. Lipids were detected using ultra high-performance liquid chromatography coupled with quadruple time-of-flight mass spectrometry. 2326 metabolic features were detected. Out of a total of 298 metabolites that were annotated we report here 185 that showed a technical variation of x < 30%. These metabolites included amino acids, fatty acid derivatives, carboxylic acids and phenolic compounds. Lipids predominantly belonged to the groups of diacylglycerols, triacylglycerols and ceramides. Metabolites varied between sampling areas, some were broadly homogeneous, others varied 80%. A LASSO-computed network using metabolites present in all areas showed two main clusters describing the system, DAG lipids and phenyllactic acid. In feces from healthy participants, the main groups detected were phenolic compounds, ceramides, diacylglycerols and triacylglycerols.
Collapse
Affiliation(s)
| | - Linda Ahonen
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Biosyntia ApS, Copenhagen, Denmark
| | | | | | - Trine Nielsen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maja Thiele
- Department of Gastroenterology and Hepatology and Odense Patient Data Exploratory Network (OPEN), Odense University Hospital, Odense, Denmark.,Institute for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Suganya Jacobsen
- Department of Gastroenterology and Hepatology and Odense Patient Data Exploratory Network (OPEN), Odense University Hospital, Odense, Denmark.,Institute for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Aleksander Krag
- Department of Gastroenterology and Hepatology and Odense Patient Data Exploratory Network (OPEN), Odense University Hospital, Odense, Denmark.,Institute for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Ove Dragsted
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Legido-Quigley
- Steno Diabetes Center Copenhagen, Gentofte, Denmark. .,Institute of Pharmaceutical Science, King's College London, London, UK.
| |
Collapse
|
398
|
Valerio A, Casadei L, Giuliani A, Valerio M. Fecal Metabolomics as a Novel Noninvasive Method for Short-Term Stress Monitoring in Beef Cattle. J Proteome Res 2020; 19:845-853. [PMID: 31873020 DOI: 10.1021/acs.jproteome.9b00655] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Traditional measures of short-term stress response such as fecal glucocorticoid metabolites (FGM) are widely used in controlled settings to quantify the intensity of stimulation to which cattle are exposed. However, FGMs present several methodological and interpretation pitfalls when applied on animals in free-ranging conditions. In this study, we proposed an NMR-based fecal metabolomics strategy for noninvasive stress detection in beef cattle. Using a longitudinal sample collection, we monitored the changes in the fecal metabolome and FGM concentrations before and after an acute stressful event. Our results showed that while the fecal metabolome changed as a function of stress (p < 0.001), the mean concentrations of FGM did not change (Levene's test: F-ratio: 0.87, p-value: 0.44). Furthermore, we showed that the interanimal variability observed in the stress response was correlated with the individual fecal microbiota. This result was in line with recent findings, indicating the gut microbiome as a crucial mediator of stress response. We conclude that NMR-based fecal metabolomics proved to be a reliable methodology to assess stress response and that its future applicability to studies for stress monitoring in range animals may be more appropriate than FGM analysis.
Collapse
Affiliation(s)
- Azzurra Valerio
- School of the Environment , Washington State University , Pullman , Washington 99164 , United States
| | - Luca Casadei
- School of the Environment , Washington State University , Pullman , Washington 99164 , United States
| | - Alessandro Giuliani
- Department of Environment and Health , National Institute of Health , Rome 00161 , Italy
| | - Mariacristina Valerio
- School of the Environment , Washington State University , Pullman , Washington 99164 , United States
| |
Collapse
|
399
|
Liu Y, Wang Y, Ni Y, Cheung CKY, Lam KSL, Wang Y, Xia Z, Ye D, Guo J, Tse MA, Panagiotou G, Xu A. Gut Microbiome Fermentation Determines the Efficacy of Exercise for Diabetes Prevention. Cell Metab 2020; 31:77-91.e5. [PMID: 31786155 DOI: 10.1016/j.cmet.2019.11.001] [Citation(s) in RCA: 233] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/24/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023]
Abstract
Exercise is an effective strategy for diabetes management but is limited by the phenomenon of exercise resistance (i.e., the lack of or the adverse response to exercise on metabolic health). Here, in 39 medication-naive men with prediabetes, we found that exercise-induced alterations in the gut microbiota correlated closely with improvements in glucose homeostasis and insulin sensitivity (clinicaltrials.gov entry NCT03240978). The microbiome of responders exhibited an enhanced capacity for biosynthesis of short-chain fatty acids and catabolism of branched-chain amino acids, whereas those of non-responders were characterized by increased production of metabolically detrimental compounds. Fecal microbial transplantation from responders, but not non-responders, mimicked the effects of exercise on alleviation of insulin resistance in obese mice. Furthermore, a machine-learning algorithm integrating baseline microbial signatures accurately predicted personalized glycemic response to exercise in an additional 30 subjects. These findings raise the possibility of maximizing the benefits of exercise by targeting the gut microbiota.
Collapse
Affiliation(s)
- Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China; Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Yao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, the University of Hong Kong, Hong Kong, China
| | - Yueqiong Ni
- Systems Biology & Bioinformatics Group, School of the Biological Sciences, Faculty of Sciences, the University of Hong Kong, Hong Kong, China; Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute, Jena, Germany
| | - Cynthia K Y Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China; Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China; Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, the University of Hong Kong, Hong Kong, China
| | - Zhengyuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Dewei Ye
- Joint Laboratory between Guangdong and Hong Kong on Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiao Guo
- Joint Laboratory between Guangdong and Hong Kong on Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Michael Andrew Tse
- Active Health Clinic, Centre for Sports and Exercise, the University of Hong Kong, Hong Kong, China.
| | - Gianni Panagiotou
- Systems Biology & Bioinformatics Group, School of the Biological Sciences, Faculty of Sciences, the University of Hong Kong, Hong Kong, China; Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute, Jena, Germany; Department of Microbiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China.
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China; Department of Medicine, the University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, the University of Hong Kong, Hong Kong, China.
| |
Collapse
|
400
|
Landberg R, Hanhineva K. Biomarkers of a Healthy Nordic Diet-From Dietary Exposure Biomarkers to Microbiota Signatures in the Metabolome. Nutrients 2019; 12:E27. [PMID: 31877633 PMCID: PMC7019922 DOI: 10.3390/nu12010027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/02/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Whole diets and dietary patterns are increasingly highlighted in modern nutrition and health research instead of single food items or nutrients alone. The Healthy Nordic Diet is a dietary pattern typically associated with beneficial health outcomes in observational studies, but results from randomized controlled trials are mixed. Dietary assessment is one of the greatest challenges in observational studies and compliance is a major challenge in dietary interventions. During the last decade, research has shown the great importance of the gut microbiota in health and disease. Studies have have both shown that the Nordic diet affects the gut microbiota and that the gut microbiota predicts the effects of such a diet. Rapid technique developments in the area of high-throughput mass spectrometry have enabled the large-scale use of metabolomics both as an objective measurement of dietary intake as well as in providing the final readout of the endogenous metabolic processes and the impact of the gut microbiota. In this review, we give an update on the current status on biomarkers that reflect a Healthy Nordic Diet or individual components thereof (food intake biomarkers), biomarkers that show the effects of a Healthy Nordic Diet and biomarkers reflecting the role of a Healthy Nordic Diet on the gut microbiota as well as how the gut microbiota or derived molecules may be used to predict the effects of a Healthy Nordic Diet on different outcomes.
Collapse
Affiliation(s)
- Rikard Landberg
- Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden;
| | - Kati Hanhineva
- Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden;
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, P.O. Box 1627, 70210 Kuopio, Finland
| |
Collapse
|