801
|
Koay YC, Wali JA, Luk AWS, Macia L, Cogger VC, Pulpitel TJ, Wahl D, Solon-Biet SM, Holmes A, Simpson SJ, O'Sullivan JF. Ingestion of resistant starch by mice markedly increases microbiome-derived metabolites. FASEB J 2019; 33:8033-8042. [PMID: 30925066 DOI: 10.1096/fj.201900177r] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent research has shown significant health benefits deriving from high-dietary fiber or microbiome-accessible carbohydrate consumption. Compared with native starch (NS), dietary resistant starch (RS) is a high microbiome-accessible carbohydrate that significantly alters the gut microbiome. The aim of this study was to determine the systemic metabolic effects of high microbiome-accessible carbohydrate. Male C57BL/6 mice were divided into 2 groups and fed either NS or RS for 18 wk (n = 20/group). Metabolomic analyses revealed that plasma levels of numerous metabolites were significantly different between the RS-fed and NS-fed mice, many of which are microbiome-derived. Most strikingly, we observed a 22-fold increase in gut microbiome-derived tryptophan metabolite indole-3-propionate (IPA), which was positively correlated with several gut microbiota, including Allobaculum, Bifidobacterium, and Lachnospiraceae, with Allobaculum having the most consistently increased abundance of all the IPA-associated taxa across all RS-fed mice. In addition, major changes were observed for metabolites solely or primarily metabolized in the gut (e.g., trimethylamine-N-oxide), metabolites that have a significant entero-hepatic circulation (i.e., bile acids), lipid metabolites (e.g., cholesterol sulfate), metabolites indicating increased energy turnover (e.g., tricarboxylic acid cycle intermediates and ketone bodies), and increased antioxidants such as reduced glutathione. Our findings reveal potentially novel mediators of high microbiome-accessible carbohydrate-derived health benefits.-Koay,Y. C., Wali. J. A., Luk, A. W. S., Macia, L., Cogger, V. C., Pulpitel, T. J., Wahl, D., Solon-Biet, S. M., Holmes, A., Simpson, S. J., O'Sullivan, J. F. Ingestion of resistant starch by mice markedly increases microbiome-derived metabolites.
Collapse
Affiliation(s)
- Yen Chin Koay
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Jibran A Wali
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Alison W S Luk
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Ageing and Alzheimer's Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord, New South Wales, Australia
| | - Tamara J Pulpitel
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Devin Wahl
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew Holmes
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - John F O'Sullivan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
802
|
Sun M, Ma N, He T, Johnston LJ, Ma X. Tryptophan (Trp) modulates gut homeostasis via aryl hydrocarbon receptor (AhR). Crit Rev Food Sci Nutr 2019; 60:1760-1768. [PMID: 30924357 DOI: 10.1080/10408398.2019.1598334] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The intestinal homeostasis is an orchestrated dynamic equilibrium state composed of the coexistence and interactions among the nutrients, microbial flora, and immune system. The intestinal balance disorder can trigger a series of diseases, such as inflammatory bowel disease (IBD). Many of tryptophan (Trp) metabolites, such as kynurenine and indole, generated under a series of endogenous enzymes or microbial metabolism, have been reported enable to bind and activate the aryl hydrocarbon receptor (AhR), this series of process is termed the Trp-AhR pathway. The activated Trp-AhR pathway can induce the expression of downstream cytokines such as interleukin-22 (IL-22) and interleukin-17 (IL-17), thereby regulating the intestinal homeostasis. This review highlights the advance of Trp-AhR pathway in the regulation of intestinal homeostasis and provides some insights for the clinical strategies that expect to effectively prevent and treat gut diseases via intervening the Trp-AhR pathway.
Collapse
Affiliation(s)
- Meige Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J Johnston
- Swine Nutrition and Production, West Central Research and Outreach Center, University of Minnesota, Morris, MN, USA
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Department of Internal Medicine Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
803
|
Gut Microbiota Metabolite Indole Propionic Acid Targets Tryptophan Biosynthesis in Mycobacterium tuberculosis. mBio 2019; 10:mBio.02781-18. [PMID: 30914514 PMCID: PMC6437058 DOI: 10.1128/mbio.02781-18] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
New drugs against tuberculosis are urgently needed. The tryptophan (Trp) analog indole propionic acid (IPA) is the first antitubercular metabolite produced by human gut bacteria. Here, we show that this antibiotic blocks Trp synthesis, an in vivo essential biosynthetic pathway in M. tuberculosis. Intriguingly, IPA acts by decoupling a bacterial feedback regulatory mechanism: it mimics Trp as allosteric inhibitor of anthranilate synthase, thereby switching off Trp synthesis regardless of intracellular Trp levels. The identification of IPA’s target paves the way for the discovery of more potent TrpE ligands employing rational, target-based lead optimization. Indole propionic acid (IPA), produced by the gut microbiota, is active against Mycobacterium tuberculosisin vitro and in vivo. However, its mechanism of action is unknown. IPA is the deamination product of tryptophan (Trp) and thus a close structural analog of this essential aromatic amino acid. De novo Trp biosynthesis in M. tuberculosis is regulated through feedback inhibition: Trp acts as an allosteric inhibitor of anthranilate synthase TrpE, which catalyzes the first committed step in the Trp biosynthesis pathway. Hence, we hypothesized that IPA may mimic Trp as an allosteric inhibitor of TrpE and exert its antimicrobial effect by blocking synthesis of Trp at the TrpE catalytic step. To test our hypothesis, we carried out metabolic, chemical rescue, genetic, and biochemical analyses. Treatment of mycobacteria with IPA inhibited growth and reduced the intracellular level of Trp, an effect abrogated upon supplementation of Trp in the medium. Missense mutations at the allosteric Trp binding site of TrpE eliminated Trp inhibition and caused IPA resistance. In conclusion, we have shown that IPA blocks Trp biosynthesis in M. tuberculosis via inhibition of TrpE by mimicking the physiological allosteric inhibitor of this enzyme.
Collapse
|
804
|
Yuan X, Kang Y, Zhuo C, Huang XF, Song X. The gut microbiota promotes the pathogenesis of schizophrenia via multiple pathways. Biochem Biophys Res Commun 2019; 512:373-380. [PMID: 30898321 DOI: 10.1016/j.bbrc.2019.02.152] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a severe mental disorder with unknown etiology. Many mechanisms, including dysregulation of neurotransmitters, immune disturbance, and abnormal neurodevelopment, are proposed for the pathogenesis of schizophrenia. The significance of communication between intestinal flora and the central nervous system through the gut-brain axis is increasingly being recognized. The intestinal microbiota plays an important role in regulating neurotransmission, immune homeostasis, and brain development. We hypothesize that an imbalance in intestinal flora causes immune activation and dysfunction in the gut-brain axis, contributing to schizophrenia. In this review, we examine recent studies that explore the intestinal flora and immune-mediated neurodevelopment of schizophrenia. We conclude that an imbalance in intestinal flora may reduce protectants and increase neurotoxin and inflammatory mediators, causing neuronal and synaptic damage, which induces schizophrenia.
Collapse
Affiliation(s)
- Xiuxia Yuan
- The First Affiliated Hospital/Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Yulin Kang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Chuanjun Zhuo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, NSW, 2522, Australia.
| | - Xueqin Song
- The First Affiliated Hospital/Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
805
|
|
806
|
Wang L, Jin L, Xue B, Wang Z, Peng Q. Characterizing the bacterial community across the gastrointestinal tract of goats: Composition and potential function. Microbiologyopen 2019; 8:e00820. [PMID: 30829000 PMCID: PMC6741129 DOI: 10.1002/mbo3.820] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 12/28/2022] Open
Abstract
The composition and function of the microbial community in the gastrointestinal tract (GIT) have increasingly captured the attention of nutritionists because these traits affect the nutrient utilization efficiency and health of host animals. Little information has been reported on these aspects of the goat GIT. This study used 12 female goats (weighing 20.70 ± 1.60 kg and 10 months of age) to examine the composition and function of the microbiota in the rumen, abomasum, jejunum, cecum, and colon. Total genomic DNA was extracted from chyme samples from different sections of the GIT, and the hypervariable region of the 16S rRNA gene was amplified by PCR using bacterial universal primers. The amplicons were sequenced on an Illumina MiSeq platform, and the biological information was analyzed using QIIME software. A total of 857 genera that belonged to 39 phyla were observed across the goat GIT, with Bacteroidetes and Firmicutes dominating. Our results revealed significant differences in the composition, diversity, and species abundance of the bacterial communities in the different sections of the GIT. However, the compositions of the bacterial communities in adjacent GIT segments showed similarities in addition to differences. The study indicated that there were significant differences in microbial function among the GIT regions. In particular, the relative abundances of genes involved in energy metabolism, amino acid metabolism, nucleotide metabolism, and glycan metabolism were overrepresented in samples from the forestomach, and genes related to energy metabolism, amino acid metabolism, and glycan metabolism were mainly enriched in samples from the small intestine. Additionally, the relative abundances of bacteria at the phylum and genus levels were significantly correlated with these metabolic functions. In general, there were significant differences in composition and potential function among the bacterial communities in the goat GIT.
Collapse
Affiliation(s)
- Lizhi Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, PR China
| | - Lei Jin
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, PR China
| | - Bai Xue
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, PR China
| | - Zhisheng Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, PR China
| | - Quanhui Peng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, PR China
| |
Collapse
|
807
|
Ruebel ML, Piccolo BD, Mercer KE, Pack L, Moutos D, Shankar K, Andres A. Obesity leads to distinct metabolomic signatures in follicular fluid of women undergoing in vitro fertilization. Am J Physiol Endocrinol Metab 2019; 316:E383-E396. [PMID: 30601701 PMCID: PMC6459300 DOI: 10.1152/ajpendo.00401.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/26/2018] [Accepted: 12/31/2018] [Indexed: 02/07/2023]
Abstract
Although obesity negatively influences the metabolic homeostasis of cells within a broad range of tissues, its impact on oocyte metabolism is not fully understood. Prior evidence suggests that obesity increases expression of oocyte genes associated with inflammation, oxidative stress, and lipid metabolism; however, the metabolic impact of these genetic differences is not known. To address this gap, we conducted an exploratory assessment of the follicular fluid (FF) metabolome in eight overweight/obese (OW) and nine normal-weight (NW) women undergoing in vitro fertilization. FF and serum were collected and analyzed by untargeted metabolomics using gas chromatography-quadrupole time-of-flight mass spectrometry and charged-surface hybrid column-electrospray ionization quadrupole time-of-flight tandem mass spectrometry. Untargeted metabolomics identified obesity-associated changes in FF metabolites related to oxidative stress/antioxidant capacity, xenometabolism/amino acid biosynthesis, and lipid metabolism. Discriminant FF metabolites included elevated uric acid, isothreonic acid, one unknown primary metabolite, and six unknown complex lipids in OW compared with NW women. Conversely, 2-ketoglucose dimethylacetal, aminomalonate, two unknown primary metabolites, and two unknown complex lipids were decreased in FF of OW relative to NW women. Indole-3-propionic acid (IPA), a bacteria-derived metabolite, was also decreased in both FF and serum of OW women ( P < 0.05). The significant correlation between antioxidant IPA in serum and FF ( R = 0.95, P < 0.0001) suggests a potential serum biomarker of FF antioxidant status or reflection of the gut metabolism interaction with the follicle. These results suggest that obesity has important consequences for the follicular environment during the preconception period, a window of time that may be important for lifestyle interventions to ameliorate obesity-associated risk factors.
Collapse
Affiliation(s)
- Meghan L Ruebel
- Department of Animal Science and Reproductive and Developmental Sciences Program, Michigan State University , East Lansing, Michigan
- Arkansas Children's Nutrition Center , Little Rock, Arkansas
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center , Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Kelly E Mercer
- Arkansas Children's Nutrition Center , Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Lindsay Pack
- Arkansas Children's Nutrition Center , Little Rock, Arkansas
| | - Dean Moutos
- Arkansas Fertility and Gynecology Associates , Little Rock, Arkansas
| | - Kartik Shankar
- Arkansas Children's Nutrition Center , Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Aline Andres
- Arkansas Children's Nutrition Center , Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| |
Collapse
|
808
|
Zhang X, Shao H, Zheng X. Amino acids at the intersection of nutrition and insulin sensitivity. Drug Discov Today 2019; 24:1038-1043. [PMID: 30818029 DOI: 10.1016/j.drudis.2019.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/06/2019] [Accepted: 02/19/2019] [Indexed: 01/02/2023]
Abstract
A systems network that is coordinated in the sensing and management of nutrient signals is paramount to energy homeostasis, and its dysfunction induces metabolic stress and insulin resistance. Amino acids have recently emerged as a collection of signaling metabolites that underlie the metabolic impacts of different dietary patterns and life styles. This relationship is beginning to be understood from the close coupling of immune and metabolic systems, and serves to enrich our understanding of metabolic diseases, such as type 2 diabetes mellitus. In this review, we provide an overview of several amino acids or their metabolites that link nutrients with insulin sensitivity and discuss how they integrate into organ crosstalk pathways to influence physiological or pathological metabolic states.
Collapse
Affiliation(s)
- Xueli Zhang
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Hua Shao
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiao Zheng
- School of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
809
|
Seth P, Hsieh PN, Jamal S, Wang L, Gygi SP, Jain MK, Coller J, Stamler JS. Regulation of MicroRNA Machinery and Development by Interspecies S-Nitrosylation. Cell 2019; 176:1014-1025.e12. [PMID: 30794773 PMCID: PMC6559381 DOI: 10.1016/j.cell.2019.01.037] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/20/2018] [Accepted: 01/23/2019] [Indexed: 02/04/2023]
Abstract
Bioactive molecules can pass between microbiota and host to influence host cellular functions. However, general principles of interspecies communication have not been discovered. We show here in C. elegans that nitric oxide derived from resident bacteria promotes widespread S-nitrosylation of the host proteome. We further show that microbiota-dependent S-nitrosylation of C. elegans Argonaute protein (ALG-1)-at a site conserved and S-nitrosylated in mammalian Argonaute 2 (AGO2)-alters its function in controlling gene expression via microRNAs. By selectively eliminating nitric oxide generation by the microbiota or S-nitrosylation in ALG-1, we reveal unforeseen effects on host development. Thus, the microbiota can shape the post-translational landscape of the host proteome to regulate microRNA activity, gene expression, and host development. Our findings suggest a general mechanism by which the microbiota may control host cellular functions, as well as a new role for gasotransmitters.
Collapse
Affiliation(s)
- Puneet Seth
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Paishiun N Hsieh
- Department of Medicine, Case Cardiovascular Research Institute, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, 2103 Cornell Road, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Suhib Jamal
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Liwen Wang
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Mukesh K Jain
- Department of Medicine, Case Cardiovascular Research Institute, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, 2103 Cornell Road, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Jeff Coller
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
810
|
Zeng Y, Lin Y, Li L, Li Y, Zhang X, Wang M, Chen Y, Luo L, Lu B, Xie Z, Liao Q. Targeted metabolomics for the quantitative measurement of 9 gut microbiota-host co-metabolites in rat serum, urine and feces by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1110-1111:133-143. [PMID: 30807966 DOI: 10.1016/j.jchromb.2019.02.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 12/21/2018] [Accepted: 02/18/2019] [Indexed: 12/16/2022]
Abstract
Gut microbiota-host co-metabolites play an essential role in maintaining homeostasis, and their concentration changes are closely related to a variety of diseases. Developing a targeted metabolomics analytical platform for these co-metabolites will help to elucidate the relationship between intestinal flora and host. Here we present a simple and sensitive liquid chromatography-tandem mass spectrometry method for the analysis of nine gut microbiota-host co-metabolites in rat serum, urine and feces. The compounds were separated on a reversed-phase C18 column using gradient elution with a solvent system consisting of methanol and water (containing 0.05% formic acid) and a 7-min run time. All of the calibration curves exhibited good linear relationships (R2 ≥ 0.9984, Percent Residual Accuracy ≥93.27%). The intra- and interday precision, expressed as relative standard deviation (RSD), was ≤ 14.84%. The accuracy was within 100 ± 13.16% for all analytes. The recovery of the nine compounds in biological samples was ≥ 85.80% with an appropriate RSD (≤12.04%). The validated method was successfully applied to monitor the global changes of these metabolites in obesity. Taken together, these results demonstrate that the method can simultaneously determine the nine co-metabolites in multiple biological matrices and is an essential part of the targeted metabolomics analytical platform, which may become an approach to evaluate the occurrence, development and therapeutic effects of metabolic diseases.
Collapse
Affiliation(s)
- Yifeng Zeng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacy, Zengcheng District People's Hospital of Guangzhou, Guangzhou, China
| | - Yixuan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojun Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengxia Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongxiong Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liang Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Biyu Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, China.
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
811
|
Schroeder BO. Fight them or feed them: how the intestinal mucus layer manages the gut microbiota. Gastroenterol Rep (Oxf) 2019; 7:3-12. [PMID: 30792861 PMCID: PMC6375348 DOI: 10.1093/gastro/goy052] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
The intestinal tract is inhabited by a tremendous number of microorganisms, termed the gut microbiota. These microorganisms live in a mutualistic relationship with their host and assist in the degradation of complex carbohydrates. Although the gut microbiota is generally considered beneficial, the vast number of microbial cells also form a permanent threat to the host. Thus, the intestinal epithelium is covered with a dense layer of mucus to prevent translocation of the gut microbiota into underlying tissues. Intestinal mucus is an organized glycoprotein network with a host-specific glycan structure. While the mucus layer has long been considered a passive, host-designed barrier, recent studies showed that maturation and function of the mucus layer are strongly influenced by the gut microbiota. In return, the glycan repertoire of mucins can select for distinct mucosa-associated bacteria that are able to bind or degrade specific mucin glycans as a nutrient source. Because the intestinal mucus layer is at the crucial interface between host and microbes, its breakdown leads to gut bacterial encroachment that can eventually cause inflammation and infection. Accordingly, a dysfunctional mucus layer has been observed in colitis in mice and humans. Moreover, the increased consumption of a low-fiber Western-style diet in our modern society has recently been demonstrated to cause bacteria-mediated defects of the intestinal mucus layer. Here, I will review current knowledge on the interaction between gut bacteria and the intestinal mucus layer in health and disease. Understanding the molecular details of this host–microbe interaction may contribute to the development of novel treatment options for diseases involving a dysfunctional mucus layer, such as ulcerative colitis.
Collapse
Affiliation(s)
- Bjoern O Schroeder
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Bruna Stråket 16, University of Gothenburg, SE 413 45 Gothenburg, Sweden
| |
Collapse
|
812
|
Yeruva T, Lee CH. Regulation of Vaginal Microbiome by Nitric Oxide. Curr Pharm Biotechnol 2019; 20:17-31. [PMID: 30727888 DOI: 10.2174/1389201020666190207092850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/18/2018] [Accepted: 01/30/2019] [Indexed: 12/15/2022]
Abstract
In this review, the composition and regulation of vaginal microbiome that displays an apparent microbial diversity and interacts with other microbiota in the body are presented. The role of nitric oxide (NO) in the regulation of vaginal microflora in which lactobacillus species typically dominate has been delineated from the perspective of maintaining gynecologic ecosystem and prevention of onset of bacteriostatic vaginosis (BV) and/or sexually transmitted diseases (STD) including HIV-1 transmission. The interactions between NO and vaginal microbiome and its influence on the levels of Lactobacillus, hormones and other components are described. The recent progress, such as NO drugs, probiotic Lactobacilli and Lactobacillus microbots, that can be explored to alleviate abnormality of vagina microbiome, is also discussed. An identification of Oral-GI-Vagina axis, as well as the relationship between NO and Lactobacillus regulation in the healthy or pathological status of vagina microbiome, surely offers the advanced drug delivery option against BV or STD including AIDS.
Collapse
Affiliation(s)
- Taj Yeruva
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri, Kansas City, MO, 64108, United States
| | - Chi H Lee
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri, Kansas City, MO, 64108, United States
| |
Collapse
|
813
|
Heianza Y, Sun D, Li X, DiDonato JA, Bray GA, Sacks FM, Qi L. Gut microbiota metabolites, amino acid metabolites and improvements in insulin sensitivity and glucose metabolism: the POUNDS Lost trial. Gut 2019; 68:263-270. [PMID: 29860242 PMCID: PMC6275143 DOI: 10.1136/gutjnl-2018-316155] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Alterations in gut microbiota have been linked to host insulin resistance, diabetes and impaired amino acid metabolism. We investigated whether changes in gut microbiota-dependent metabolite of trimethylamine N-oxide (TMAO) and its nutrient precursors (choline and L-carnitine) were associated with improvements in glucose metabolism and diabetes-related amino acids in a weight-loss diet intervention. DESIGN We included 504 overweight and obese adults who were randomly assigned to one of four energy-reduced diets varying in macronutrient intake. The 6-month changes (Δ) in TMAO, choline and L-carnitine levels after the intervention were calculated. RESULTS Greater decreases in choline and L-carnitine were significantly (p<0.05) associated with greater improvements in fasting insulin concentrations and homeostasis model assessment of insulin resistance (HOMA-IR) at 6 months. The reduction of choline was significantly related to 2-year improvements in glucose and insulin resistance. We found significant linkages between dietary fat intake and ΔTMAO for changes in fasting glucose, insulin and HOMA-IR (pinteraction <0.05); a greater increase in TMAO was related to lesser improvements in the outcomes among participants who consumed a high-fat diet. In addition, ΔL-carnitine and Δcholine were significantly related to changes in amino acids (including branched-chain and aromatic amino acids). Interestingly, the associations of ΔTMAO, Δcholine and ΔL-carnitine with diabetes-related traits were independent of the changes in amino acids. CONCLUSION Our findings underscore the importance of changes in TMAO, choline and L-carnitine in improving insulin sensitivity during a weight-loss intervention for obese patients. Dietary fat intake may modify the associations of TMAO with insulin sensitivity and glucose metabolism. TRIAL REGISTRATION NUMBER NCT00072995.
Collapse
Affiliation(s)
- Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Dianjianyi Sun
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Joseph A. DiDonato
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH
| | - George A Bray
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
814
|
Abstract
Our understanding of the human gut microbiome continues to evolve at a rapid pace, but practical application of thisknowledge is still in its infancy. This review discusses the type of studies that will be essential for translating microbiome research into targeted modulations with dedicated benefits for the human host.
Collapse
Affiliation(s)
- Thomas S B Schmidt
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
| | - Jeroen Raes
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Rega Institute, Herestraat 49, 3000 Leuven, Belgium; VIB, Center for Microbiology, Heerestraat 49, 3000 Leuven, Belgium.
| | - Peer Bork
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany; Molecular Medicine Partnership Unit, University of Heidelberg and European Molecular Biology Laboratory, 69120 Heidelberg, Germany; Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany.
| |
Collapse
|
815
|
Microbiome control of innate reactivity. Curr Opin Immunol 2019; 56:107-113. [PMID: 30677624 DOI: 10.1016/j.coi.2018.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/23/2018] [Accepted: 12/08/2018] [Indexed: 12/19/2022]
Abstract
Numerous scientific disciplines, including immunology, are now positioned to fully realize the potential of the intestinal microbiome to modulate a wide array of basic processes. Increasingly, microbiota-derived metabolites are being recognized for mediating these effects. Coupled with advances in large scale sequencing and mass spectrometry, research into the microbiota and their metabolites has entered into an era of rapid discovery. Here, we review recent studies that have shown how-specific metabolic products of the microbiome alter properties of the innate immune system that in turn modulate response to infection and immunity.
Collapse
|
816
|
Sprouse ML, Bates NA, Felix KM, Wu HJJ. Impact of gut microbiota on gut-distal autoimmunity: a focus on T cells. Immunology 2019; 156:305-318. [PMID: 30560993 DOI: 10.1111/imm.13037] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/07/2018] [Accepted: 11/25/2018] [Indexed: 12/14/2022] Open
Abstract
The immune system is essential for maintaining a delicate balance between eliminating pathogens and maintaining tolerance to self-tissues to avoid autoimmunity. An enormous and complex community of gut microbiota provides essential health benefits to the host, particularly by regulating immune homeostasis. Many of the metabolites derived from commensals can impact host health by directly regulating the immune system. Many autoimmune diseases arise from an imbalance between pathogenic effector T cells and regulatory T (Treg) cells. Recent interest has emerged in understanding how cross-talk between gut microbiota and the host immune system promotes autoimmune development by controlling the differentiation and plasticity of T helper and Treg cells. At the molecular level, our recent study, along with others, demonstrates that asymptomatic colonization by commensal bacteria in the gut is capable of triggering autoimmune disease by molecular mimicking self-antigen and skewing the expression of dual T-cell receptors on T cells. Dysbiosis, an imbalance of the gut microbiota, is involved in autoimmune development in both mice and humans. Although it is well known that dysbiosis can impact diseases occurring within the gut, growing literature suggests that dysbiosis also causes the development of gut-distal/non-gut autoimmunity. In this review, we discuss recent advances in understanding the potential molecular mechanisms whereby gut microbiota induces autoimmunity, and the evidence that the gut microbiota triggers gut-distal autoimmune diseases.
Collapse
Affiliation(s)
- Maran L Sprouse
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Nicholas A Bates
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Krysta M Felix
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Hsin-Jung Joyce Wu
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA.,Arizona Arthritis Center, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
817
|
Abstract
Natural products have long played a pivotal role in the development of therapeutics for a variety of diseases. Traditionally, soil and marine environments have provided a rich reservoir from which diverse chemical scaffolds could be discovered. Recently, the human microbiome has been recognized as a promising niche from which secondary metabolites with therapeutic potential have begun to be isolated. In this Review, we address how the expansive history of identifying bacterial natural products in other environments is informing the approaches being brought to bear on the study of the human microbiota. We also touch on how these tools can lead to insights about microbe-microbe and host-microbe interactions and help generate biological hypotheses that may lead to developments of new therapeutic modalities.
Collapse
|
818
|
Abstract
The gut microbiota is a crucial actor in human physiology. Many of these effects are mediated by metabolites that are either produced by the microbes or derived from the transformation of environmental or host molecules. Among the array of metabolites at the interface between these microorganisms and the host is the essential aromatic amino acid tryptophan (Trp). In the gut, the three major Trp metabolism pathways leading to serotonin (5-hydroxytryptamine), kynurenine (Kyn), and indole derivatives are under the direct or indirect control of the microbiota. In this review, we gather the most recent advances concerning the central role of Trp metabolism in microbiota-host crosstalk in health and disease. Deciphering the complex equilibrium between these pathways will facilitate a better understanding of the pathogenesis of human diseases and open therapeutic opportunities.
Collapse
|
819
|
Cao X, Hamilton JJ, Venturelli OS. Understanding and Engineering Distributed Biochemical Pathways in Microbial Communities. Biochemistry 2019; 58:94-107. [PMID: 30457843 PMCID: PMC6733022 DOI: 10.1021/acs.biochem.8b01006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microbiomes impact nearly every environment on Earth by modulating the molecular composition of the environment. Temporally changing environmental stimuli and spatial organization are major variables shaping the structure and function of microbiomes. The web of interactions among members of these communities and between the organisms and the environment dictates microbiome functions. Microbial interactions are major drivers of microbiomes and are modulated by spatiotemporal parameters. A mechanistic and quantitative understanding of ecological, molecular, and environmental forces shaping microbiomes could inform strategies to control microbiome dynamics and functions. Major challenges for harnessing the potential of microbiomes for diverse applications include the development of predictive modeling frameworks and tools for precise manipulation of microbiome behaviors.
Collapse
Affiliation(s)
| | | | - Ophelia S. Venturelli
- Department of Biochemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
820
|
Naskali E, Dettmer K, Oefner PJ, Pereira PAB, Krohn K, Auvinen P, Ranki A, Kluger N. Serotonin and tryptophan metabolites, autoantibodies and gut microbiome in APECED. Endocr Connect 2019; 8:69-77. [PMID: 30608907 PMCID: PMC6365670 DOI: 10.1530/ec-18-0513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 01/04/2019] [Indexed: 12/28/2022]
Abstract
Objective Intestinal autoimmunity with gastrointestinal (GI) dysfunction has been shown in patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). Patients lack entero-endocrine (EE) cells and have circulating autoantibodies (Aabs) against critical enzymes in serotonin (5-HT) biosynthesis. Design We sought to determine the serum levels of 5-HT, tryptophan (Trp) metabolites and L-DOPA in 37 Finnish APECED patients and to correlate their abundance with the presence of TPH and AADC Aabs, GI dysfunction and depressive symptoms. We also performed an exploratory analysis of the gut microbiome. Methods Serum 5-HT, L-DOPA and Trp metabolite levels were determined by liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS). TPH and AADC Aabs were measured by ELISA. Depression was assessed with a structured RBDI questionnaire. The V3-V4 regions of the bacterial 16S rRNA gene were sequenced for gut microbiome exploration. Results Serum 5-HT levels were significantly decreased (130 ± 131 nmol/L vs 686 ± 233 nmol/L, P < 0.0001) in APECED patients with TPH-1 (±AADC) Aabs compared to controls and patients with only AADC Aabs. Reduced 5-HT levels correlated with constipation. The genus Escherichia/Shigella was overrepresented in the intestinal microbiome. No correlation between serum Trp, 5-HT or l-DOPA levels and the RBDI total score, fatigue or sleep disorders was found. Conclusions This exploratory study found low serum levels of 5-HT to be associated with constipation and the presence of TPH-1 and AADC Aabs, but not with symptoms of depression. Hence, serum 5-HT, TPH1 and AADC Aabs should be determined in APECED patients presenting with GI symptoms.
Collapse
Affiliation(s)
- Emmi Naskali
- Department of Dermatology, Allergology and Venereology, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Pedro A B Pereira
- Institute of Biotechnology, DNA Sequencing and Genomics Laboratory, University of Helsinki, Helsinki, Finland
| | - Kai Krohn
- Clinical Research Institute HUCH Ltd, Biomedicum Helsinki 1, Helsinki, Finland
| | - Petri Auvinen
- Institute of Biotechnology, DNA Sequencing and Genomics Laboratory, University of Helsinki, Helsinki, Finland
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
| | - Nicolas Kluger
- Department of Dermatology, Allergology and Venereology, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
- Correspondence should be addressed to N Kluger:
| |
Collapse
|
821
|
Cani PD, Van Hul M, Lefort C, Depommier C, Rastelli M, Everard A. Microbial regulation of organismal energy homeostasis. Nat Metab 2019; 1:34-46. [PMID: 32694818 DOI: 10.1038/s42255-018-0017-4] [Citation(s) in RCA: 344] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/15/2018] [Indexed: 12/13/2022]
Abstract
The gut microbiome has emerged as a key regulator of host metabolism. Here we review the various mechanisms through which the gut microbiome influences the energy metabolism of its host, highlighting the complex interactions between gut microbes, their metabolites and host cells. Among the most important bacterial metabolites are short-chain fatty acids, which serve as a direct energy source for host cells, stimulate the production of gut hormones and act in the brain to regulate food intake. Other microbial metabolites affect systemic energy expenditure by influencing thermogenesis and adipose tissue browning. Both direct and indirect mechanisms of action are known for specific metabolites, such as bile acids, branched chain amino acids, indole propionic acid and endocannabinoids. We also discuss the roles of specific bacteria in the production of specific metabolites and explore how external factors, such as antibiotics and exercise, affect the microbiome and thereby energy homeostasis. Collectively, we present a large body of evidence supporting the concept that gut microbiota-based therapies can be used to modulate host metabolism, and we expect to see such approaches moving from bench to bedside in the near future.
Collapse
Affiliation(s)
- Patrice D Cani
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Charlotte Lefort
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Clara Depommier
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marialetizia Rastelli
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
822
|
Yang X, Xu M, Huang G, Zhang C, Pang Y, Cheng Y. Effect of dietary L-tryptophan on the survival, immune response and gut microbiota of the Chinese mitten crab, Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2019; 84:1007-1017. [PMID: 30381266 DOI: 10.1016/j.fsi.2018.10.076] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 10/17/2018] [Accepted: 10/26/2018] [Indexed: 06/08/2023]
Abstract
This study investigated the influence of L-tryptophan (L-trp) on the survival, immune response and gut microbiota of the Chinese mitten crab, Eriocheir sinensis (with an average weight of 16.58 ± 2.20 g). After 30 days of feeding with diets supplemented with L-trp at 0.36%, 0.47%, 0.73% and 1.05% (groups 1, 2, 3 and 4, respectively), the survival rate and bacterial challenge (Aeromonas hydrophila) were evaluated, the activities of antioxidant and phosphatase enzymes in the serum were assessed, and the gut microbiota were measured via high-throughput Illumina sequencing. The results showed that the supplementation of L-trp significantly improved the survival rate of crabs (P < 0.05). After feeding for 7 days, it was observed that a high L-trp diet significantly increase the survival rate relative to a basal diet after a 96-h post-challenge with A. hydrophila (P < 0.05). The activity of CAT and AKP in the serum were increased by the addition of L-trp. The activity of CAT and AKP in the serum in group 4 were higher than those in group 1 (P < 0.05). Furthermore, we observed that adjunction of the L-trp can significantly increase the richness and diversity of the gut microbiota. The dominant phylum in the intestine of the Chinese mitten crab were Tenericutes, Proteobacteria, Firmicutes, Chloroflexi and Actinobacteria. The L-trp in the diets increased the richness of Proteobacteria, Firmicutes and Actinobacteria in the intestine significantly. These bacteria were all dominant bacteria and had a specific role in promoting the immunity of E. sinensis. Therefore, it could be inferred that L-trp supplementation is beneficial in the diet of E. sinensis. Based in these results, the dietary 0.47% or 0.73%L-trp supplemented is found to be optimum to improve E. sinensis survival.
Collapse
Affiliation(s)
- Xiaozhen Yang
- Key Laboratory of Freshwater Aquatic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Minjie Xu
- Key Laboratory of Freshwater Aquatic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Genyong Huang
- Key Laboratory of Freshwater Aquatic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Cong Zhang
- Key Laboratory of Freshwater Aquatic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Yangyang Pang
- Key Laboratory of Freshwater Aquatic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Yongxu Cheng
- Key Laboratory of Freshwater Aquatic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
823
|
Wu XM, Tan RX. Interaction between gut microbiota and ethnomedicine constituents. Nat Prod Rep 2019; 36:788-809. [DOI: 10.1039/c8np00041g] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This highlight reviews the interaction processes between gut microbiota and ethnomedicine constituents, which may conceptualize future therapeutic strategies.
Collapse
Affiliation(s)
- Xue Ming Wu
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Ren Xiang Tan
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
- State Key Laboratory of Pharmaceutical Biotechnology
| |
Collapse
|
824
|
Sorbara MT, Pamer EG. Interbacterial mechanisms of colonization resistance and the strategies pathogens use to overcome them. Mucosal Immunol 2019; 12:1-9. [PMID: 29988120 PMCID: PMC6312114 DOI: 10.1038/s41385-018-0053-0] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/15/2018] [Accepted: 05/27/2018] [Indexed: 02/08/2023]
Abstract
The communities of bacteria that reside in the intestinal tract are in constant competition within this dynamic and densely colonized environment. At homeostasis, the equilibrium that exists between these species and strains is shaped by their metabolism and also by pathways of active antagonism, which drive competition with related and unrelated strains. Importantly, these normal activities contribute to colonization resistance by the healthy microbiota, which includes the ability to prevent the expansion of potential pathogens. Disruption of the microbiota, resulting from, for example, inflammation or antibiotic use, can reduce colonization resistance. Pathogens that engraft following disruption of the microbiota are often adapted to expand into newly created niches and compete in an altered gut environment. In this review, we examine both the interbacterial mechanisms of colonization resistance and the strategies of pathogenic strains to exploit gaps in colonization resistance.
Collapse
Affiliation(s)
- Matthew T. Sorbara
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Eric G. Pamer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
- Center for Microbes, Inflammation and Cancer, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
825
|
Wang Y, Liu J, Zhao X, Yang C, Ozaki Y, Xu Z, Zhao B, Yu Z. A chiral signal-amplified sensor for enantioselective discrimination of amino acids based on charge transfer-induced SERS. Chem Commun (Camb) 2019; 55:9697-9700. [DOI: 10.1039/c9cc04665h] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An ultra-high sensitivity enantioselective sensor with excellent discrimination performance for trace amino acids by using charge transfer-induced SERS.
Collapse
Affiliation(s)
- Yue Wang
- Department of Chemistry
- College of Sciences
- Northeastern University
- Shenyang 110819
- People's Republic of China
| | - Jing Liu
- Department of Chemistry
- College of Sciences
- Northeastern University
- Shenyang 110819
- People's Republic of China
| | - Xueqi Zhao
- Department of Chemistry
- College of Sciences
- Northeastern University
- Shenyang 110819
- People's Republic of China
| | - Chunguang Yang
- Department of Chemistry
- College of Sciences
- Northeastern University
- Shenyang 110819
- People's Republic of China
| | - Yukihiro Ozaki
- Department of Chemistry
- School of Science and Technology
- Kwansei Gakuin University
- Sanda
- Japan
| | - Zhangrun Xu
- Department of Chemistry
- College of Sciences
- Northeastern University
- Shenyang 110819
- People's Republic of China
| | - Bing Zhao
- State Key Laboratory of Supramolecular Structure and Materials
- Jilin University
- Changchun 130012
- People's Republic of China
| | - Zhi Yu
- The Guo China-US Photonics Laboratory
- Changchun Institute of Optics
- Fine Mechanics and Physics
- Changchun
- People's Republic of China
| |
Collapse
|
826
|
Bin P, Tang Z, Liu S, Chen S, Xia Y, Liu J, Wu H, Zhu G. Intestinal microbiota mediates Enterotoxigenic Escherichia coli-induced diarrhea in piglets. BMC Vet Res 2018; 14:385. [PMID: 30518356 PMCID: PMC6282381 DOI: 10.1186/s12917-018-1704-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/21/2018] [Indexed: 12/31/2022] Open
Abstract
Background Enterotoxigenic Escherichia coli (ETEC) causes diarrhea in humans, cows, and pigs. The gut microbiota underlies pathology of several infectious diseases yet the role of the gut microbiota in the pathogenesis of ETEC-induced diarrhea is unknown. Results By using an ETEC induced diarrheal model in piglet, we profiled the jejunal and fecal microbiota using metagenomics and 16S rRNA sequencing. A jejunal microbiota transplantation experiment was conducted to determine the role of the gut microbiota in ETEC-induced diarrhea. ETEC-induced diarrhea influenced the structure and function of gut microbiota. Diarrheal piglets had lower Bacteroidetes: Firmicutes ratio and microbiota diversity in the jejunum and feces, and lower percentage of Prevotella in the feces, but higher Lactococcus in the jejunum and higher Escherichia-Shigella in the feces. The transplantation of the jejunal microbiota from diarrheal piglets to uninfected piglets leaded to diarrhea after transplantation. Microbiota transplantation experiments also supported the notion that dysbiosis of gut microbiota is involved in the immune responses in ETEC-induced diarrhea. Conclusion We conclude that ETEC infection influences the gut microbiota and the dysbiosis of gut microbiota after ETEC infection mediates the immune responses in ETEC infection. Electronic supplementary material The online version of this article (10.1186/s12917-018-1704-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peng Bin
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiyi Tang
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shaojuan Liu
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shuai Chen
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, People's Republic of China
| | - Yaoyao Xia
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, People's Republic of China
| | - Jiaqi Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Hucong Wu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Guoqiang Zhu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.
| |
Collapse
|
827
|
Zeng H, Zheng Y, Lin Y, Huang C, Lin S, Zheng B, Zhang Y. Effect of fractionated lotus seed resistant starch on proliferation of Bifidobacterium longum and Lactobacillus delbrueckii subsp. bulgaricus and its structural changes following fermentation. Food Chem 2018; 268:134-142. [DOI: 10.1016/j.foodchem.2018.05.070] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/07/2018] [Accepted: 05/15/2018] [Indexed: 12/23/2022]
|
828
|
Vieira-Potter VJ, Cross TWL, Swanson KS, Sarma SJ, Lei Z, Sumner LW, Rosenfeld CS. Soy-Induced Fecal Metabolome Changes in Ovariectomized and Intact Female Rats: Relationship with Cardiometabolic Health. Sci Rep 2018; 8:16896. [PMID: 30442926 PMCID: PMC6237990 DOI: 10.1038/s41598-018-35171-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022] Open
Abstract
Phytoestrogens are plant-derived compounds found in a variety of foods, most notably, soy. These compounds have been shown to improve immuno-metabolic health, yet mechanisms remain uncertain. We demonstrated previously that dietary phytoestrogen-rich soy (SOY) rescued metabolic dysfunction/inflammation following ovariectomy (OVX) in female rats; we also noted remarkable shifts in gut microbiota in SOY vs control diet-fed rats. Importantly, specific bacteria that significantly increased in those fed the SOY correlated positively with several favorable host metabolic parameters. One mechanism by which gut microbes might lead to such host effects is through production of bacterial metabolites. To test this possibility, we utilized non-targeted gas chromatography-mass spectrometry (GCMS) to assess the fecal metabolome in those previously studied animals. Partial least square discriminant analysis (PLSDA) revealed clear separation of fecal metabolomes based on diet and ovarian state. In particular, SOY-fed animals had greater fecal concentrations of the beneficial bacterial metabolite, S-equol, which was positively associated with several of the bacteria upregulated in the SOY group. S-equol was inversely correlated with important indicators of metabolic dysfunction and inflammation, suggesting that this metabolite might be a key mediator between SOY and gut microbiome-positive host health outcomes.
Collapse
Affiliation(s)
- Victoria J Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, 65211, USA
| | - Tzu-Wen L Cross
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Kelly S Swanson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Saurav J Sarma
- MU Metabolomics Center, University of Missouri, Columbia, MO, 65211, USA
- Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Zhentian Lei
- MU Metabolomics Center, University of Missouri, Columbia, MO, 65211, USA
- Biochemistry, University of Missouri, Columbia, MO, 65211, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
| | - Lloyd W Sumner
- MU Metabolomics Center, University of Missouri, Columbia, MO, 65211, USA
- Biochemistry, University of Missouri, Columbia, MO, 65211, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
| | - Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA.
- Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA.
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, 65211, USA.
- Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
829
|
Xiao J, Peng Z, Liao Y, Sun H, Chen W, Chen X, Wei Z, Yang C, Nüssler AK, Liu J, Yang W. Organ transplantation and gut microbiota: current reviews and future challenges. Am J Transl Res 2018; 10:3330-3344. [PMID: 30662590 PMCID: PMC6291689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 10/21/2018] [Indexed: 06/09/2023]
Abstract
Organ transplantation is often the only effective treatment for patients with end-stage diseases, such as heart, liver, kidney and small bowel failure and is carried out frequently worldwide. Still the post-transplantation complications remain health- and life-threatening outcome that needed to be resolved. With the rapid development of molecular technologies in recent years, more and more researchers realize that the gut microbiota may play a critical role in human diseases. The intestinal microbiome has been proved to provide a lot of functions to the host, such as digesting food, modulating metabolism, promoting angiogenesis and regulating the immune system. Several studies have investigated the alteration of intestinal microbiota in post-transplantation patients and observed significant changes in the intestinal microbiome compared to the pre-transplant condition. Due to the abovementioned features that the gut microbiota may be used in the prognosis of clinical outcome of organ transplantation. In addition, the FMT (fecal microbiota transplantation), probiotics and prebiotics as the newest therapy methods, effectiveness of which has been verified in some diseases, such as Clostridium difficile infection, inflammatory bowel disease and other chronic disorders, might be used as the prognosis tool in organ transplantation as well. The purpose of this present review is to elucidate the relationship between gut microbiota and organ transplantation as well as the potential use of new therapies like fecal microbiota transplantation, probiotic and prebiotic administration after the transplantation, and provide some ideas for future researches in field of organ transplantation.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Department of Cardio-Thoracic Surgery, Ganzhou People’s HospitalGanzhou 341000, Jiangxi, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology13 Hangkong Road, Wuhan 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology13 Hangkong Road, Wuhan 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology13 Hangkong Road, Wuhan 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology13 Hangkong Road, Wuhan 430030, China
| | - Hui Sun
- Department of Cardio-Thoracic Surgery, Ganzhou People’s HospitalGanzhou 341000, Jiangxi, China
| | - Weiqiang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Zhanjie Wei
- Department of Thyroid Surgery, Central Hospital of WuhanWuhan 430022, Hubei, China
| | - Chuanlei Yang
- Department of Cardiovascular Surgery, Central Hospital of WuhanWuhan 430022, Hubei, China
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Center, University of TübingenSchnarrenbergstr. 95, Tübingen 72076, Germany
| | - Jinping Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology13 Hangkong Road, Wuhan 430030, China
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology13 Hangkong Road, Wuhan 430030, China
| |
Collapse
|
830
|
Koh A, Molinaro A, Ståhlman M, Khan MT, Schmidt C, Mannerås-Holm L, Wu H, Carreras A, Jeong H, Olofsson LE, Bergh PO, Gerdes V, Hartstra A, de Brauw M, Perkins R, Nieuwdorp M, Bergström G, Bäckhed F. Microbially Produced Imidazole Propionate Impairs Insulin Signaling through mTORC1. Cell 2018; 175:947-961.e17. [DOI: 10.1016/j.cell.2018.09.055] [Citation(s) in RCA: 344] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 06/30/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
|
831
|
Cheng D, Chang H, Ma S, Guo J, She G, Zhang F, Li L, Li X, Lu Y. Tiansi Liquid Modulates Gut Microbiota Composition and Tryptophan⁻Kynurenine Metabolism in Rats with Hydrocortisone-Induced Depression. Molecules 2018; 23:molecules23112832. [PMID: 30384480 PMCID: PMC6278342 DOI: 10.3390/molecules23112832] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/21/2018] [Accepted: 10/27/2018] [Indexed: 12/17/2022] Open
Abstract
Tiansi Liquid is a traditional Chinese herbal medicine used to treat depression; however, the underlying mechanisms remain unclear. Here, we examined the effect of Tiansi Liquid in a rat model of hydrocortisone-induced depression using behavioral testing, 16S rRNA high-throughput pyrosequencing and high-performance liquid chromatography-mass spectrometry-based metabolomics of the tryptophan (TRP)–kynurenine (KYN) pathway. Tiansi Liquid significantly improved the sucrose preference and exploratory behavior of the depressive rats. The richness of intestinal mucosa samples from the model (depressive) group tended to be higher than that from the control group, while the richness was higher in the Tiansi Liquid-treated group than in the model group. Tiansi Liquid increased the relative abundance of some microbiota (Ruminococcaceae, Lactococcus, Lactobacillus, Lachnospiraceae_NK4A136_group). Metabolomics showed that Tiansi Liquid reduced the levels of tryptophan 2,3 dioxygenase, indoleamine 2,3-dioxygenase, quinoline and the KYN/TRP ratio, while increasing kynurenic acid and 5-HT levels. Correlation analysis revealed a negative relationship between the relative abundance of the Lachnospiraceae_NK4A136_group and quinoline content. Collectively, these findings suggest that Tiansi Liquid ameliorates depressive symptoms in rats by modulating the gut microbiota composition and metabolites in the TRP–KYN pathway.
Collapse
Affiliation(s)
- Dan Cheng
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Hongsheng Chang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Suya Ma
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Jian Guo
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Gaimei She
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Feilong Zhang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Lingling Li
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Xinjie Li
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yi Lu
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
832
|
Indoleacetate decarboxylase is a glycyl radical enzyme catalysing the formation of malodorant skatole. Nat Commun 2018; 9:4224. [PMID: 30310076 PMCID: PMC6181972 DOI: 10.1038/s41467-018-06627-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/07/2018] [Indexed: 12/30/2022] Open
Abstract
Skatole is a malodorous compound that contributes to the characteristic smell of animal faeces. Although skatole has long been known to originate from bacterial tryptophan fermentation, the enzyme catalysing its formation has so far remained elusive. Here we report the use of comparative genomics for the discovery of indoleacetate decarboxylase, an O2-sensitive glycyl radical enzyme catalysing the decarboxylation of indoleacetate to form skatole as the terminal step of tryptophan fermentation in certain anaerobic bacteria. We describe its biochemical characterization and compare it to other glycyl radical decarboxylases. Indoleacetate decarboxylase may serve as a genetic marker for the identification of skatole-producing environmental and human-associated bacteria, with impacts on human health and the livestock industry. Skatole is a bacterial metabolite responsible for boar taint and the objectionable smell of manure. Here, the authors elucidate the final step of skatole biosynthesis, describing the discovery and biochemical characterization of the enzyme catalysing the conversion of indoleacetate into skatole.
Collapse
|
833
|
Chen MX, Wang SY, Kuo CH, Tsai IL. Metabolome analysis for investigating host-gut microbiota interactions. J Formos Med Assoc 2018; 118 Suppl 1:S10-S22. [PMID: 30269936 DOI: 10.1016/j.jfma.2018.09.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
Dysbiosis of the gut microbiome is associated with host health conditions. Many diseases have shown to have correlations with imbalanced microbiota, including obesity, inflammatory bowel disease, cancer, and even neurodegeneration disorders. Metabolomics studies targeting small molecule metabolites that impact the host metabolome and their biochemical functions have shown promise for studying host-gut microbiota interactions. Metabolome analysis determines the metabolites being discussed for their biological implications in host-gut microbiota interactions. To facilitate understanding the critical aspects of metabolome analysis, this article reviewed (1) the sample types used in host-gut microbiome studies; (2) mass spectrometry (MS)-based analytical methods and (3) useful tools for MS-based data processing/analysis. In addition to the most frequently used sample type, feces, we also discussed others biosamples, such as urine, plasma/serum, saliva, cerebrospinal fluid, exhaled breaths, and tissues, to better understand gut metabolite systemic effects on the whole organism. Gas chromatography-mass spectrometry (GC-MS), liquid chromatography-mass spectrometry (LC-MS), and capillary electrophoresis-mass spectrometry (CE-MS), three powerful tools that can be utilized to study host-gut microbiota interactions, are included with examples of their applications. After obtaining big data from MS-based instruments, noise removal, peak detection, missing value imputation, and data analysis are all important steps for acquiring valid results in host-gut microbiome research. The information provided in this review will help new researchers aiming to join this field by providing a global view of the analytical aspects involved in gut microbiota-related metabolomics studies.
Collapse
Affiliation(s)
- Michael X Chen
- Department of Laboratory Medicine and Pathology, The University of British Columbia, Canada; Island Medical Program, University of Victoria, Canada
| | - San-Yuan Wang
- Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan; The Metabolomics Core Laboratory, NTU Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan; Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| | - I-Lin Tsai
- Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
834
|
Kim S, Kerns SJ, Ziesack M, Bry L, Gerber GK, Way JC, Silver PA. Quorum Sensing Can Be Repurposed To Promote Information Transfer between Bacteria in the Mammalian Gut. ACS Synth Biol 2018; 7:2270-2281. [PMID: 30125499 DOI: 10.1021/acssynbio.8b00271] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The gut microbiome is intricately involved with establishing and maintaining the health of the host. Engineering of gut microbes aims to add new functions and expand the scope of control over the gut microbiome. To create systems that can perform increasingly complex tasks in the gut, it is necessary to harness the ability of the bacteria to communicate in the gut environment. Interestingly, acyl-homoserine lactone (acyl-HSL)-mediated Gram-negative bacterial quorum sensing, a widely used mode of intercellular signaling system in nature, has not been identified in normal healthy mammalian gut. It remains unknown whether the gut bacteria that do not natively use quorum sensing can be engineered to successfully signal to other bacteria using acyl-HSLs in the gut environment. Here, we repurposed quorum sensing to create an information transfer system between native gut Escherichia coli and attenuated Salmonella enterica serovar Typhimurium. Specifically, we functionalized one species with inducible signal production and the other with signal detection and recording using genomically integrated circuits. The information transfer system demonstrated successful intra- and interspecies signaling in the murine gut. This study provides a basis for further understanding of interbacterial interactions in an otherwise hard-to-study environment as well as a basis for further investigation of the potential of acyl-HSLs as intercellular signaling molecules of engineered gut consortia.
Collapse
Affiliation(s)
- Suhyun Kim
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - S. Jordan Kerns
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Marika Ziesack
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Lynn Bry
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts 02115, United States
| | - Georg K. Gerber
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts 02115, United States
| | - Jeffrey C. Way
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Pamela A. Silver
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| |
Collapse
|
835
|
Blachier F, Beaumont M, Portune KJ, Steuer N, Lan A, Audebert M, Khodorova N, Andriamihaja M, Airinei G, Benamouzig R, Davila AM, Armand L, Rampelli S, Brigidi P, Tomé D, Claus SP, Sanz Y. High-protein diets for weight management: Interactions with the intestinal microbiota and consequences for gut health. A position paper by the my new gut study group. Clin Nutr 2018; 38:1012-1022. [PMID: 30274898 DOI: 10.1016/j.clnu.2018.09.016] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS This review examines to what extent high-protein diets (HPD), which may favor body weight loss and improve metabolic outcomes in overweight and obese individuals, may also impact the gut environment, shaping the microbiota and the host-microbe (co)metabolic pathways and products, possibly affecting large intestine mucosa homeostasis. METHODS PubMed-referenced publications were analyzed with an emphasis on dietary intervention studies involving human volunteers in order to clarify the beneficial vs. deleterious effects of HPD in terms of both metabolic and gut-related health parameters; taking into account the interactions with the gut microbiota. RESULTS HPD generally decrease body weight and improve blood metabolic parameters, but also modify the fecal and urinary contents in various bacterial metabolites and co-metabolites. The effects of HPD on the intestinal microbiota composition appear rather heterogeneous depending on the type of dietary intervention. Recently, HPD consumption was shown to modify the expression of genes playing key roles in homeostatic processes in the rectal mucosa, without evidence of intestinal inflammation. Importantly, the effects of HPD on the gut were dependent on the protein source (i.e. from plant or animal sources), a result which should be considered for further investigations. CONCLUSION Although HPD appear to be efficient for weight loss, the effects of HPD on microbiota-derived metabolites and gene expression in the gut raise new questions on the impact of HPD on the large intestine mucosa homeostasis leading the authors to recommend some caution regarding the utilization of HPD, notably in a recurrent and/or long-term ways.
Collapse
Affiliation(s)
- François Blachier
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France.
| | - Martin Beaumont
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Kevin Joseph Portune
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agronomy and Food Technology, Spanish National Research Council, Valencia, Spain
| | - Nils Steuer
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Annaïg Lan
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Marc Audebert
- Toxalim, Research Centre in Food Toxicology, INRA, Toulouse, France
| | - Nadezda Khodorova
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | | | - Gheorghe Airinei
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Robert Benamouzig
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Anne-Marie Davila
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Lucie Armand
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Daniel Tomé
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris, France
| | - Sandrine Paule Claus
- Department of Food Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agronomy and Food Technology, Spanish National Research Council, Valencia, Spain
| |
Collapse
|
836
|
Abstract
Tuberculosis (TB) is an ancient infectious disease of humans that has been extensively studied both clinically and experimentally. Although susceptibility to Mycobacterium tuberculosis infection is clearly influenced by factors such as nutrition, immune status, and both mycobacterial and host genetics, the variable pathogenesis of TB in infected individuals remains poorly understood. Tuberculosis (TB) is an ancient infectious disease of humans that has been extensively studied both clinically and experimentally. Although susceptibility to Mycobacterium tuberculosis infection is clearly influenced by factors such as nutrition, immune status, and both mycobacterial and host genetics, the variable pathogenesis of TB in infected individuals remains poorly understood. During the past two decades, it has become clear that the microbiota—the trillion organisms that reside at mucosal surfaces within and on the body—can exert a major influence on disease outcome through its effects on host innate and adaptive immune function and metabolism. This new recognition of the potentially pleiotropic participation of the microbiome in immune responses has raised the possibility that the microbiota may influence M. tuberculosis infection and/or disease. Similarly, treatment of TB may alter the healthy steady-state composition and function of the microbiome, possibly affecting treatment outcome in addition to other host physiological parameters. Herein, we review emerging evidence for how the microbiota may influence the transition points in the life cycle of TB infection, including (i) resistance to initial infection, (ii) initial infection to latent tuberculosis (LTBI), (iii) LTBI to reactivated disease, and (iv) treatment to cure. A major goal of this review is to frame questions to guide future scientific and clinical studies in this largely unexplored but increasingly important area of TB research.
Collapse
|
837
|
Vijayakumar V, Vanhove AS, Pickering BS, Liao J, Tierney BT, Asara JM, Bronson R, Watnick PI. Removal of a Membrane Anchor Reveals the Opposing Regulatory Functions of Vibrio cholerae Glucose-Specific Enzyme IIA in Biofilms and the Mammalian Intestine. mBio 2018; 9:e00858-18. [PMID: 30181246 PMCID: PMC6123446 DOI: 10.1128/mbio.00858-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022] Open
Abstract
The Vibrio cholerae phosphoenolpyruvate phosphotransferase system (PTS) is a well-conserved, multicomponent phosphotransfer cascade that coordinates the bacterial response to carbohydrate availability through direct interactions of its components with protein targets. One such component, glucose-specific enzyme IIA (EIIAGlc), is a master regulator that coordinates bacterial metabolism, nutrient uptake, and behavior by direct interactions with cytoplasmic and membrane-associated protein partners. Here, we show that an amphipathic helix (AH) at the N terminus of V. cholerae EIIAGlc serves as a membrane association domain that is dispensable for interactions with cytoplasmic partners but essential for regulation of integral membrane protein partners. By deleting this AH, we reveal previously unappreciated opposing regulatory functions for EIIAGlc at the membrane and in the cytoplasm and show that these opposing functions are active in the laboratory biofilm and the mammalian intestine. Phosphotransfer through the PTS proceeds in the absence of the EIIAGlc AH, while PTS-dependent sugar transport is blocked. This demonstrates that the AH couples phosphotransfer to sugar transport and refutes the paradigm of EIIAGlc as a simple phosphotransfer component in PTS-dependent transport. Our findings show that Vibrio cholerae EIIAGlc, a central regulator of pathogen metabolism, contributes to optimization of bacterial physiology by integrating metabolic cues arising from the cytoplasm with nutritional cues arising from the environment. Because pathogen carbon metabolism alters the intestinal environment, we propose that it may be manipulated to minimize the metabolic cost of intestinal infection.IMPORTANCE The V. cholerae phosphoenolpyruvate phosphotransferase system (PTS) is a well-conserved, multicomponent phosphotransfer cascade that regulates cellular physiology and virulence in response to nutritional signals. Glucose-specific enzyme IIA (EIIAGlc), a component of the PTS, is a master regulator that coordinates bacterial metabolism, nutrient uptake, and behavior by direct interactions with protein partners. We show that an amphipathic helix (AH) at the N terminus of V. cholerae EIIAGlc serves as a membrane association domain that is dispensable for interactions with cytoplasmic partners but essential for regulation of integral membrane protein partners. By removing this amphipathic helix, hidden, opposing roles for cytoplasmic partners of EIIAGlc in both biofilm formation and metabolism within the mammalian intestine are revealed. This study defines a novel paradigm for AH function in integrating opposing regulatory functions in the cytoplasm and at the bacterial cell membrane and highlights the PTS as a target for metabolic modulation of the intestinal environment.
Collapse
Affiliation(s)
- Vidhya Vijayakumar
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Audrey S Vanhove
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bradley S Pickering
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Liao
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Braden T Tierney
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Roderick Bronson
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Paula I Watnick
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
838
|
Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun 2018; 9:3294. [PMID: 30120222 PMCID: PMC6098093 DOI: 10.1038/s41467-018-05470-4] [Citation(s) in RCA: 1229] [Impact Index Per Article: 175.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence implicates metabolites produced by gut microbes as crucial mediators of diet-induced host-microbial cross-talk. Here, we review emerging data suggesting that microbial tryptophan catabolites resulting from proteolysis are influencing host health. These metabolites are suggested to activate the immune system through binding to the aryl hydrocarbon receptor (AHR), enhance the intestinal epithelial barrier, stimulate gastrointestinal motility, as well as secretion of gut hormones, exert anti-inflammatory, anti-oxidative or toxic effects in systemic circulation, and putatively modulate gut microbial composition. Tryptophan catabolites thus affect various physiological processes and may contribute to intestinal and systemic homeostasis in health and disease.
Collapse
Affiliation(s)
- Henrik M Roager
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, DK-1958, Frederiksberg, Denmark.
- National Food Institute, Technical University of Denmark, DK-2800 Kgs, Lyngby, Denmark.
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, DK-2800 Kgs, Lyngby, Denmark.
| |
Collapse
|
839
|
Kim DJ, Yoon S, Ji SC, Yang J, Kim YK, Lee S, Yu KS, Jang IJ, Chung JY, Cho JY. Ursodeoxycholic acid improves liver function via phenylalanine/tyrosine pathway and microbiome remodelling in patients with liver dysfunction. Sci Rep 2018; 8:11874. [PMID: 30089798 PMCID: PMC6082879 DOI: 10.1038/s41598-018-30349-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023] Open
Abstract
Ursodeoxycholic acid (UDCA) is a metabolic by-product of intestinal bacteria, showing hepatoprotective effects. However, its underlying molecular mechanisms remain unclear. The purpose of this study was to elucidate the action mechanisms underlying the protective effects of UDCA and vitamin E against liver dysfunction using metabolomics and metagenomic analysis. In this study, we analysed blood and urine samples from patients with obesity and liver dysfunction. Nine patients were randomly assigned to receive UDCA (300 mg twice daily), and 10 subjects received vitamin E (400 IU twice daily) for 8 weeks. UDCA significantly improved the liver function scores after 4 weeks of treatment and effectively reduced hepatic deoxycholic acid and serum microRNA-122 levels. To better understand its protective mechanism, a global metabolomics study was conducted, and we found that UDCA regulated uremic toxins (hippuric acid, p-cresol sulphate, and indole-derived metabolites), antioxidants (ascorbate sulphate and N-acetyl-L-cysteine), and the phenylalanine/tyrosine pathway. Furthermore, microbiome involvement, particularly of Lactobacillus and Bifidobacterium, was demonstrated through metagenomic analysis of bacteria-derived extracellular vesicles. Meanwhile, vitamin E treatment did not result in such alterations, except that it reduced uremic toxins and liver dysfunction. Our findings suggested that both treatments were effective in improving liver function, albeit via different mechanisms.
Collapse
Affiliation(s)
- Da Jung Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Seonghae Yoon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Bundang Hospital, Seongnam, Korea
| | - Sang Chun Ji
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | | | | | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Bundang Hospital, Seongnam, Korea.
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea. .,Metabolomics Medical Research Center, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
840
|
Kell DB, Pretorius E. No effects without causes: the Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93:1518-1557. [PMID: 29575574 PMCID: PMC6055827 DOI: 10.1111/brv.12407] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022]
Abstract
Since the successful conquest of many acute, communicable (infectious) diseases through the use of vaccines and antibiotics, the currently most prevalent diseases are chronic and progressive in nature, and are all accompanied by inflammation. These diseases include neurodegenerative (e.g. Alzheimer's, Parkinson's), vascular (e.g. atherosclerosis, pre-eclampsia, type 2 diabetes) and autoimmune (e.g. rheumatoid arthritis and multiple sclerosis) diseases that may appear to have little in common. In fact they all share significant features, in particular chronic inflammation and its attendant inflammatory cytokines. Such effects do not happen without underlying and initially 'external' causes, and it is of interest to seek these causes. Taking a systems approach, we argue that these causes include (i) stress-induced iron dysregulation, and (ii) its ability to awaken dormant, non-replicating microbes with which the host has become infected. Other external causes may be dietary. Such microbes are capable of shedding small, but functionally significant amounts of highly inflammagenic molecules such as lipopolysaccharide and lipoteichoic acid. Sequelae include significant coagulopathies, not least the recently discovered amyloidogenic clotting of blood, leading to cell death and the release of further inflammagens. The extensive evidence discussed here implies, as was found with ulcers, that almost all chronic, infectious diseases do in fact harbour a microbial component. What differs is simply the microbes and the anatomical location from and at which they exert damage. This analysis offers novel avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Douglas B. Kell
- School of ChemistryThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- The Manchester Institute of BiotechnologyThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| | - Etheresia Pretorius
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| |
Collapse
|
841
|
|
842
|
Zhou P, Zhou N, Shao L, Li J, Liu S, Meng X, Duan J, Xiong X, Huang X, Chen Y, Fan X, Zheng Y, Ma S, Li C, Wu A. Diagnosis of Clostridium difficile infection using an UPLC-MS based metabolomics method. Metabolomics 2018; 14:102. [PMID: 30830376 DOI: 10.1007/s11306-018-1397-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 07/10/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The fecal metabolome of Clostridium difficile (CD) infection is far from being understood, particularly its non-volatile organic compounds. The drawbacks of current tests used to diagnose CD infection hinder their application. OBJECTIVE The aims of this study were to find new characteristic fecal metabolites of CD infection and develop a metabolomics model for the diagnosis of CD infection. METHODS Ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) was used to characterize the fecal metabolome of CD positive and negative diarrhea and healthy control stool samples. RESULTS Diarrhea and healthy control samples showed distinct clusters in the principal components analysis score plot, and CD positive group and CD negative group demonstrated clearer separation in a partial least squares discriminate analysis model. The relative abundance of sphingosine, chenodeoxycholic acid, phenylalanine, lysophosphatidylcholine (C16:0), and propylene glycol stearate was higher, and the relative abundance of fatty amide, glycochenodeoxycholic acid, tyrosine, linoleyl carnitine, and sphingomyelin was lower in CD positive diarrhea groups, than in the CD negative group. A linear discriminant analysis model based on capsiamide, dihydrosphingosine, and glycochenodeoxycholic acid was further constructed to identify CD infection in diarrhea. The leave-one-out cross-validation accuracy and area under receiver operating characteristic curve for the training set/external validation set were 90.00/78.57%, and 0.900/0.7917 respectively. CONCLUSIONS Compared with other hospital-onset diarrhea, CD diarrhea has distinct fecal metabolome characteristics. Our UPLC-MS metabolomics model might be useful tool for diagnosing CD diarrhea.
Collapse
Affiliation(s)
- Pengcheng Zhou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Ning Zhou
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Li Shao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Jianzhou Li
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, People's Republic of China
| | - Sidi Liu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xiujuan Meng
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Juping Duan
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xinrui Xiong
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xun Huang
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Yuhua Chen
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xuegong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yixiang Zheng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Shujuan Ma
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Chunhui Li
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| | - Anhua Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| |
Collapse
|
843
|
Zhou X, Du L, Shi R, Chen Z, Zhou Y, Li Z. Early-life food nutrition, microbiota maturation and immune development shape life-long health. Crit Rev Food Sci Nutr 2018; 59:S30-S38. [PMID: 29874476 DOI: 10.1080/10408398.2018.1485628] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The current knowledge about early-life nutrition and environmental factors that affect the interaction between the symbiotic microbiota and the host immune system has demonstrated novel regulatory target for treating allergic diseases, autoimmune disorders and metabolic syndrome. Various kinds of food nutrients (such as dietary fiber, starch, polyphenols and proteins) can provide energy resources for both intestinal microbiota and the host. The indigestible food components are fermented by the indigenous gut microbiota to produce diverse metabolites, including short-chain fatty acids, bile acids and trimethylamine-N-oxide, which can regulate the host metabolized physiology, immunity homeostasis and health state. Therefore it is commonly believed early-life perturbation of the microbial community structure and the dietary nutrition interference on the child mucosal immunity contribute to the whole life susceptibility to chronic diseases. In all, the combined interrelationship between food ingredients nutrition, intestinal microbiota configurations and host system immunity provides new therapeutic targets to treat various kinds of pathogenic inflammations and chronic diseases.
Collapse
Affiliation(s)
- Xiaoli Zhou
- a Shanghai Institute of Technology , Shanghai , China
| | - Lina Du
- a Shanghai Institute of Technology , Shanghai , China
| | - Ronghua Shi
- a Shanghai Institute of Technology , Shanghai , China
| | - Zhidong Chen
- a Shanghai Institute of Technology , Shanghai , China
| | - Yiming Zhou
- a Shanghai Institute of Technology , Shanghai , China
| | - Zongjie Li
- a Shanghai Institute of Technology , Shanghai , China
| |
Collapse
|
844
|
Silva JCP, Mota M, Martins FO, Nogueira C, Gonçalves T, Carneiro T, Pinto J, Duarte D, Barros AS, Jones JG, Gil AM. Intestinal Microbial and Metabolic Profiling of Mice Fed with High-Glucose and High-Fructose Diets. J Proteome Res 2018; 17:2880-2891. [PMID: 29923728 DOI: 10.1021/acs.jproteome.8b00354] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Increased sugar intake is implicated in Type-2 diabetes and fatty liver disease; however, the mechanisms through which glucose and fructose promote these conditions are unclear. We hypothesize that alterations in intestinal metabolite and microbiota profiles specific to each monosaccharide are involved. Two groups of six adult C57BL/6 mice were fed for 10-weeks with diets with glucose (G) or fructose (F) as sole carbohydrates, and a third group was fed with a normal chow carbohydrate mixture (N). Fecal metabolites were profiled by nuclear magnetic resonance (NMR) and microbial composition by real-time polymerase chain reaction (qPCR). Although N, G and F mice exhibited similar weight gains (with slight slower gains for F) and glucose tolerance, multivariate analysis of NMR data indicated that F mice were separated from N and G, with decreased butyrate and glutamate and increased fructose, succinate, taurine, tyrosine, and xylose. The different sugar diets also resulted in distinct intestinal microbiota profiles. That associated with fructose seemed to hold more potential to induce host metabolic disturbances compared to glucose, mainly by promoting bile acid deconjugation and taurine release and compromising intestinal barrier integrity. This may reflect the noted nonquantitative intestinal fructose absorption hence increasing its availability for microbial metabolism, a subject for further investigation.
Collapse
Affiliation(s)
- João C P Silva
- CNC - Centre for Neuroscience and Cell Biology , University of Coimbra , Coimbra , Portugal
| | - Marta Mota
- CNC - Centre for Neuroscience and Cell Biology , University of Coimbra , Coimbra , Portugal.,Institute of Microbiology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Fátima O Martins
- CNC - Centre for Neuroscience and Cell Biology , University of Coimbra , Coimbra , Portugal.,CEDOC, NOVA Medical School , Universidade NOVA de Lisboa , Rua Câmara Pestana, n°6, 6A, edifício II, piso 3 , 1150-082 Lisbon , Portugal
| | - Célia Nogueira
- CNC - Centre for Neuroscience and Cell Biology , University of Coimbra , Coimbra , Portugal.,Institute of Microbiology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Teresa Gonçalves
- CNC - Centre for Neuroscience and Cell Biology , University of Coimbra , Coimbra , Portugal.,Institute of Microbiology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Tatiana Carneiro
- CICECO-Aveiro Institute of Materials and Department of Chemistry , University of Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal
| | - Joana Pinto
- CICECO-Aveiro Institute of Materials and Department of Chemistry , University of Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal.,UCIBIO@REQUIMTE/Toxicological Laboratory, Biological Science Department, Faculty of Pharmacy , University of Porto , 4050-313 Porto , Portugal
| | - Daniela Duarte
- CICECO-Aveiro Institute of Materials and Department of Chemistry , University of Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal
| | - António S Barros
- CICECO-Aveiro Institute of Materials and Department of Chemistry , University of Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal.,Department of Cardiothoracic Surgery and Physiology, Faculty of Medicine , University of Porto , 4200-319 , Porto , Portugal
| | - John G Jones
- CNC - Centre for Neuroscience and Cell Biology , University of Coimbra , Coimbra , Portugal.,CEDOC, NOVA Medical School , Universidade NOVA de Lisboa , Rua Câmara Pestana, n°6, 6A, edifício II, piso 3 , 1150-082 Lisbon , Portugal.,APDP - Portuguese Diabetes Association , Lisbon , Portugal
| | - Ana M Gil
- CICECO-Aveiro Institute of Materials and Department of Chemistry , University of Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal
| |
Collapse
|
845
|
Waclawiková B, El Aidy S. Role of Microbiota and Tryptophan Metabolites in the Remote Effect of Intestinal Inflammation on Brain and Depression. Pharmaceuticals (Basel) 2018; 11:ph11030063. [PMID: 29941795 PMCID: PMC6160932 DOI: 10.3390/ph11030063] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022] Open
Abstract
The human gastrointestinal tract is inhabited by trillions of commensal bacteria collectively known as the gut microbiota. Our recognition of the significance of the complex interaction between the microbiota, and its host has grown dramatically over the past years. A balanced microbial community is a key regulator of the immune response, and metabolism of dietary components, which in turn, modulates several brain processes impacting mood and behavior. Consequently, it is likely that disruptions within the composition of the microbiota would remotely affect the mental state of the host. Here, we discuss how intestinal bacteria and their metabolites can orchestrate gut-associated neuroimmune mechanisms that influence mood and behavior leading to depression. In particular, we focus on microbiota-triggered gut inflammation and its implications in shifting the tryptophan metabolism towards kynurenine biosynthesis while disrupting the serotonergic signaling. We further investigate the gaps to be bridged in this exciting field of research in order to clarify our understanding of the multifaceted crosstalk in the microbiota–gut–brain interphase, bringing about novel, microbiota-targeted therapeutics for mental illnesses.
Collapse
Affiliation(s)
- Barbora Waclawiková
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| | - Sahar El Aidy
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| |
Collapse
|
846
|
Abstract
Mammalian immune systems evolved within a diverse world dominated by microbes, making interactions between these two life-forms inevitable. Adaptive immunity protects against microbes through antigen-specific responses. In classical studies, these responses were investigated in the context of pathogenicity; however, we now know that they have significant effects on our resident microbes. In turn, microbes employ an arsenal of mechanisms to influence development and specificity of host immunity. Understanding these complex reactions will be necessary to develop microbiota-based strategies to prevent or treat disease. Here we review the literature detailing the cross talk between resident microbes with a focus on the specificity of host responses and the microbial molecules that influence them.
Collapse
Affiliation(s)
- Kyla S Ost
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Utah 84211, USA;
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Utah 84211, USA;
| |
Collapse
|
847
|
Inshaw JRJ, Cutler AJ, Burren OS, Stefana MI, Todd JA. Approaches and advances in the genetic causes of autoimmune disease and their implications. Nat Immunol 2018; 19:674-684. [PMID: 29925982 DOI: 10.1038/s41590-018-0129-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/04/2018] [Indexed: 12/18/2022]
Abstract
Genome-wide association studies are transformative in revealing the polygenetic basis of common diseases, with autoimmune diseases leading the charge. Although the field is just over 10 years old, advances in understanding the underlying mechanistic pathways of these conditions, which result from a dense multifactorial blend of genetic, developmental and environmental factors, have already been informative, including insights into therapeutic possibilities. Nevertheless, the challenge of identifying the actual causal genes and pathways and their biological effects on altering disease risk remains for many identified susceptibility regions. It is this fundamental knowledge that will underpin the revolution in patient stratification, the discovery of therapeutic targets and clinical trial design in the next 20 years. Here we outline recent advances in analytical and phenotyping approaches and the emergence of large cohorts with standardized gene-expression data and other phenotypic data that are fueling a bounty of discovery and improved understanding of human physiology.
Collapse
Affiliation(s)
- Jamie R J Inshaw
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Antony J Cutler
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Oliver S Burren
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - M Irina Stefana
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
848
|
Palau-Rodriguez M, Tulipani S, Marco-Ramell A, Miñarro A, Jauregui O, Gonzalez-Dominguez R, Sanchez-Pla A, Ramos-Molina B, Tinahones FJ, Andres-Lacueva C. Characterization of Metabolomic Profile Associated with Metabolic Improvement after Bariatric Surgery in Subjects with Morbid Obesity. J Proteome Res 2018; 17:2704-2714. [DOI: 10.1021/acs.jproteome.8b00144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Magali Palau-Rodriguez
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| | - Sara Tulipani
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- Biomedical Research Institute [IBIMA], Service of Endocrinology and Nutrition, Malaga Hospital Complex [Virgen de la Victoria], Campus de Teatinos s/n, 29010 Malaga, Spain
| | - Anna Marco-Ramell
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| | - Antonio Miñarro
- Genetics, Microbiology and Statistics Department, Biology Faculty, University of Barcelona, 08028 Barcelona, Spain
| | - Olga Jauregui
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- Scientific and Technological Centres of the University of Barcelona (CCIT-UB), 08028 Barcelona, Spain
| | - Raul Gonzalez-Dominguez
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| | - Alex Sanchez-Pla
- Genetics, Microbiology and Statistics Department, Biology Faculty, University of Barcelona, 08028 Barcelona, Spain
- Statistics and Bioinformatics Unit, Vall d’Hebron Institut de Recerca [VHIR], 08035 Barcelona, Spain
| | - Bruno Ramos-Molina
- Biomedical Research Institute [IBIMA], Service of Endocrinology and Nutrition, Malaga Hospital Complex [Virgen de la Victoria], Campus de Teatinos s/n, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición [CIBERobn], Instituto de Salud Carlos III [ISCIII], 28029 Barcelona, Spain
| | - Francisco J. Tinahones
- Biomedical Research Institute [IBIMA], Service of Endocrinology and Nutrition, Malaga Hospital Complex [Virgen de la Victoria], Campus de Teatinos s/n, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición [CIBERobn], Instituto de Salud Carlos III [ISCIII], 28029 Barcelona, Spain
| | - Cristina Andres-Lacueva
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| |
Collapse
|
849
|
The Microbiome and Metabolome of Preterm Infant Stool Are Personalized and Not Driven by Health Outcomes, Including Necrotizing Enterocolitis and Late-Onset Sepsis. mSphere 2018; 3:3/3/e00104-18. [PMID: 29875143 PMCID: PMC5990886 DOI: 10.1128/msphere.00104-18] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/17/2018] [Indexed: 01/28/2023] Open
Abstract
Preterm infants face health problems likely related to microbial exposures, including sepsis and necrotizing enterocolitis. However, the role of the gut microbiome in preterm infant health is poorly understood. Microbial colonization differs from that of healthy term babies because it occurs in the NICU and is often perturbed by antibiotics. We measured bacterial compositions and metabolomic profiles of 77 fecal samples from 32 preterm infants to investigate the differences between microbiomes in health and disease. Rather than finding microbial signatures of disease, we found that both the preterm infant microbiome and the metabolome were personalized and that the preterm infant gut microbiome is enriched in microbes that commonly dominate in the presence of antibiotics. These results contribute to the growing knowledge of the preterm infant microbiome and emphasize that a personalized view will be important to disentangle the health consequences of the preterm infant microbiome. The assembly and development of the gut microbiome in infants have important consequences for immediate and long-term health. Preterm infants represent an abnormal case for bacterial colonization because of early exposure to bacteria and frequent use of antibiotics. To better understand the assembly of the gut microbiota in preterm infants, fecal samples were collected from 32 very low birth weight preterm infants over the first 6 weeks of life. Infant health outcomes included health, late-onset sepsis, and necrotizing enterocolitis (NEC). We characterized bacterial compositions by 16S rRNA gene sequencing and metabolomes by untargeted gas chromatography-mass spectrometry. Preterm infant fecal samples lacked beneficial Bifidobacterium spp. and were dominated by Enterobacteriaceae, Enterococcus, and Staphylococcus organisms due to nearly uniform antibiotic administration. Most of the variance between the microbial community compositions could be attributed to the baby from which the sample derived (permutational multivariate analysis of variance [PERMANOVA] R2 = 0.48, P < 0.001), while clinical status (health, NEC, or late-onset sepsis) and overlapping times in the neonatal intensive care unit (NICU) did not explain a significant amount of variation in bacterial composition. Fecal metabolomes were also found to be unique to the individual (PERMANOVA R2 = 0.43, P < 0.001) and weakly associated with bacterial composition (Mantel statistic r = 0.23 ± 0.05, P < 0.05). No measured metabolites were found to be associated with necrotizing enterocolitis, late-onset sepsis, or a healthy outcome. Overall, preterm infant gut microbial communities were personalized and reflected antibiotic usage. IMPORTANCE Preterm infants face health problems likely related to microbial exposures, including sepsis and necrotizing enterocolitis. However, the role of the gut microbiome in preterm infant health is poorly understood. Microbial colonization differs from that of healthy term babies because it occurs in the NICU and is often perturbed by antibiotics. We measured bacterial compositions and metabolomic profiles of 77 fecal samples from 32 preterm infants to investigate the differences between microbiomes in health and disease. Rather than finding microbial signatures of disease, we found that both the preterm infant microbiome and the metabolome were personalized and that the preterm infant gut microbiome is enriched in microbes that commonly dominate in the presence of antibiotics. These results contribute to the growing knowledge of the preterm infant microbiome and emphasize that a personalized view will be important to disentangle the health consequences of the preterm infant microbiome.
Collapse
|
850
|
Pimentel G, Burton KJ, von Ah U, Bütikofer U, Pralong FP, Vionnet N, Portmann R, Vergères G. Metabolic Footprinting of Fermented Milk Consumption in Serum of Healthy Men. J Nutr 2018; 148:851-860. [PMID: 29788433 PMCID: PMC5991204 DOI: 10.1093/jn/nxy053] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/20/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Background Fermentation is a widely used method of natural food preservation that has consequences on the nutritional value of the transformed food. Fermented dairy products are increasingly investigated in view of their ability to exert health benefits beyond their nutritional qualities. Objective To explore the mechanisms underpinning the health benefits of fermented dairy intake, the present study followed the effects of milk fermentation, from changes in the product metabolome to consequences on the human serum metabolome after its ingestion. Methods A randomized crossover study design was conducted in 14 healthy men [mean age: 24.6 y; mean body mass index (in kg/m2): 21.8]. At the beginning of each test phase, serum samples were taken 6 h postprandially after the ingestion of 800 g of a nonfermented milk or a probiotic yogurt. During the 2-wk test phases, subjects consumed 400 g of the assigned test product daily (200 g, 2 times/d). Serum samples were taken from fasting participants at the end of each test phase. The serum metabolome was assessed through the use of LC-MS-based untargeted metabolomics. Results Postprandial serum metabolomes after milk or yogurt intake could be differentiated [orthogonal projections to latent structures discriminant analysis (OPLS-DA) Q2 = 0.74]. Yogurt intake was characterized by higher concentrations of 7 free amino acids (including proline, P = 0.03), reduced concentrations of 5 bile acids (including glycocholic acid, P = 0.04), and modulation of 4 indole derivative compounds (including indole lactic acid, P = 0.01). Fasting serum samples after 2 wk of daily intake of milk or yogurt could also be differentiated based on their metabolic profiles (OPLS-DA Q2 = 0.56) and were discussed in light of the postprandial results. Conclusion Metabolic pathways related to amino acids, indole derivatives, and bile acids were modulated in healthy men by the intake of yogurt. Further investigation to explore novel health effects of fermented dairy products is warranted.This trial was registered at clinicaltrials.gov as NCT02230345.
Collapse
Affiliation(s)
- Grégory Pimentel
- Federal Department of Economic Affairs, Education and Research (EAER), Agroscope, Bern, Switzerland
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Kathryn J Burton
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Ueli von Ah
- Federal Department of Economic Affairs, Education and Research (EAER), Agroscope, Bern, Switzerland
| | - Ueli Bütikofer
- Federal Department of Economic Affairs, Education and Research (EAER), Agroscope, Bern, Switzerland
| | - François P Pralong
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Nathalie Vionnet
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Reto Portmann
- Federal Department of Economic Affairs, Education and Research (EAER), Agroscope, Bern, Switzerland
| | - Guy Vergères
- Federal Department of Economic Affairs, Education and Research (EAER), Agroscope, Bern, Switzerland
| |
Collapse
|