101
|
Jiang X, Shi P, Jiang L, Qiu J, Xu B, Pan Y, Zhou Q. In vivo toxicity evaluations of halophenolic disinfection byproducts in drinking water: A multi-omics analysis of toxic mechanisms. WATER RESEARCH 2022; 218:118431. [PMID: 35468502 DOI: 10.1016/j.watres.2022.118431] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/18/2022] [Accepted: 04/06/2022] [Indexed: 06/14/2023]
Abstract
Halophenolic disinfection byproducts (DBPs) in drinking water have attracted considerable concerns in recent years due to their wide occurrence and high toxicity. The liver has been demonstrated as a major target organ for several halophenolic DBPs. However, little is known about the underlying mechanisms of liver damage caused by halophenolic DBPs. In this study, 2,4,6-trichlorophenol (TCP), 2,4,6-tribromophenol (TBP) and 2,4,6-triiodiophenol (TIP) were selected as representative halophenolic DBPs and exposed to C57BL/6 mice at an environmentally-relevant concentration (0.5 μg/L) and two toxicological concentrations (10 and 200 μg/L) for 12 weeks. Then, a combination of histopathologic and biochemical examination, liver transcriptome, serum metabolome, and gut microbiome was adopted. It was found that trihalophenol exposure significantly elevated the serum levels of alkaline phosphatase and albumin. Liver inflammation was observed at toxicological concentrations in the histopathological examination. Transcriptome results showed that the three trihalophenols could impact immune-related pathways at 0.5 μg/L, which further contributed to the disturbance of pathways in infectious diseases and cancers. Notably, TBP and TIP had higher immunosuppressive effects than TCP, which might lead to uncontrolled infection and cancer. In terms of serum metabolic profiles, energy metabolism pathway of citrate cycle and amino acid metabolism pathways of valine, leucine, and isoleucine were also significantly affected. Integration of the metabolomic and transcriptomic data suggested that a 12-week trihalophenol exposure could prominently disturb the glutathione metabolism pathway, indicating the impaired antioxidation and detoxification abilities in liver. Moreover, the disorder of the intestinal flora could interfere with immune regulation and host metabolism. This study reveals the toxic effects of halophenolic DBPs on mammalian liver and provides novel insights into the underlying mechanisms of hepatotoxicity.
Collapse
Affiliation(s)
- Xiaoqin Jiang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Peng Shi
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Liujing Jiang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Jingfan Qiu
- Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Bin Xu
- State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Yang Pan
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu, China.
| | - Qing Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
102
|
Meta-Analysis of Altered Gut Microbiota Reveals Microbial and Metabolic Biomarkers for Colorectal Cancer. Microbiol Spectr 2022; 10:e0001322. [PMID: 35766483 PMCID: PMC9431300 DOI: 10.1128/spectrum.00013-22] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer mortality worldwide. The dysbiotic gut microbiota and its metabolite secretions play a significant role in CRC development and progression. In this study, we identified microbial and metabolic biomarkers applicable to CRC using a meta-analysis of metagenomic datasets from diverse geographical regions. We used LEfSe, random forest (RF), and co-occurrence network methods to identify microbial biomarkers. Geographic dataset-specific markers were identified and evaluated using area under the ROC curve (AUC) scores and random effect size. Co-occurrence networks analysis showed a reduction in the overall microbial associations and the presence of oral pathogenic microbial clusters in CRC networks. Analysis of predicted metabolites from CRC datasets showed the enrichment of amino acids, cadaverine, and creatine in CRC, which were positively correlated with CRC-associated microbes (Peptostreptococcus stomatis, Gemella morbillorum, Bacteroides fragilis, Parvimonas spp., Fusobacterium nucleatum, Solobacterium moorei, and Clostridium symbiosum), and negatively correlated with control-associated microbes. Conversely, butyrate, nicotinamide, choline, tryptophan, and 2-hydroxybutanoic acid showed positive correlations with control-associated microbes (P < 0.05). Overall, our study identified a set of global CRC biomarkers that are reproducible across geographic regions. We also reported significant differential metabolites and microbe-metabolite interactions associated with CRC. This study provided significant insights for further investigations leading to the development of noninvasive CRC diagnostic tools and therapeutic interventions. IMPORTANCE Several studies showed associations between gut dysbiosis and CRC. Yet, the results are not conclusive due to cohort-specific associations that are influenced by genomic, dietary, and environmental stimuli and associated reproducibility issues with various analysis approaches. Emerging evidence suggests the role of microbial metabolites in modulating host inflammation and DNA damage in CRC. However, the experimental validations have been hindered by cost, resources, and cumbersome technical expertise required for metabolomic investigations. In this study, we performed a meta-analysis of CRC microbiota data from diverse geographical regions using multiple methods to achieve reproducible results. We used a computational approach to predict the metabolomic profiles using existing CRC metagenomic datasets. We identified a reliable set of CRC-specific biomarkers from this analysis, including microbial and metabolite markers. In addition, we revealed significant microbe-metabolite associations through correlation analysis and microbial gene families associated with dysregulated metabolic pathways in CRC, which are essential in understanding the vastly sporadic nature of CRC development and progression.
Collapse
|
103
|
Monfort-Ferré D, Caro A, Menacho M, Martí M, Espina B, Boronat-Toscano A, Nuñez-Roa C, Seco J, Bautista M, Espín E, Megía A, Vendrell J, Fernández-Veledo S, Serena C. The Gut Microbiota Metabolite Succinate Promotes Adipose Tissue Browning in Crohn's Disease. J Crohns Colitis 2022; 16:1571-1583. [PMID: 35554517 PMCID: PMC9624294 DOI: 10.1093/ecco-jcc/jjac069] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/17/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Crohn's disease [CD] is associated with complex microbe-host interactions, involving changes in microbial communities, and gut barrier defects, leading to the translocation of microorganisms to surrounding adipose tissue [AT]. We evaluated the presence of beige AT depots in CD and questioned whether succinate and/or bacterial translocation promotes white-to-beige transition in adipocytes. METHODS Visceral [VAT] and subcutaneous [SAT] AT biopsies, serum and plasma were obtained from patients with active [n = 21] or inactive [n = 12] CD, and from healthy controls [n = 15]. Adipose-derived stem cells [ASCs] and AT macrophages [ATMs] were isolated from VAT biopsies. RESULTS Plasma succinate levels were significantly higher in patients with active CD than in controls and were intermediate in those with inactive disease. Plasma succinate correlated with the inflammatory marker high-sensitivity C-reactive protein. Expression of the succinate receptor SUCNR1 was higher in VAT, ASCs and ATMs from the active CD group than from the inactive or control groups. Succinate treatment of ASCs elevated the expression of several beige AT markers from controls and from patients with inactive disease, including uncoupling protein-1 [UCP1]. Notably, beige AT markers were prominent in ASCs from patients with active CD. Secretome profiling revealed that ASCs from patients with active disease secrete beige AT-related proteins, and co-culture assays showed that bacteria also trigger the white-to-beige switch of ASCs from patients with CD. Finally, AT depots from patients with CD exhibited a conversion from white to beige AT together with high UCP1 expression, which was corroborated by in situ thermal imaging analysis. CONCLUSIONS Succinate and bacteria trigger white-to-beige AT transition in CD. Understanding the role of beige AT in CD might aid in the development of therapeutic or diagnostic interventions.
Collapse
Affiliation(s)
- Diandra Monfort-Ferré
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Aleidis Caro
- Colorectal Surgery Unit, Hospital Universitari Joan XXIII, Tarragona, Spain
| | | | - Marc Martí
- Colorectal Surgery Unit, General Surgery Service, Hospital Valle de Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Beatriz Espina
- Colorectal Surgery Unit, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Albert Boronat-Toscano
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Cati Nuñez-Roa
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Seco
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Michelle Bautista
- Digestive Unit, Hospital Universitari Joan XXIII, 43007, Tarragona, Spain
| | - Eloy Espín
- Colorectal Surgery Unit, General Surgery Service, Hospital Valle de Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Ana Megía
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Vendrell
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain,Universitat Rovira i Virgili, Tarragona, Spain
| | - Sonia Fernández-Veledo
- Corresponding authors: Sonia Fernández-Veledo, PhD, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain. ;
| | - Carolina Serena
- Carolina Serena, PhD, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain. ;
| |
Collapse
|
104
|
Gut microbial metabolites in Parkinson's disease: Association with lifestyle, disease characteristics, and treatment status. Neurobiol Dis 2022; 170:105780. [PMID: 35654277 PMCID: PMC9241494 DOI: 10.1016/j.nbd.2022.105780] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022] Open
Abstract
There is growing appreciation of the importance of the intestinal microbiota in Parkinson’s disease (PD), and one potential mechanism by which the intestinal microbiota can communicate with the brain is via bacteria-derived metabolites. In this study, plasma levels of bacterial-derived metabolites including trimethylamine-N-oxide (TMAO), short chain fatty acids (SCFA), the branched chain fatty acid isovalerate, succinate, and lactate were evaluated in PD subjects (treatment naïve and treated) which were compared to (1) population controls, (2) spousal / household controls (similar lifestyle to PD subjects), and (3) subjects with multiple system atrophy (MSA). Analyses revealed an increase in the TMAO pathway in PD subjects which was independent of medication status, disease characteristics, and lifestyle. Lactic acid was decreased in treated PD subjects, succinic acid positively correlated with disease severity, and the ratio of pro-inflammatory TMAO to the putative anti-inflammatory metabolite butyric acid was significantly higher in PD subjects compared to controls indicating a pro-inflammatory shift in the metabolite profile in PD subjects. Finally, acetic and butyric acid were different between PD and MSA subjects indicating that metabolites may differentiate these synucleinopathies. In summary, (1) TMAO is elevated in PD subjects, a phenomenon independent of disease characteristics, treatment status, and lifestyle and (2) metabolites may differentiate PD and MSA subjects. Additional studies to understand the potential of TMAO and other bacterial metabolites to serve as a biomarker or therapeutic targets are warranted.
Collapse
|
105
|
Abstract
The gut microbiota is now considered as one of the key elements contributing to the regulation of host health. Virtually all our body sites are colonised by microbes suggesting different types of crosstalk with our organs. Because of the development of molecular tools and techniques (ie, metagenomic, metabolomic, lipidomic, metatranscriptomic), the complex interactions occurring between the host and the different microorganisms are progressively being deciphered. Nowadays, gut microbiota deviations are linked with many diseases including obesity, type 2 diabetes, hepatic steatosis, intestinal bowel diseases (IBDs) and several types of cancer. Thus, suggesting that various pathways involved in immunity, energy, lipid and glucose metabolism are affected.In this review, specific attention is given to provide a critical evaluation of the current understanding in this field. Numerous molecular mechanisms explaining how gut bacteria might be causally linked with the protection or the onset of diseases are discussed. We examine well-established metabolites (ie, short-chain fatty acids, bile acids, trimethylamine N-oxide) and extend this to more recently identified molecular actors (ie, endocannabinoids, bioactive lipids, phenolic-derived compounds, advanced glycation end products and enterosynes) and their specific receptors such as peroxisome proliferator-activated receptor alpha (PPARα) and gamma (PPARγ), aryl hydrocarbon receptor (AhR), and G protein-coupled receptors (ie, GPR41, GPR43, GPR119, Takeda G protein-coupled receptor 5).Altogether, understanding the complexity and the molecular aspects linking gut microbes to health will help to set the basis for novel therapies that are already being developed.
Collapse
Affiliation(s)
- Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| |
Collapse
|
106
|
Díez-Sainz E, Milagro FI, Riezu-Boj JI, Lorente-Cebrián S. Effects of gut microbiota-derived extracellular vesicles on obesity and diabetes and their potential modulation through diet. J Physiol Biochem 2022; 78:485-499. [PMID: 34472032 PMCID: PMC8410452 DOI: 10.1007/s13105-021-00837-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022]
Abstract
Obesity and diabetes incidence rates are increasing dramatically, reaching pandemic proportions. Therefore, there is an urgent need to unravel the mechanisms underlying their pathophysiology. Of particular interest is the close interconnection between gut microbiota dysbiosis and obesity and diabetes progression. Hence, microbiota manipulation through diet has been postulated as a promising therapeutic target. In this regard, secretion of gut microbiota-derived extracellular vesicles is gaining special attention, standing out as key factors that could mediate gut microbiota-host communication. Extracellular vesicles (EVs) derived from gut microbiota and probiotic bacteria allow to encapsulate a wide range of bioactive molecules (such as/or including proteins and nucleic acids) that could travel short and long distances to modulate important biological functions with the overall impact on the host health. EV-derived from specific bacteria induce differential physiological responses. For example, a high-fat diet-induced increase of the proteobacterium Pseudomonas panacis-derived EV is closely associated with the progression of metabolic dysfunction in mice. In contrast, Akkermansia muciniphila EV are linked with the alleviation of high-fat diet-induced obesity and diabetes in mice. Here, we review the newest pieces of evidence concerning the potential role of gut microbiota and probiotic-derived EV on obesity and diabetes onset, progression, and management, through the modulation of inflammation, metabolism, and gut permeability. In addition, we discuss the role of certain dietary patterns on gut microbiota-derived EV profile and the clinical implication that dietary habits could have on metabolic diseases progression through the shaping of gut microbiota-derived EV.
Collapse
Affiliation(s)
- Ester Díez-Sainz
- Department of Nutrition, Food Science and Physiology/Center for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Fermín I Milagro
- Department of Nutrition, Food Science and Physiology/Center for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red Fisiopatología de La Obesidad Y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.
| | - José I Riezu-Boj
- Department of Nutrition, Food Science and Physiology/Center for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Silvia Lorente-Cebrián
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Health and Sport Science, University of Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain
- Aragón Health Research Institute (IIS-Aragon), Zaragoza, Spain
| |
Collapse
|
107
|
Abstract
The gut microbiota is now considered as one of the key elements contributing to the regulation of host health. Virtually all our body sites are colonised by microbes suggesting different types of crosstalk with our organs. Because of the development of molecular tools and techniques (ie, metagenomic, metabolomic, lipidomic, metatranscriptomic), the complex interactions occurring between the host and the different microorganisms are progressively being deciphered. Nowadays, gut microbiota deviations are linked with many diseases including obesity, type 2 diabetes, hepatic steatosis, intestinal bowel diseases (IBDs) and several types of cancer. Thus, suggesting that various pathways involved in immunity, energy, lipid and glucose metabolism are affected.In this review, specific attention is given to provide a critical evaluation of the current understanding in this field. Numerous molecular mechanisms explaining how gut bacteria might be causally linked with the protection or the onset of diseases are discussed. We examine well-established metabolites (ie, short-chain fatty acids, bile acids, trimethylamine N-oxide) and extend this to more recently identified molecular actors (ie, endocannabinoids, bioactive lipids, phenolic-derived compounds, advanced glycation end products and enterosynes) and their specific receptors such as peroxisome proliferator-activated receptor alpha (PPARα) and gamma (PPARγ), aryl hydrocarbon receptor (AhR), and G protein-coupled receptors (ie, GPR41, GPR43, GPR119, Takeda G protein-coupled receptor 5).Altogether, understanding the complexity and the molecular aspects linking gut microbes to health will help to set the basis for novel therapies that are already being developed.
Collapse
Affiliation(s)
- Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| |
Collapse
|
108
|
Pérez Escriva P, Fuhrer T, Sauer U. Distinct N and C Cross-Feeding Networks in a Synthetic Mouse Gut Consortium. mSystems 2022; 7:e0148421. [PMID: 35357218 PMCID: PMC9040589 DOI: 10.1128/msystems.01484-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/03/2022] [Indexed: 11/20/2022] Open
Abstract
The complex interactions between the gut microbiome and host or pathogen colonization resistance cannot be understood solely from community composition. Missing are causal relationships, such as metabolic interactions among species, to better understand what shapes the microbiome. Here, we focused on metabolic niches generated and occupied by the Oligo-Mouse-Microbiota (OMM) consortium, a synthetic community composed of 12 members that is increasingly used as a model for the mouse gut microbiome. Combining monocultures and spent medium experiments with untargeted metabolomics revealed broad metabolic diversity in the consortium, constituting a dense cross-feeding network with more than 100 pairwise interactions. Quantitative analysis of the cross-feeding network revealed distinct C and N food webs, highlighting the two Bacteroidetes members Bacteroides caecimuris and Muribaculum intestinale as primary suppliers of carbon and a more diverse group as nitrogen providers. Cross-fed metabolites were mainly carboxylic acids, amino acids, and the so far not reported nucleobases. In particular, the dicarboxylic acids malate and fumarate provided a strong physiological benefit to consumers, presumably used in anaerobic respiration. Isotopic tracer experiments validated the fate of a subset of cross-fed metabolites, such as the conversion of the most abundant cross-fed compound succinate to butyrate. Thus, we show that this consortium is tailored to produce the anti-inflammatory metabolite butyrate. Overall, we provide evidence for metabolic niches generated and occupied by OMM members that lays a metabolic foundation to facilitate an understanding of the more complex in vivo behavior of this consortium in the mouse gut. IMPORTANCE This article maps out the cross-feeding network among 10 members of a synthetic consortium that is increasingly used as the model mouse gut microbiota. Combining metabolomics with in vitro cultivations, two dense networks of carbon and nitrogen exchange are described. The vast majority of the ∼100 interactions are synergistic in nature, in several cases providing distinct physiological benefits to the recipient species. These networks lay the groundwork toward understanding gut community dynamics and host-gut microbe interactions.
Collapse
Affiliation(s)
- Pau Pérez Escriva
- Institute of Molecular Systems Biology, D-BIOL, ETH Zurich, Zurich, Switzerland
- Systems Biology Graduate School, Zurich, Switzerland
| | - Tobias Fuhrer
- Institute of Molecular Systems Biology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, D-BIOL, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
109
|
Schubert C, Unden G. C 4-Dicarboxylates as Growth Substrates and Signaling Molecules for Commensal and Pathogenic Enteric Bacteria in Mammalian Intestine. J Bacteriol 2022; 204:e0054521. [PMID: 34978458 PMCID: PMC9017328 DOI: 10.1128/jb.00545-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The C4-dicarboxylates (C4-DC) l-aspartate and l-malate have been identified as playing an important role in the colonization of mammalian intestine by enteric bacteria, such as Escherichia coli and Salmonella enterica serovar Typhimurium, and succinate as a signaling molecule for host-enteric bacterium interaction. Thus, endogenous and exogenous fumarate respiration and related functions are required for efficient initial growth of the bacteria. l-Aspartate represents a major substrate for fumarate respiration in the intestine and a high-quality substrate for nitrogen assimilation. During nitrogen assimilation, DcuA catalyzes an l-aspartate/fumarate antiport and serves as a nitrogen shuttle for the net uptake of ammonium only, whereas DcuB acts as a redox shuttle that catalyzes the l-malate/succinate antiport during fumarate respiration. The C4-DC two-component system DcuS-DcuR is active in the intestine and responds to intestinal C4-DC levels. Moreover, in macrophages and in mice, succinate is a signal that promotes virulence and survival of S. Typhimurium and pathogenic E. coli. On the other hand, intestinal succinate is an important signaling molecule for the host and activates response and protective programs. Therefore, C4-DCs play a major role in supporting colonization of enteric bacteria and as signaling molecules for the adaptation of host physiology.
Collapse
Affiliation(s)
- Christopher Schubert
- Institute for Molecular Physiology (IMP), Microbiology and Wine Research, Johannes Gutenberg University, Mainz, Germany
| | - Gottfried Unden
- Institute for Molecular Physiology (IMP), Microbiology and Wine Research, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
110
|
Imdad S, Lim W, Kim JH, Kang C. Intertwined Relationship of Mitochondrial Metabolism, Gut Microbiome and Exercise Potential. Int J Mol Sci 2022; 23:ijms23052679. [PMID: 35269818 PMCID: PMC8910986 DOI: 10.3390/ijms23052679] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
The microbiome has emerged as a key player contributing significantly to the human physiology over the past decades. The potential microbial niche is largely unexplored in the context of exercise enhancing capacity and the related mitochondrial functions. Physical exercise can influence the gut microbiota composition and diversity, whereas a sedentary lifestyle in association with dysbiosis can lead to reduced well-being and diseases. Here, we have elucidated the importance of diverse microbiota, which is associated with an individual's fitness, and moreover, its connection with the organelle, the mitochondria, which is the hub of energy production, signaling, and cellular homeostasis. Microbial by-products, such as short-chain fatty acids, are produced during regular exercise that can enhance the mitochondrial capacity. Therefore, exercise can be employed as a therapeutic intervention to circumvent or subside various metabolic and mitochondria-related diseases. Alternatively, the microbiome-mitochondria axis can be targeted to enhance exercise performance. This review furthers our understanding about the influence of microbiome on the functional capacity of the mitochondria and exercise performance, and the interplay between them.
Collapse
Affiliation(s)
- Saba Imdad
- Molecular Metabolism in Health & Disease, Exercise Physiology Laboratory, Sport Science Research Institute, Inha University, Incheon 22212, Korea;
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Korea
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju 28503, Korea;
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Korea
- Correspondence: (J.-H.K.); (C.K.)
| | - Chounghun Kang
- Molecular Metabolism in Health & Disease, Exercise Physiology Laboratory, Sport Science Research Institute, Inha University, Incheon 22212, Korea;
- Department of Physical Education, College of Education, Inha University, Incheon 22212, Korea
- Correspondence: (J.-H.K.); (C.K.)
| |
Collapse
|
111
|
Zhang FQ, McMullen JG, Douglas AE, Ankrah NYD. Succinate: A microbial product that modulates Drosophila nutritional physiology. INSECT SCIENCE 2022; 29:315-318. [PMID: 33629498 PMCID: PMC8382807 DOI: 10.1111/1744-7917.12905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/10/2021] [Accepted: 01/17/2021] [Indexed: 05/16/2023]
Affiliation(s)
| | | | - Angela E Douglas
- Department of Entomology
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Nana Y D Ankrah
- Department of Entomology
- Department of Biological Sciences, State University of New York at Plattsburgh, Plattsburgh, NY, 12901, USA
| |
Collapse
|
112
|
Tochitani S, Maehara Y, Kawase T, Tsukahara T, Shimizu R, Watanabe T, Maehara K, Asaoka K, Matsuzaki H. Fermented rice bran supplementation ameliorates obesity via gut microbiota and metabolism modification in female mice. J Clin Biochem Nutr 2022; 70:160-174. [PMID: 35400825 PMCID: PMC8921717 DOI: 10.3164/jcbn.21-96] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/21/2021] [Indexed: 12/12/2022] Open
Abstract
We investigated the effects of fermented rice bran (FRB) administration in two groups of C57BL/6J mice. The first group was fed with a high-fat diet, and the second group was fed with a high-fat diet supplemented with the FRB for 8 weeks. FRB supplementation suppressed the high-fat-induced weight gain and considerable alterations in the intestinal microbiota profile in the second group. Among 27 bacterial genera detected in the FRB, only Enterococcus, Lactobacillus, Bacteroides, Prevotella, and the unclassified family Peptostreptococcaceae were detected in mice feces. Their abundances were not particularly increased by FRB supplementation. The abundances of Enterococcus and the unclassified family Peptostreptococcaceae were even suppressed in the second group, suggesting that FRB supplementation didn’t cause an addition of beneficial microbiome but inhibit the proliferation of specific bacteria. Fecal succinic acid concentration was significantly decreased in the second group and highly correlated with the relative abundances of Turicibacter, Enterococcus, and the unclassified family Peptostreptococcaceae. A significant increase in fumaric acid and a decrease in xylitol, sorbitol, uracil, glutamic acid, and malic acid levels were observed in the peripheral blood of the second group. FRB supplementation counteracted the high-fat-induced obesity in mice by modulating the gut microbiota and the host metabolism.
Collapse
Affiliation(s)
- Shiro Tochitani
- Department of Radiological Technology, Faculty of Health Science, Suzuka University of Medical Science
| | | | | | | | | | | | | | | | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui
| |
Collapse
|
113
|
Li JS, Su SL, Xu Z, Zhao LH, Fan RY, Guo JM, Qian DW, Duan JA. Potential roles of gut microbiota and microbial metabolites in chronic inflammatory pain and the mechanisms of therapy drugs. Ther Adv Chronic Dis 2022; 13:20406223221091177. [PMID: 35924009 PMCID: PMC9340317 DOI: 10.1177/20406223221091177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/15/2022] [Indexed: 01/21/2023] Open
Abstract
Observational findings achieved that gut microbes mediate human metabolic health
and disease risk. The types of intestinal microorganisms depend on the intake of
food and drugs and are also related to their metabolic level and genetic
factors. Recent studies have shown that chronic inflammatory pain is closely
related to intestinal microbial homeostasis. Compared with the normal intestinal
flora, the composition of intestinal flora in patients with chronic inflammatory
pain had significant changes in Actinomycetes,
Firmicutes, Bacteroidetes, etc. At the
same time, short-chain fatty acids and amino acids, the metabolites of
intestinal microorganisms, can regulate neural signal molecules and signaling
pathways, thus affecting the development trend of chronic inflammatory pain.
Glucocorticoids and non-steroidal anti-inflammatory drugs in the treatment of
chronic inflammatory pain, the main mechanism is to affect the secretion of
inflammatory factors and the abundance of intestinal bacteria. This article
reviews the relationship between intestinal microorganisms and their metabolites
on chronic inflammatory pain and the possible mechanism.
Collapse
Affiliation(s)
- Jia-Shang Li
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | | | - Zhuo Xu
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Li-Hui Zhao
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Ruo-Ying Fan
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Jian-Ming Guo
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Da-Wei Qian
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, P.R. China
| |
Collapse
|
114
|
Gutiérrez-Repiso C, Moreno-Indias I, Tinahones FJ. Shifts in gut microbiota and their metabolites induced by bariatric surgery. Impact of factors shaping gut microbiota on bariatric surgery outcomes. Rev Endocr Metab Disord 2021; 22:1137-1156. [PMID: 34287758 DOI: 10.1007/s11154-021-09676-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
Evidence suggests that bariatric surgery alters gut microbiota, although its impact at compositional and functional level is not well described. In this review, the most relevant findings, mainly described in Roux-en-Y gastric bypass and sleeve gastrectomy, are outlined. Although the number of studies has increased in the last years, conclusive assertions cannot be elaborated. An issue to address is to know the influence of these alterations on host metabolism and the contribution of gut microbiota derived metabolites. New lines of research have been focusing on analysing gut microbiota functionality rather than evaluating changes at compositional level, and the functions of gut microbiota metabolites in host metabolism, what will bring more relevant information about the influence of gut microbiota in bariatric surgery outcomes. Personalized medicine, because of the predictive value of gut microbiota, is another promising field. The possibility of a specific gut microbiota pattern that could predict type 2 diabetes remission or weight loss failure after bariatric surgery is a matter of great interest. However, little is known about how gut microbiota manipulation could contribute to the beneficial effects of bariatric surgery. Peri-operative antibiotics prophylaxis or probiotic supplementation early after surgery, are strategies barely studied so far, and could constitute a novel tool in the management of weight loss and metabolic profile improvement after surgery.
Collapse
Affiliation(s)
- Carolina Gutiérrez-Repiso
- Unidad de Gestión Clínica de Endocrinología Y Nutrición del Hospital Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA) Málaga. Centro de Investigación Biomédica en Red de Fisiopatología de La Obesidad Y La Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.
| | - Isabel Moreno-Indias
- Unidad de Gestión Clínica de Endocrinología Y Nutrición del Hospital Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA) Málaga. Centro de Investigación Biomédica en Red de Fisiopatología de La Obesidad Y La Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J Tinahones
- Unidad de Gestión Clínica de Endocrinología Y Nutrición del Hospital Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA) Málaga. Centro de Investigación Biomédica en Red de Fisiopatología de La Obesidad Y La Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina Y Dermatología, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
115
|
Li Y, Cao H, Wang X, Guo L, Ding X, Zhao W, Zhang F. Diet-mediated metaorganismal relay biotransformation: health effects and pathways. Crit Rev Food Sci Nutr 2021:1-19. [PMID: 34802351 DOI: 10.1080/10408398.2021.2004993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In recent years, the concept of metaorganism expands our insight into how diet-microbe-host interactions contribute to human health and diseases. We realized that many biological metabolic processes in the host can be summarized into metaorganismal relay pathways, in which metabolites such as trimethylamine-N‑oxide, short-chain fatty acids and bile acids act as double-edged swords (beneficial or harmful effects) in the initiation and progression of diseases. Pleiotropic effects of metabolites are derived from several influencing factors including dose level, targeted organ of effect, action duration and species of these metabolites. Based on the pleiotropic effects of metabolites, personalized therapeutic approaches including microecological agents, enzymatic regulators and changes in dietary habits to govern related metabolite production may provide a new insight in promoting human health. In this review, we summarize our current knowledge of metaorganismal relay pathways and elaborate on the pleiotropic effects of metabolites in these pathways, with special emphasis on related therapeutic nutritional interventions.
Collapse
Affiliation(s)
- Yanmin Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hong Cao
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaoqian Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Lichun Guo
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoying Ding
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Feng Zhang
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
116
|
Abstract
Prednisone (PRED) is a synthetic glucocorticoid (GC) widely used in immune-mediated diseases for its immunosuppressive and anti-inflammatory properties. The effects of GC are achieved by genomic and nongenomic mechanisms. However, the nongenomic effects are largely unknown. Thus, we aimed to investigate how long-term prednisone therapy changes the composition of the gut microbiota and fecal metabolites in rats. Male Sprague-Dawley rats were randomly assigned to a control (CON) group and a PRED group, which received prednisone treatment daily for 6 weeks by gavage. The V3 to V4 regions of bacterial 16S rRNA genes were amplified and sequenced after the total bacterial DNA was extracted from fecal samples. The alpha and beta diversities were calculated. The compositional alteration of the gut microbiota at different taxonomic levels was analyzed using the Metastats method. Meanwhile, the fecal metabolites were quantitated in an ultra-performance liquid chromatography system. Similar microbial richness and diversity between the CON and PRED groups were indicated by the alpha diversity results. The gut microbial communities differed significantly between two groups. The relative abundances of the genera Eisenbergiella, Alistipes, and Clostridium XIVb decreased, whereas that of Anaerobacterium increased significantly in rats after the 6-week prednisone treatment. In total, 11 downregulated and 10 upregulated fecal metabolites were identified. Differential fecal metabolites were enriched in the pathways, including phenylalanine metabolism, butanoate metabolism, and propanoate metabolism. The lowered production of short-chain fatty acids was associated with the decreased relative abundance of the genera Alistipes and Clostridium XIVb and increased abundance of the genus Anaerobacterium. The composition of the gut microbiota and fecal metabolites was changed after long-term prednisone treatment. This may help us to understand the pharmacology of prednisone. IMPORTANCE Prednisone is widely used in chronic glomerular diseases, immunological disorders, and rheumatic diseases for its anti-inflammatory and immunosuppressive properties. It is a synthetic glucocorticoid (GC) that shows therapeutic effects after conversion to prednisolone by the liver. Prolonged GC therapy causes anti-inflammatory effects; it also results in a variety of adverse events, including obesity, hypertension, psychiatric symptoms, and dyslipidemia. The therapeutic effects and adverse events of GCs may be associated with changes in the gut microbiota, as the host might be affected by the metabolites generated by the altered gut microbes. Thus, we investigated how long-term prednisone therapy changed the composition of the gut microbiota and fecal metabolites in rats. This study may shed new light on the pharmacology of prednisone.
Collapse
|
117
|
Cui G, Qing Y, Li M, Sun L, Zhang J, Feng L, Li J, Chen T, Wang J, Wan C. Salivary Metabolomics Reveals that Metabolic Alterations Precede the Onset of Schizophrenia. J Proteome Res 2021; 20:5010-5023. [PMID: 34618462 DOI: 10.1021/acs.jproteome.1c00504] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a complex and highly heterogeneous mental illness with a prodromal period called clinical high risk (CHR) for psychosis before onset. Metabolomics is greatly promising in analyzing the pathology of complex diseases and exploring diagnostic biomarkers. Therefore, we conducted salivary metabolomics analysis in 83 first-episode schizophrenia (FES) patients, 42 CHR individuals, and 78 healthy controls with ultrahigh-performance liquid chromatography-quadrupole time-of-flight mass spectrometry. The mass spectrometry raw data have been deposited on the MetaboLights (ID: MTBLS3463). We found downregulated aromatic amino acid metabolism, disturbed glutamine and nucleotide metabolism, and upregulated tricarboxylic acid cycle in FES patients, which existed even in the CHR stage and became more intense with the onset of the schizophrenia. Moreover, differential metabolites can be considered as potential diagnostic biomarkers and indicate the severity of the different clinical stages of disease. Furthermore, three disordered pathways were closely related to peripheral indicators of inflammatory response, oxidative stress, blood-brain barrier damage, and salivary microbiota. These results indicate that the disorder of oral metabolism occurs earlier than the onset of schizophrenia and is concentrated and intensified with the onset of disease, which may originate from the dysbiotic salivary microbiota and cause the onset of schizophrenia through the peripheral inflammatory response and redox system, suggesting the importance of oral-brain connection in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Gaoping Cui
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ying Qing
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Minghui Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liya Sun
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Juan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lei Feng
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Li
- Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tianlu Chen
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jijun Wang
- Shanghai Mental Health Centre, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
118
|
An R, Wilms E, Logtenberg MJ, van Trijp MPH, Schols HA, Masclee AAM, Smidt H, Jonkers DMAE, Zoetendal EG. In vitro metabolic capacity of carbohydrate degradation by intestinal microbiota of adults and pre-frail elderly. ISME COMMUNICATIONS 2021; 1:61. [PMID: 36750618 PMCID: PMC9723549 DOI: 10.1038/s43705-021-00065-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 09/27/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022]
Abstract
Globally increased life expectancy strongly triggered interest to delay the onset of frailty, which has been associated with alterations in compositional and functional characteristics of intestinal microbiota. In the current study, we used an in vitro batch incubation model to compare the metabolic capacity of the faecal microbiota of adults (n = 6) versus pre-frail elderly (n = 6) to degrade various glycosidic carbohydrates, including galacto-oligosaccharides, 2'-fucosyllactose, chicory fructo-oligosaccharides and inulin, and isomalto/malto-polysaccharides. The in vitro metabolic capacity was also compared with an in vivo GOS intervention study based on the same subjects. Analysis of 16S rRNA gene sequences and metabolites revealed distinct portions of variation in overall microbiota and metabolite composition during incubation being explained by individuality of the subjects and carbon source. In addition, the age group of the subjects also had significant impact on microbiota variation, carbohydrate degradation and metabolite production. This was accompanied by elevated increase in the relative abundance of Bifidobacterium in the microbiota of adults compared to that of pre-frail elderly and significantly decreased effectiveness to degrade galacto-oligosaccharides by the latter group. Altogether, the carbohydrate degradation in elderly was different compared to adults, with some carbohydrates showing decreased degradation rates. Longer interventions periods may be required to enhance bifidobacterial abundance in the microbiota of pre-frail elderly and thereby to obtain associated prebiotic health benefits.
Collapse
Affiliation(s)
- Ran An
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands.
| | - Ellen Wilms
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Madelon J Logtenberg
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Mara P H van Trijp
- Nutrition, Metabolism & Genomics Group, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Ad A M Masclee
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Daisy M A E Jonkers
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
119
|
TCA cycle remodeling drives proinflammatory signaling in humans with pulmonary tuberculosis. PLoS Pathog 2021; 17:e1009941. [PMID: 34559866 PMCID: PMC8494353 DOI: 10.1371/journal.ppat.1009941] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/06/2021] [Accepted: 09/03/2021] [Indexed: 11/19/2022] Open
Abstract
The metabolic signaling pathways that drive pathologic tissue inflammation and damage in humans with pulmonary tuberculosis (TB) are not well understood. Using combined methods in plasma high-resolution metabolomics, lipidomics and cytokine profiling from a multicohort study of humans with pulmonary TB disease, we discovered that IL-1β-mediated inflammatory signaling was closely associated with TCA cycle remodeling, characterized by accumulation of the proinflammatory metabolite succinate and decreased concentrations of the anti-inflammatory metabolite itaconate. This inflammatory metabolic response was particularly active in persons with multidrug-resistant (MDR)-TB that received at least 2 months of ineffective treatment and was only reversed after 1 year of appropriate anti-TB chemotherapy. Both succinate and IL-1β were significantly associated with proinflammatory lipid signaling, including increases in the products of phospholipase A2, increased arachidonic acid formation, and metabolism of arachidonic acid to proinflammatory eicosanoids. Together, these results indicate that decreased itaconate and accumulation of succinate and other TCA cycle intermediates is associated with IL-1β-mediated proinflammatory eicosanoid signaling in pulmonary TB disease. These findings support host metabolic remodeling as a key driver of pathologic inflammation in human TB disease. Pulmonary tuberculosis (TB) often results in pathologic lung inflammation that causes tissue damage and does not control bacterial replication. This impairs the host response to antibiotic treatment and can result in long term deficits in lung function. Currently, the role of host metabolism in regulating the inflammatory response in TB disease is not well understood. Here, we use detailed immunometabolic phenotyping to show that metabolic remodeling of the tricarboxylic acid (TCA) cycle is closely associated with pathologic inflammatory signaling in humans with TB disease. Accumulation of TCA cycle intermediates in plasma, including the proinflammatory metabolite succinate, as well as decreased concentrations of the anti-inflammatory metabolite itaconate, were associated with increases in IL-1β and upregulation of proinflammatory lipid signaling cascades. This inflammatory network was upregulated following delays in appropriate anti-TB treatment and was associated with prolonged time to sputum culture clearance of Mycobacterium tuberculosis. Our study provides new insights into the metabolic reprograming that leads to pathologic inflammation in humans with pulmonary TB.
Collapse
|
120
|
Fernández-Veledo S, Ceperuelo-Mallafré V, Vendrell J. Rethinking succinate: an unexpected hormone-like metabolite in energy homeostasis. Trends Endocrinol Metab 2021; 32:680-692. [PMID: 34301438 DOI: 10.1016/j.tem.2021.06.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
There has been an explosion of interest in the signaling capacity of energy metabolites. A prime example is the Krebs cycle substrate succinate, an archetypal respiratory substrate with functions beyond energy production as an intracellular and extracellular signaling molecule. Long associated with inflammation, emerging evidence supports a key role for succinate in metabolic processes relating to energy management. As the natural ligand for SUCNR1, a G protein-coupled receptor, succinate is akin to hormones and likely functions as a reporter of metabolism and stress. In this review, we reconcile new and old observations to outline a regulatory role for succinate in metabolic homeostasis. We highlight the importance of the succinate-SUCNR1 axis in metabolic diseases as an integrator of macrophage immune response, and we discuss new metabolic functions recently ascribed to succinate in specific tissues. Because circulating succinate has emerged as a promising biomarker in chronic metabolic diseases, a better understanding of the physiopathological role of the succinate-SUCNR1 axis in metabolism might open new avenues for clinical use in patients with obesity or diabetes.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| | - Victòria Ceperuelo-Mallafré
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine and Surgery, University Rovira I Virgili, Tarragona, Spain
| | - Joan Vendrell
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine and Surgery, University Rovira I Virgili, Tarragona, Spain
| |
Collapse
|
121
|
Kilua A, Pelpolage S, Goto A, Nakayama Y, Kitazono E, Toyohara K, Nagata R, Fukuma N, Han KH, Fukushima M. Deciphering the colonic fermentation characteristics of agavin and digestion-resistant maltodextrin in a simulated batch fermentation system. Int J Biol Macromol 2021; 189:151-159. [PMID: 34400230 DOI: 10.1016/j.ijbiomac.2021.08.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 10/20/2022]
Abstract
Gut microbial fermentation of soluble dietary fibers promotes general and substrate-specific health benefits. In this study, the fermentation characteristics of two soluble branched-dietary fibers, namely, agavin (a type of agave fructans) and digestion-resistant maltodextrin (RD) were investigated against cellulose, using a simulated colonic fermenter apparatus employing a mixed culture of swine fecal bacteria. After 48 h of complete fermentation period, the microbial composition was different among all groups, where Bifidobacterium spp. and Lactobacillus spp. dominated the agavin treatment, while the members of the families Lachnospiraceae and Prevotellaceae dominated the RD treatment. Agavin treatment exhibited a clearly segregated two-phased prolonged fermentation trend compared to RD treatment as manifested by the fermentation rates. Further, the highest short-chain fatty acids production even at the end of the fermentation cycle, acidic pH, and the negligible concentration of ammonia accumulation demonstrated favorable fermentation attributes of agavin compared to RD. Therefore, agavin might be an effective and desirable substrate for the colonic microbiota than RD with reference to the expressed microbial taxa and fermentation attributes. This study revealed a notable significance of the structural differences of fermentable fibers on the subsequent fermentation characteristics.
Collapse
Affiliation(s)
- Aldrine Kilua
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Samanthi Pelpolage
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Aki Goto
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Yasunori Nakayama
- Healthcare New Business Division, Teijin Limited, 4-3-2 Asahigaoka, Hino, Tokyo 191-8512, Japan
| | - Eiichi Kitazono
- Healthcare New Business Division, Teijin Limited, 4-3-2 Asahigaoka, Hino, Tokyo 191-8512, Japan
| | - Kiyotsuna Toyohara
- Healthcare New Business Division, Teijin Limited, 4-3-2 Asahigaoka, Hino, Tokyo 191-8512, Japan
| | - Ryuji Nagata
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Naoki Fukuma
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan; Research Center for Global Agromedicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Kyu-Ho Han
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan; Research Center for Global Agromedicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| | - Michihiro Fukushima
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
122
|
Chang WC, So J, Lamon-Fava S. Differential and shared effects of eicosapentaenoic acid and docosahexaenoic acid on serum metabolome in subjects with chronic inflammation. Sci Rep 2021; 11:16324. [PMID: 34381108 PMCID: PMC8357808 DOI: 10.1038/s41598-021-95590-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/22/2021] [Indexed: 12/03/2022] Open
Abstract
The omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) affect cell function and metabolism, but the differential effects of EPA and DHA are not known. In a randomized, controlled, double-blind, crossover study, we assessed the effects of 10-week supplementation with EPA-only and DHA-only (3 g/d), relative to a 4-week lead-in phase of high oleic acid sunflower oil (3 g/day, defined as baseline), on fasting serum metabolites in 21 subjects (9 men and 12 post-menopausal women) with chronic inflammation and some characteristics of metabolic syndrome. Relative to baseline, EPA significantly lowered the tricarboxylic acid (TCA) cycle intermediates fumarate and α-ketoglutarate and increased glucuronate, UDP-glucuronate, and non-esterified DHA. DHA significantly lowered the TCA cycle intermediates pyruvate, citrate, isocitrate, fumarate, α-ketoglutarate, and malate, and increased succinate and glucuronate. Pathway analysis showed that both EPA and DHA significantly affected the TCA cycle, the interconversion of pentose and glucuronate, and alanine, and aspartate and glutamate pathways (FDR < 0.05) and that DHA had a significantly greater effect on the TCA cycle than EPA. Our results indicate that EPA and DHA exhibit both common and differential effects on cell metabolism in subjects with chronic inflammation and some key aspects of metabolic syndrome.
Collapse
Affiliation(s)
- Wan-Chi Chang
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center On Aging, Tufts University, 711 Washington Street, Boston, MA, 02111, USA.,Gerard J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, USA
| | - Jisun So
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center On Aging, Tufts University, 711 Washington Street, Boston, MA, 02111, USA.,Gerard J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, USA
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center On Aging, Tufts University, 711 Washington Street, Boston, MA, 02111, USA. .,Gerard J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, USA.
| |
Collapse
|
123
|
Osuna-Prieto FJ, Martinez-Tellez B, Ortiz-Alvarez L, Di X, Jurado-Fasoli L, Xu H, Ceperuelo-Mallafré V, Núñez-Roa C, Kohler I, Segura-Carretero A, García-Lario JV, Gil A, Aguilera CM, Llamas-Elvira JM, Rensen PCN, Vendrell J, Ruiz JR, Fernández-Veledo S. Elevated plasma succinate levels are linked to higher cardiovascular disease risk factors in young adults. Cardiovasc Diabetol 2021; 20:151. [PMID: 34315463 PMCID: PMC8314524 DOI: 10.1186/s12933-021-01333-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Succinate is produced by both host and microbiota, with a key role in the interplay of immunity and metabolism and an emerging role as a biomarker for inflammatory and metabolic disorders in middle-aged adults. The relationship between plasma succinate levels and cardiovascular disease (CVD) risk in young adults is unknown. METHODS Cross-sectional study in 100 (65% women) individuals aged 18-25 years from the ACTIvating Brown Adipose Tissue through Exercise (ACTIBATE) study cohort. CVD risk factors, body composition, dietary intake, basal metabolic rate, and cardiorespiratory fitness were assessed by routine methods. Plasma succinate was measured with an enzyme-based assay. Brown adipose tissue (BAT) was evaluated by positron emission tomography, and circulating oxylipins were assessed by targeted metabolomics. Fecal microbiota composition was analyzed in a sub-sample. RESULTS Individuals with higher succinate levels had higher levels of visceral adipose tissue (VAT) mass (+ 42.5%), triglycerides (+ 63.9%), C-reactive protein (+ 124.2%), diastolic blood pressure (+ 5.5%), and pro-inflammatory omega-6 oxylipins than individuals with lower succinate levels. Succinate levels were also higher in metabolically unhealthy individuals than in healthy overweight/obese peers. Succinate levels were not associated with BAT volume or activity or with fecal microbiota composition and diversity. CONCLUSIONS Plasma succinate levels are linked to a specific pro-inflammatory omega-6 signature pattern and higher VAT levels, and seem to reflect the cardiovascular status of young adults.
Collapse
Affiliation(s)
- Francisco J Osuna-Prieto
- PROFITH (PROmoting FITness and Health Through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
- Department of Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Center (CIDAF), Granada, Spain
| | - Borja Martinez-Tellez
- PROFITH (PROmoting FITness and Health Through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
- Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lourdes Ortiz-Alvarez
- PROFITH (PROmoting FITness and Health Through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology II, "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Xinyu Di
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Lucas Jurado-Fasoli
- PROFITH (PROmoting FITness and Health Through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - Huiwen Xu
- PROFITH (PROmoting FITness and Health Through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology II, "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Victoria Ceperuelo-Mallafré
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d ́Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Rovira i Virgili University, Tarragona, Spain
| | - Catalina Núñez-Roa
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d ́Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Isabelle Kohler
- Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Amsterdam, The Netherlands
- Center for Analytical Sciences Amsterdam, Amsterdam, The Netherlands
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Center (CIDAF), Granada, Spain
| | | | - Angel Gil
- Department of Biochemistry and Molecular Biology II, "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Biohealth Research Institute in Granada (Ibs, GRANADA), Granada, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Concepción M Aguilera
- Department of Biochemistry and Molecular Biology II, "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Biohealth Research Institute in Granada (Ibs, GRANADA), Granada, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Jose M Llamas-Elvira
- Nuclear Medicine Service, Virgen de las Nieves University Hospital, Biohealth Research Institute in Granada (Ibs. GRANADA), Granada, Spain
| | - Patrick C N Rensen
- Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Joan Vendrell
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d ́Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Rovira i Virgili University, Tarragona, Spain
| | - Jonatan R Ruiz
- PROFITH (PROmoting FITness and Health Through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain.
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d ́Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
124
|
Hagihara M, Ariyoshi T, Kuroki Y, Eguchi S, Higashi S, Mori T, Nonogaki T, Iwasaki K, Yamashita M, Asai N, Koizumi Y, Oka K, Takahashi M, Yamagishi Y, Mikamo H. Clostridium butyricum enhances colonization resistance against Clostridioides difficile by metabolic and immune modulation. Sci Rep 2021; 11:15007. [PMID: 34294848 PMCID: PMC8298451 DOI: 10.1038/s41598-021-94572-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/07/2021] [Indexed: 01/21/2023] Open
Abstract
Clostridioides difficile infection (CDI) represents the leading cause of nosocomial diarrhea worldwide and is associated with gut dysbiosis and intestinal damage. Clostridium butyricum MIYAIRI 588 (CBM 588) contributes significantly to reduce epithelial damage. However, the impacts of CBM 588 on antibacterial therapy for CDI are not clear. Here we show that CBM 588 enhanced the antibacterial activity of fidaxomicin against C. difficile and negatively modulated gut succinate levels to prevent C. difficile proliferation and downregulate tumor necrosis factor-α (TNF-α) producing macrophages in the colon lumina propria (cLP), resulting in a significant decrease in colon epithelial damage. Additionally, CBM 588 upregulated T cell-dependent pathogen specific immunoglobulin A (IgA) via interleukin (IL)-17A producing CD4+ cells and plasma B cells in the cLP, and Th17 cells in the cLP enhanced the gut epithelial barrier function. IL-17A and succinic acid modulations with CBM 588 enhance gut colonization resistance to C. difficile and protect the colon tissue from CDI.
Collapse
Affiliation(s)
- Mao Hagihara
- Department of Molecular Epidemiology and Biomedical Sciences, Aichi Medical University, Nagakute, 480-1195, Japan.,Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan
| | - Tadashi Ariyoshi
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan.,Miyarisan Pharmaceutical Co., Ltd., Saitama, 114-0016, Japan
| | - Yasutoshi Kuroki
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan.,Miyarisan Pharmaceutical Co., Ltd., Saitama, 114-0016, Japan
| | - Shuhei Eguchi
- Miyarisan Pharmaceutical Co., Ltd., Saitama, 114-0016, Japan
| | - Seiya Higashi
- Miyarisan Pharmaceutical Co., Ltd., Saitama, 114-0016, Japan
| | - Takeshi Mori
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan
| | - Tsunemasa Nonogaki
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, 463-8521, Japan
| | - Kenta Iwasaki
- Departments of Kidney Disease and Transplant Immunology, Aichi Medical University, Nagakute, 480-1195, Japan
| | - Makoto Yamashita
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan
| | - Nobuhiro Asai
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan
| | - Yusuke Koizumi
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan
| | - Kentaro Oka
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan.,Miyarisan Pharmaceutical Co., Ltd., Saitama, 114-0016, Japan
| | - Motomichi Takahashi
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan.,Miyarisan Pharmaceutical Co., Ltd., Saitama, 114-0016, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, 480-1195, Japan.
| |
Collapse
|
125
|
Wale KR, Cottam C, Connolly JP, Roe AJ. Transcriptional and metabolic regulation of EHEC and Citrobacter rodentium pathogenesis. Curr Opin Microbiol 2021; 63:70-75. [PMID: 34224961 DOI: 10.1016/j.mib.2021.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/04/2021] [Accepted: 06/16/2021] [Indexed: 11/24/2022]
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) is a gastrointestinal pathogen that colonizes the colonic epithelium of humans and ruminants using a Type Three Secretion System (T3SS). This system is indispensable for disease and is regulated in response to a plethora of host and microbiota derived signals. The murine pathogen, Citrobacter rodentium, has become an instrumental tool in studying EHEC infection mechanisms in vivo, given its natural ability to infect mice and reliance on the same colonisation machinery. Here, we provide a review of the most recent advancements in EHEC infection biology, focusing on transcriptional regulation of the T3SS in response to physiologically relevant signals and how colonisation impacts on the metabolic micro-environment of the host niche. We pay particular attention to studies that have employed the C. rodentium model for elucidation of such mechanisms in vivo.
Collapse
Affiliation(s)
- Kabo R Wale
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Curtis Cottam
- Newcastle University Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - James Pr Connolly
- Newcastle University Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Andrew J Roe
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
126
|
Catarino MD, Marçal C, Bonifácio-Lopes T, Campos D, Mateus N, Silva AMS, Pintado MM, Cardoso SM. Impact of Phlorotannin Extracts from Fucus vesiculosus on Human Gut Microbiota. Mar Drugs 2021; 19:375. [PMID: 34209623 PMCID: PMC8306378 DOI: 10.3390/md19070375] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
Recent studies indicate that plant polyphenols could be pointed as potential prebiotic candidates since they may interact with the gut microbiota, stimulating its growth and the production of metabolites. However, little is known about the fate of brown seaweeds' phlorotannins during their passage throughout the gastrointestinal tract. This work aimed to evaluate the stability and bioaccessibility of Fucus vesiculosus phlorotannins after being submitted to a simulated digestive process, as well as their possible modulatory effects on gut microbiota and short-chain fatty acids production following a fermentation procedure using fecal inoculates to mimic the conditions of the large intestine. The stability of phlorotannins throughout the gastrointestinal tract was reduced, with a bioaccessibility index between 2 and 14%. Moreover, slight alterations in the growth of certain commensal bacteria were noticed, with Enterococcus spp. being the most enhanced group. Likewise, F. vesiculosus phlorotannins displayed striking capacity to enhance the levels of propionate and butyrate, which are two important short-chain fatty acids known for their role in intestinal homeostasis. In summary, this work provides valuable information regarding the behavior of F. vesiculosus phlorotannins along the gastrointestinal tract, presenting clear evidence that these compounds can positively contribute to the maintenance of a healthy gastrointestinal condition.
Collapse
Affiliation(s)
- Marcelo D. Catarino
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| | - Catarina Marçal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| | - Teresa Bonifácio-Lopes
- CBQF-Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (D.C.); (M.M.P.)
| | - Débora Campos
- CBQF-Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (D.C.); (M.M.P.)
| | - Nuno Mateus
- REQUIMTE/LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal;
| | - Artur M. S. Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| | - Maria Manuela Pintado
- CBQF-Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (D.C.); (M.M.P.)
| | - Susana M. Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| |
Collapse
|
127
|
Steiner TM, Lettl C, Schindele F, Goebel W, Haas R, Fischer W, Eisenreich W. Substrate usage determines carbon flux via the citrate cycle in Helicobacter pylori. Mol Microbiol 2021; 116:841-860. [PMID: 34164854 DOI: 10.1111/mmi.14775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/07/2021] [Accepted: 06/19/2021] [Indexed: 12/31/2022]
Abstract
Helicobacter pylori displays a worldwide infection rate of about 50%. The Gram-negative bacterium is the main reason for gastric cancer and other severe diseases. Despite considerable knowledge about the metabolic inventory of H. pylori, carbon fluxes through the citrate cycle (TCA cycle) remained enigmatic. In this study, different 13 C-labeled substrates were supplied as carbon sources to H. pylori during microaerophilic growth in a complex medium. After growth, 13 C-excess and 13 C-distribution were determined in multiple metabolites using GC-MS analysis. [U-13 C6 ]Glucose was efficiently converted into glyceraldehyde but only less into TCA cycle-related metabolites. In contrast, [U-13 C5 ]glutamate, [U-13 C4 ]succinate, and [U-13 C4 ]aspartate were incorporated at high levels into intermediates of the TCA cycle. The comparative analysis of the 13 C-distributions indicated an adaptive TCA cycle fully operating in the closed oxidative direction with rapid equilibrium fluxes between oxaloacetate-succinate and α-ketoglutarate-citrate. 13 C-Profiles of the four-carbon intermediates in the TCA cycle, especially of malate, together with the observation of an isocitrate lyase activity by in vitro assays, suggested carbon fluxes via a glyoxylate bypass. In conjunction with the lack of enzymes for anaplerotic CO2 fixation, the glyoxylate bypass could be relevant to fill up the TCA cycle with carbon atoms derived from acetyl-CoA.
Collapse
Affiliation(s)
- Thomas M Steiner
- Bavarian NMR Center-Structural Membrane Biochemistry, Department of Chemistry, Technische Universität München, Garching, Germany
| | - Clara Lettl
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, München, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, München, Germany
| | - Franziska Schindele
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, München, Germany
| | - Werner Goebel
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, München, Germany
| | - Rainer Haas
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, München, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, München, Germany
| | - Wolfgang Fischer
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, München, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, München, Germany
| | - Wolfgang Eisenreich
- Bavarian NMR Center-Structural Membrane Biochemistry, Department of Chemistry, Technische Universität München, Garching, Germany
| |
Collapse
|
128
|
Shinno-Hashimoto H, Hashimoto Y, Wei Y, Chang L, Fujita Y, Ishima T, Matsue H, Hashimoto K. Abnormal composition of microbiota in the gut and skin of imiquimod-treated mice. Sci Rep 2021; 11:11265. [PMID: 34050205 PMCID: PMC8163751 DOI: 10.1038/s41598-021-90480-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023] Open
Abstract
Psoriasis is a chronic, inflammatory skin disease. Although the precise etiology of psoriasis remains unclear, gut-microbiota axis might play a role in the pathogenesis of the disease. Here we investigated whether the composition of microbiota in the intestine and skin is altered in the imiquimod (IMQ)-treated mouse model of psoriasis. Topical application of IMQ to back skin caused significant changes in the composition of microbiota in the intestine and skin of IMQ-treated mice compared to control mice. The LEfSe algorithm identified the species Staphylococcus lentus as potential skin microbial marker for IMQ group. Furthermore, there were correlations for several microbes between the intestine and skin, suggesting a role of skin-gut-microbiota in IMQ-treated mice. Levels of succinic acid and lactic acid in feces from IMQ-treated mice were significantly higher than control mice. Moreover, the predictive functional analysis of the microbiota in the intestine and skin showed that IMQ caused alterations in several KEGG pathways. In conclusion, the current data indicated that topical application with IMQ to skin alters the composition of the microbiota in the gut and skin of host. It is likely that skin-gut microbiota axis plays a role in pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Hiroyo Shinno-Hashimoto
- Department of Dermatology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Yaeko Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
- Department of Respirology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Yan Wei
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Tamaki Ishima
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Hiroyuki Matsue
- Department of Dermatology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
129
|
Morishima S, Aoi W, Kawamura A, Kawase T, Takagi T, Naito Y, Tsukahara T, Inoue R. Intensive, prolonged exercise seemingly causes gut dysbiosis in female endurance runners. J Clin Biochem Nutr 2021. [PMID: 34025029 DOI: 10.3164/jcbn.20.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intensive, prolonged exercise is known to induce gastrointestinal disorders such as diarrhea, with gut dysbiosis suggested as being one of the causatives. In the present study, we wanted to investigate the relationship between intensive exercise and the gut microbiota status. To that end, the microbiota, the moisture content and the bacterial metabolites (e.g., organic acids) of female endurance runners (n = 15) and those of non-athletic but healthy, age-matching female controls (n = 14) were compared. The analysis of the gut microbiota analysis showed that, unlike control subjects, female endurance runners had distinct microbiotas, with some bacteria found in higher abundances likely being involved in gut inflammation. The concentration of succinate, a gut bacterial metabolite regarded as undesirable when accumulated in the lumen, was significantly (p<0.05) higher in the female endurance runners. Faecalibacterium, that was significantly (p<0.05) abundant in female endurance runners, can produce succinate in certain environments and hence may contribute to succinate accumulation, at least partly. The present work suggested that the gut microbiotas of female endurance runners are seemingly dysbiotic when compared with those of control subjects. Further investigation of the mechanism by which intensive, prolonged exercise affects the gut microbiota is recommended.
Collapse
Affiliation(s)
- So Morishima
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan.,Laboratory of Animal Science, Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho, Sakyo-ku, Kyoto 606-8522, Japan
| | - Wataru Aoi
- Laboratory of Nutrition Science, Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho, Sakyo-ku, Kyoto 606-8522, Japan
| | - Aki Kawamura
- Sports Science Research Promotion Center, Nippon Sport Science University, 7-1-1 Fukasawa, Setagaya-ku, Tokyo 158-8508, Japan
| | - Takahiro Kawase
- Kyoto Institute of Nutrition & Pathology, Ujitawara, Kyoto 610-0231, Japan
| | - Tomohisa Takagi
- Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan.,Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan.,Laboratory of Animal Science, Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho, Sakyo-ku, Kyoto 606-8522, Japan
| |
Collapse
|
130
|
Wang J, Li X, Wu X, Wang Z, Wu X, Wang S, Jing G, Yan T. Fecal Microbiota Transplantation as an Effective Treatment for Carbapenem-Resistant Klebsiella pneumoniae Infection in a Renal Transplant Patient. Infect Drug Resist 2021; 14:1805-1811. [PMID: 34017186 PMCID: PMC8131010 DOI: 10.2147/idr.s308308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/29/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND In renal transplant recipients, carbapenem-resistant Klebsiella pneumoniae (CRKP) infection is a common complication, and usually associated with severe clinical outcomes due to a lack of effective treatment. CASE PRESENTATION A 37-year-old woman with CRKP infection one month after kidney transplantation was involved in this study. Microbial characteristics of fecal samples from the patient were analyzed. Fecal microbiota transplantation (FMT) was performed for treating the CRKP infection. One week after FMT, the patient's urine and anal swab cultures returned negative for CRKP, and 17 days after FMT, the incision showed complete healing. Moreover, the patient had no symptoms of infection two months after FMT. Alpha diversity analyses showed that before FMT, the patient was associated with obviously lower species richness and diversity than the donor, which significantly increased at one week, three weeks and two months after FMT. Beta diversity analyses showed that though the patient's microbial community post-FMT still differed from the donor composition, their distances decreased visibly, especially at one week and three weeks after FMT. Obvious shift in microbial composition could be observed before and after FMT. The microbial composition of the patient post FMT resembled the donor composition. Relative abundance of genera such as Phascolarctobacterium and Lachnoclostridium increased after FMT, while the relative abundance of Klebsiella significantly decreased. CONCLUSION This study demonstrated the therapeutic effect of FMT on infections caused by CRKP for a renal transplant patient. Further studies are required to confirm the findings of this study.
Collapse
Affiliation(s)
- Junpeng Wang
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xin Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, People’s Republic of China
| | - Xiaoqiang Wu
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Zhiwei Wang
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Xuan Wu
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Shanmei Wang
- Department of Clinical Microbiology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Gaopeng Jing
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Tianzhong Yan
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| |
Collapse
|
131
|
Biemann R, Buß E, Benndorf D, Lehmann T, Schallert K, Püttker S, Reichl U, Isermann B, Schneider JG, Saake G, Heyer R. Fecal Metaproteomics Reveals Reduced Gut Inflammation and Changed Microbial Metabolism Following Lifestyle-Induced Weight Loss. Biomolecules 2021; 11:biom11050726. [PMID: 34066026 PMCID: PMC8150863 DOI: 10.3390/biom11050726] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Gut microbiota-mediated inflammation promotes obesity-associated low-grade inflammation, which represents a hallmark of metabolic syndrome. To investigate if lifestyle-induced weight loss (WL) may modulate the gut microbiome composition and its interaction with the host on a functional level, we analyzed the fecal metaproteome of 33 individuals with metabolic syndrome in a longitudinal study before and after lifestyle-induced WL in a well-defined cohort. The 6-month WL intervention resulted in reduced BMI (-13.7%), improved insulin sensitivity (HOMA-IR, -46.1%), and reduced levels of circulating hsCRP (-39.9%), indicating metabolic syndrome reversal. The metaprotein spectra revealed a decrease of human proteins associated with gut inflammation. Taxonomic analysis revealed only minor changes in the bacterial composition with an increase of the families Desulfovibrionaceae, Leptospiraceae, Syntrophomonadaceae, Thermotogaceae and Verrucomicrobiaceae. Yet we detected an increased abundance of microbial metaprotein spectra that suggest an enhanced hydrolysis of complex carbohydrates. Hence, lifestyle-induced WL was associated with reduced gut inflammation and functional changes of human and microbial enzymes for carbohydrate hydrolysis while the taxonomic composition of the gut microbiome remained almost stable. The metaproteomics workflow has proven to be a suitable method for monitoring inflammatory changes in the fecal metaproteome.
Collapse
Affiliation(s)
- Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Paul-List-Str. 13/15, 04103 Leipzig, Germany;
- Correspondence: (R.B.); (D.B.); (R.H.)
| | - Enrico Buß
- Bioprocess Engineering, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (E.B.); (T.L.); (K.S.); (S.P.); (U.R.)
| | - Dirk Benndorf
- Bioprocess Engineering, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (E.B.); (T.L.); (K.S.); (S.P.); (U.R.)
- Microbiology, Anhalt University of Applied Sciences, Bernburger Straße 55, 06354 Köthen, Germany
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106 Magdeburg, Germany
- Correspondence: (R.B.); (D.B.); (R.H.)
| | - Theresa Lehmann
- Bioprocess Engineering, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (E.B.); (T.L.); (K.S.); (S.P.); (U.R.)
| | - Kay Schallert
- Bioprocess Engineering, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (E.B.); (T.L.); (K.S.); (S.P.); (U.R.)
| | - Sebastian Püttker
- Bioprocess Engineering, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (E.B.); (T.L.); (K.S.); (S.P.); (U.R.)
| | - Udo Reichl
- Bioprocess Engineering, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (E.B.); (T.L.); (K.S.); (S.P.); (U.R.)
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106 Magdeburg, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Paul-List-Str. 13/15, 04103 Leipzig, Germany;
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Jochen G. Schneider
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, Avenue du Swing, L-4367 Belvaux, Luxembourg;
- Department of Internal Medicine II, Saarland University Medical Center, Kirrberger Str., 66424 Homburg Saar, Germany
| | - Gunter Saake
- Database and Software Engineering Group, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany;
| | - Robert Heyer
- Bioprocess Engineering, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (E.B.); (T.L.); (K.S.); (S.P.); (U.R.)
- Database and Software Engineering Group, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany;
- Correspondence: (R.B.); (D.B.); (R.H.)
| |
Collapse
|
132
|
Galié S, García-Gavilán J, Papandreou C, Camacho-Barcía L, Arcelin P, Palau-Galindo A, Rabassa A, Bulló M. Effects of Mediterranean Diet on plasma metabolites and their relationship with insulin resistance and gut microbiota composition in a crossover randomized clinical trial. Clin Nutr 2021; 40:3798-3806. [PMID: 34130026 DOI: 10.1016/j.clnu.2021.04.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS The Mediterranean Diet (MedDiet) may decrease the cardiometabolic risk through modulation of metabolic pathways. Furthermore, the interplay between MedDiet, metabolites and microbial metabolism may improve our understanding on the metabolic effects of this diet. We aimed to evaluate the effect of the MedDiet compared to nuts supplementation on circulating metabolites and their relationship with cardiometabolic health. We further examined whether changes in the metabolomic profiles were associated with changes in gut microbiota composition in a multi-omics integrative approach. METHODS Forty-four adults with Metabolic Syndrome (MetS), (aged 37-65) participated in a randomized controlled, crossover 2-months dietary-intervention trial with a 1-month wash-out period, consuming a MedDiet or a non MedDiet plus nuts (50 g/day). Nutritional data were collected at the beginning and the end of each intervention period using 3-day dietary records, as well as fasting blood and fecal samples. Plasma metabolites (m = 378) were profiled using targeted metabolomics. Associations of these metabolites with the interventions were assessed with elastic net regression analyses. Gut microbiota composition was assessed by 16S rRNA sequencing. A sparse least regression analysis combined with a canonical correlation analysis was conducted between the plasma selected metabolites and genera in order to identify the relevant dual-omics signatures discriminating the dietary interventions. RESULTS Changes in 65 circulating metabolites were significantly associated with the MedDiet (mainly lipids, acylcarnitines, amino acids, steroids and TCA intermediates). Importantly, these changes were associated with decreases in glucose, insulin and HOMA-IR. The network analysis identified two main clusters of genera with an opposite behaviour towards selected metabolites, mainly PC species, ChoE(20:5), TGs and medium/long-chain acylcarnitines. CONCLUSION Following a MedDiet, rather than consuming nuts in the context of a non-MedDiet was associated with a specific plasma metabolomic profile, which was also related to metabolic improvements in adults with MetS. The identified correlated network between specific bacteria and metabolites suggests interplay between diet, circulating metabolites and gut microbiota. The trial was registered in the ISRCTN with identifier ISRCTN88780852, https://doi.org/10.1186/ISRCTN88780852.
Collapse
Affiliation(s)
- Serena Galié
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University Rovira i Virgili (URV), Reus, Spain; Institute of Health Pere Virgili, IISPV, University Hospital Sant Joan, Reus, Spain; Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús García-Gavilán
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University Rovira i Virgili (URV), Reus, Spain; Institute of Health Pere Virgili, IISPV, University Hospital Sant Joan, Reus, Spain; Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Christopher Papandreou
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University Rovira i Virgili (URV), Reus, Spain; Institute of Health Pere Virgili, IISPV, University Hospital Sant Joan, Reus, Spain; Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Lucía Camacho-Barcía
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University Rovira i Virgili (URV), Reus, Spain; Institute of Health Pere Virgili, IISPV, University Hospital Sant Joan, Reus, Spain; Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Pierre Arcelin
- ABS Reus V. Centre d'Assistència Primària Marià Fortuny, SAGESSA, Reus, Spain
| | | | - Antoni Rabassa
- Institute of Health Pere Virgili, IISPV, University Hospital Sant Joan, Reus, Spain
| | - Mònica Bulló
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University Rovira i Virgili (URV), Reus, Spain; Institute of Health Pere Virgili, IISPV, University Hospital Sant Joan, Reus, Spain; Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
133
|
Therdtatha P, Song Y, Tanaka M, Mariyatun M, Almunifah M, Manurung NEP, Indriarsih S, Lu Y, Nagata K, Fukami K, Ikeda T, Lee YK, Rahayu ES, Nakayama J. Gut Microbiome of Indonesian Adults Associated with Obesity and Type 2 Diabetes: A Cross-Sectional Study in an Asian City, Yogyakarta. Microorganisms 2021; 9:897. [PMID: 33922321 PMCID: PMC8147061 DOI: 10.3390/microorganisms9050897] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Indonesia is a developing country facing the national problem of the growing obesity and diabetes in its population due to recent drastic dietary and lifestyle changes. To understand the link between the gut microbiome, diet, and health of Indonesian people, fecal microbiomes and metabolomes of 75 Indonesian adults in Yogyakarta City, including obese people (n = 21), type 2 diabetes (T2D) patients (n = 25), and the controls (n = 29) were characterized together with their dietary and medical records. Variations of microbiomes showed a triangular distribution in the principal component analysis, driven by three dominant bacterial genera, namely Bacteroides, Prevotella, and Romboutsia. The Romboutsia-driven microbiome, characterized by low bacterial diversity and high primary bile acids, was associated with fat-driven obesity. The Bacteroides-driven microbiome, which counteracted Prevotella but was associated with Ruminococcaceae concomitantly increased with high-carbohydrate diets, showed positive correlation with T2D indices but negative correlation with body mass index. Notably, Bacteroides fragilis was increased in T2D patients with a decrease in fecal conjugated bile acids, particularly tauroursodeoxycholic acid (TUDCA), a farnesoid X receptor (FXR) antagonist with anti-diabetic activity, while these features disappeared in patients administered metformin. These results indicate that the gut microbiome status of Indonesian adults is differently associated with obesity and T2D under their varied dietary habits.
Collapse
Affiliation(s)
- Phatthanaphong Therdtatha
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (P.T.); (Y.S.); (M.T.)
| | - Yayi Song
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (P.T.); (Y.S.); (M.T.)
| | - Masaru Tanaka
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (P.T.); (Y.S.); (M.T.)
| | - Mariyatun Mariyatun
- Faculty of Agricultural Technology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia; (M.M.); (M.A.); (N.E.P.M.); (S.I.); (E.S.R.)
| | - Maisaroh Almunifah
- Faculty of Agricultural Technology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia; (M.M.); (M.A.); (N.E.P.M.); (S.I.); (E.S.R.)
| | - Nancy Eka Putri Manurung
- Faculty of Agricultural Technology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia; (M.M.); (M.A.); (N.E.P.M.); (S.I.); (E.S.R.)
| | - Siska Indriarsih
- Faculty of Agricultural Technology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia; (M.M.); (M.A.); (N.E.P.M.); (S.I.); (E.S.R.)
| | - Yi Lu
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (Y.L.); (K.N.)
| | - Koji Nagata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (Y.L.); (K.N.)
| | - Katsuya Fukami
- Material Management Center of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
| | - Tetsuo Ikeda
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
- Endoscopy and Endoscopic Surgery, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Yuan-Kun Lee
- Department of Microbiology and Immunology, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore;
| | - Endang Sutriswati Rahayu
- Faculty of Agricultural Technology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia; (M.M.); (M.A.); (N.E.P.M.); (S.I.); (E.S.R.)
| | - Jiro Nakayama
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (P.T.); (Y.S.); (M.T.)
| |
Collapse
|
134
|
Zhang X, Cheng K, Ning Z, Mayne J, Walker K, Chi H, Farnsworth CL, Lee K, Figeys D. Exploring the Microbiome-Wide Lysine Acetylation, Succinylation, and Propionylation in Human Gut Microbiota. Anal Chem 2021; 93:6594-6598. [PMID: 33885279 DOI: 10.1021/acs.analchem.1c00962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lysine acylations are important post-translational modifications that are present in both eukaryotes and prokaryotes and regulate diverse cellular functions. Our knowledge of the microbiome lysine acylation remains limited due to the lack of efficient analytical and bioinformatics methods for complex microbial communities. Here, we show that the serial enrichment using motif antibodies successfully captures peptides containing lysine acetylation, propionylation, and succinylation from human gut microbiome samples. A new bioinformatic workflow consisting of an unrestricted database search confidently identified >60,000 acetylated, and ∼20,000 propionylated and succinylated gut microbial peptides. The characterization of these identified modification-specific metaproteomes, i.e., meta-PTMomes, demonstrates that lysine acylations are differentially distributed in microbial species with different metabolic capabilities. This study provides an analytical framework for the study of lysine acylations in the microbiome, which enables functional microbiome studies at the post-translational level.
Collapse
Affiliation(s)
- Xu Zhang
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Kai Cheng
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Zhibin Ning
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Janice Mayne
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Krystal Walker
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Hao Chi
- Institute of Computing Technology, Chinese Academy of Sciences, No. 6 Zhongguancun South Road, Beijing 100190, China
| | | | - Kimberly Lee
- Cell Signaling Technology Inc., Danvers, Massachusetts 01923, United States
| | - Daniel Figeys
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
135
|
Fernandez-Julia PJ, Munoz-Munoz J, van Sinderen D. A comprehensive review on the impact of β-glucan metabolism by Bacteroides and Bifidobacterium species as members of the gut microbiota. Int J Biol Macromol 2021; 181:877-889. [PMID: 33864864 DOI: 10.1016/j.ijbiomac.2021.04.069] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/01/2021] [Accepted: 04/10/2021] [Indexed: 12/16/2022]
Abstract
β-glucans are polysaccharides which can be obtained from different sources, and which have been described as potential prebiotics. The beneficial effects associated with β-glucan intake are that they reduce energy intake, lower cholesterol levels and support the immune system. Nevertheless, the mechanism(s) of action underpinning these health effects related to β-glucans are still unclear, and the precise impact of β-glucans on the gut microbiota has been subject to debate and revision. In this review, we summarize the most recent advances involving structurally different types of β-glucans as fermentable substrates for Bacteroidetes (mainly Bacteroides) and Bifidobacterium species as glycan degraders. Bacteroides is one of the most abundant bacterial components of the human gut microbiota, while bifidobacteria are widely employed as a probiotic ingredient. Both are generalist glycan degraders capable of using a wide range of substrates: Bacteroides spp. are specialized as primary degraders in the metabolism of complex carbohydrates, whereas Bifidobacterium spp. more commonly metabolize smaller glycans, in particular oligosaccharides, sometimes through syntrophic interactions with Bacteroides spp., in which they act as secondary degraders.
Collapse
Affiliation(s)
- Pedro J Fernandez-Julia
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne NE1 8ST, Tyne & Wear, England, United Kingdom
| | - Jose Munoz-Munoz
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne NE1 8ST, Tyne & Wear, England, United Kingdom.
| | - Douwe van Sinderen
- School of Microbiology & APC Microbiome Ireland, University College Cork, Ireland University College Cork, Cork, Ireland.
| |
Collapse
|
136
|
Butts CA, Paturi G, Hedderley DI, Martell S, Dinnan H, Stoklosinski H, Carpenter EA. Goat and cow milk differ in altering microbiota composition and fermentation products in rats with gut dysbiosis induced by amoxicillin. Food Funct 2021; 12:3104-3119. [PMID: 33725036 DOI: 10.1039/d0fo02950e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Antibiotics are effective treatments for bacterial infections, however, their oral administration can have unintended consequences and may alter the gut microbiota composition. In this study, we examined the influence of antibiotics on the induction of gut dysbiosis and then evaluated the potential of cow and goat milk to restore the microbiota composition and metabolism in newly weaned rats. In the first study (gut dysbiosis model), rats were treated with amoxicillin, a mixture of antibiotics (ampicillin, gentamicin and metronidazole) or no antibiotics (control). Antibiotics reduced the rat body weights, food intakes and faecal outputs compared to the control group. Gut length was significantly decreased after the antibiotic intake. The bacterial populations (Bifidobacterium spp., Lactobacillus spp. and total bacteria) and short-chain fatty acids (SCFAs; acetic, butyric and propionic) concentrations in rat caecum, colon and faeces were significantly altered by the antibiotic treatments. In the second study, we examined the effects of cow and goat milk in restoring bacterial populations and metabolism in rats with gut dysbiosis induced by amoxicillin. Goat milk significantly increased the numbers of Bifidobacterium spp. and Lactobacillus spp. and decreased the numbers of Clostridium perfringens in the caecum and colon of rats treated with amoxicillin. Whereas, rats fed cow milk had higher Lactobacillus spp. and lower C. perfringens in the gut. Caecal and colonic SCFAs (acetic, butyric and propionic) concentrations differed significantly between rats fed cow and goat milk diets. Overall, goat and cow milk varied in their effects on the immature gut following antibiotic-induced dysbiosis in a rat model.
Collapse
Affiliation(s)
- Christine A Butts
- The New Zealand Institute for Plant and Food Research Limited, Private Bag 11600, Palmerston North 4442, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
137
|
Rombouts C, Van Meulebroek L, De Spiegeleer M, Goethals S, Van Hecke T, De Smet S, De Vos WH, Vanhaecke L. Untargeted Metabolomics Reveals Elevated L-Carnitine Metabolism in Pig and Rat Colon Tissue Following Red Versus White Meat Intake. Mol Nutr Food Res 2021; 65:e2000463. [PMID: 33550692 DOI: 10.1002/mnfr.202000463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/29/2020] [Indexed: 12/12/2022]
Abstract
SCOPE The consumption of red and processed meat, and not white meat, associates with the development of various Western diseases such as colorectal cancer and type 2 diabetes. This work aims at unraveling novel meat-associated mechanisms that are involved in disease development. METHODS AND RESULTS A non-hypothesis driven strategy of untargeted metabolomics is applied to assess colon tissue from rats (fed a high dose of beef vs. white meat) and from pigs (fed red/processed meat vs. white meat), receiving a realistic human background diet. An increased carnitine metabolism is observed, which is reflected by higher levels of acylcarnitines and 3-dehydroxycarnitine (rats and pigs) and trimethylamine-N-oxide (rats). While 3-dehydroxycarnitine is higher in HT29 cells, incubated with colonic beef digests, acylcarnitine levels are reduced. This suggests an altered response from colon cancer cell line towards meat-induced oxidative stress. Moreover, metabolic differences between rat and pigs are observed in N-glycolylneuraminic acid incorporation, prostaglandin, and fatty acid synthesis. CONCLUSION This study demonstrates elevated (acyl)carnitine metabolism in colon tissue of animals that follow a red meat-based diet, providing mechanistic insights that may aid in explaining the nutritional-physiological correlation between red/processed meat and Western diseases.
Collapse
Affiliation(s)
- Caroline Rombouts
- Department of Veterinary Public Health and Food Safety, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Lieven Van Meulebroek
- Department of Veterinary Public Health and Food Safety, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Margot De Spiegeleer
- Department of Veterinary Public Health and Food Safety, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Sophie Goethals
- Department of Veterinary Public Health and Food Safety, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Nutrition and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Thomas Van Hecke
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Nutrition and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Stefaan De Smet
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Nutrition and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Winnok H De Vos
- Department of Molecular Biotechnology, Cell Systems & Imaging, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
- Department of Veterinary Sciences, Laboratory of Cell Biology and Histology, Faculty of Veterinary Medicine, University of Antwerp, Campus Drie Eiken Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Lynn Vanhaecke
- Department of Veterinary Public Health and Food Safety, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
- School of Biological Sciences, Queen's University Belfast, Lisburn Road 97, Belfast, UK
| |
Collapse
|
138
|
Terra X, Ceperuelo-Mallafré V, Merma C, Benaiges E, Bosch R, Castillo P, Flores JC, León X, Valduvieco I, Basté N, Cámara M, Lejeune M, Gumà J, Vendrell J, Vilaseca I, Fernández-Veledo S, Avilés-Jurado FX. Succinate Pathway in Head and Neck Squamous Cell Carcinoma: Potential as a Diagnostic and Prognostic Marker. Cancers (Basel) 2021; 13:cancers13071653. [PMID: 33916314 PMCID: PMC8037494 DOI: 10.3390/cancers13071653] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Emerging evidence points to succinate as an important oncometabolite in cancer development; however, the contribution of the succinate-SUCNR1 axis to cancer progression remains unclear. Head and neck squamous cell carcinoma (HNSCC) is associated with disease and treatment-related morbidity so there is an urgent need for innovation in treatment and diagnosis practices. Our aim was to evaluate the potential of the succinate-related pathway as a diagnostic and prognostic biomarker in HNSCC. The circulating succinate levels are increased in HNSCC, being a potential noninvasive biomarker for HNSCC diagnosis. Moreover, the succinate receptor (SUCNR1) and genes related to succinate metabolism, which are predominantly expressed in the tumoral mucosa as compared with healthy tissue, are positively associated with plasma succinate. Remarkably, we found that SUCNR1 and SDHA expression levels predict prognosis. Abstract Head and neck squamous cell carcinoma (HNSCC) is characterized by high rates of mortality and treatment-related morbidity, underscoring the urgent need for innovative and safe treatment strategies and diagnosis practices. Mitochondrial dysfunction is a hallmark of cancer and can lead to the accumulation of tricarboxylic acid cycle intermediates, such as succinate, which function as oncometabolites. In addition to its role in cancer development through epigenetic events, succinate is an extracellular signal transducer that modulates immune response, angiogenesis and cell invasion by activating its cognate receptor SUCNR1. Here, we explored the potential value of the circulating succinate and related genes in HNSCC diagnosis and prognosis. We determined the succinate levels in the serum of 66 pathologically confirmed, untreated patients with HNSCC and 20 healthy controls. We also surveyed the expression of the genes related to succinate metabolism and signaling in tumoral and nontumoral adjacent tissue and in normal mucosa from 50 patients. Finally, we performed immunohistochemical analysis of SUCNR1 in mucosal samples. The results showed that the circulating levels of succinate were higher in patients with HNSCC than in the healthy controls. Additionally, the expression of SUCNR1, HIF-1α, succinate dehydrogenase (SDH) A, and SDHB was higher in the tumor tissue than in the matched normal mucosa. Consistent with this, immunohistochemical analysis revealed an increase in SUCNR1 protein expression in tumoral and nontumoral adjacent tissue. High SUCNR1 and SDHA expression levels were associated with poor locoregional control, and the locoregional recurrence-free survival rate was significantly lower in patients with high SUCNR1 and SDHA expression than in their peers with lower levels (77.1% [95% CI: 48.9–100.0] vs. 16.7% [95% CI: 0.0–44.4], p = 0.018). Thus, the circulating succinate levels are elevated in HNSCC and high SUCNR1/SDHA expression predicts poor locoregional disease-free survival, identifying this oncometabolite as a potentially valuable noninvasive biomarker for HNSCC diagnosis and prognosis.
Collapse
Affiliation(s)
- Ximena Terra
- MoBioFood Research Group, Biochemistry and Biotechnology Department, Universitat Rovira i Virgili, Campus Sescel·lades, 43007 Tarragona, Spain;
| | - Victoria Ceperuelo-Mallafré
- Department of Endocrinology and Nutrition, Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; (V.C.-M.); (E.B.); (J.V.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - Carla Merma
- Otorhinolaryngology Head-Neck Surgery Department, Hospital Universitari de Tarragona Joan XXIII, Insitut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43005 Tarragona, Spain; (C.M.); (J.C.F.)
| | - Ester Benaiges
- Department of Endocrinology and Nutrition, Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; (V.C.-M.); (E.B.); (J.V.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
- School of Medicine, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Ramon Bosch
- Pathology Department, Plataforma de Estudios Histológicos, Citológicos y de Digitalización, Hospital de Tortosa Verge de la Cinta, Institut d’Investigació Sanitària Pere Virgili (IISPV), URV, 43500 Tortosa, Spain; (R.B.); (M.L.)
| | - Paola Castillo
- Pathology Department, Hospital Clínic de Barcelona, IDIBAPS, 08036 Barcelona, Spain;
| | - Joan Carles Flores
- Otorhinolaryngology Head-Neck Surgery Department, Hospital Universitari de Tarragona Joan XXIII, Insitut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43005 Tarragona, Spain; (C.M.); (J.C.F.)
| | - Xavier León
- Otorhinolaryngology Head-Neck Surgery Department, Hospital de la Santa Creu i Sant Pau and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN, MICINN, ISCIII), Universitat Autònoma de Barcelona, 08041 Barcelona, Spain;
| | - Izaskun Valduvieco
- Radiation Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
| | - Neus Basté
- Oncology Department, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
| | - Marina Cámara
- Maxillofacial Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
| | - Marylène Lejeune
- Pathology Department, Plataforma de Estudios Histológicos, Citológicos y de Digitalización, Hospital de Tortosa Verge de la Cinta, Institut d’Investigació Sanitària Pere Virgili (IISPV), URV, 43500 Tortosa, Spain; (R.B.); (M.L.)
| | - Josep Gumà
- Oncology Department, Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Sant Joan de Reus, 43204 Reus, Spain;
| | - Joan Vendrell
- Department of Endocrinology and Nutrition, Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; (V.C.-M.); (E.B.); (J.V.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
- School of Medicine, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Isabel Vilaseca
- Otorhinolaryngology Department, UB, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
- Head Neck Clínic, Agència de Gestió d’Ajuts Universitaris i de Recerca (AGAUR), 2017-SGR-01581 Barcelona, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition, Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, 43005 Tarragona, Spain; (V.C.-M.); (E.B.); (J.V.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
- Correspondence: (S.F.-V.); (F.X.A.-J.)
| | - Francesc Xavier Avilés-Jurado
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
- Otorhinolaryngology Department, UB, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
- Head Neck Clínic, Agència de Gestió d’Ajuts Universitaris i de Recerca (AGAUR), 2017-SGR-01581 Barcelona, Spain
- Correspondence: (S.F.-V.); (F.X.A.-J.)
| |
Collapse
|
139
|
Gatea F, Sârbu I, Vamanu E. In Vitro Modulatory Effect of Stevioside, as a Partial Sugar Replacer in Sweeteners, on Human Child Microbiota. Microorganisms 2021; 9:590. [PMID: 33805627 PMCID: PMC8000329 DOI: 10.3390/microorganisms9030590] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
The effect of stevioside on human health is still insufficiently highlighted by recent research. The total or partial replacement of sugar with sweeteners influences the general state of health, especially the human microbiota's response as a determining factor in the onset of type 2 diabetes. The present study aimed to present the long-term (one-year) in vitro effect that regular stevioside consumption had on children's pattern microbiota. A metabolomic response was established by determining the synthesis of organic acids and a correlation with antioxidant status. An increase in the number of bacterial strains and the variation of amount of butyrate and propionate to the detriment of lactic acid was observed. The effect was evidenced by the progressive pH increasing, the reduction of acetic acid, and the proliferation of Escherichia coli strains during the simulations. Synthesis of the main short-chain fatty acids (SCFAs) was interpreted as a response (adaptation) of the microbiota to the stevioside, without a corresponding increase in antioxidant status. This study demonstrated the modulatory role of stevioside on the human microbiota and on the fermentation processes that determine the essential SCFA synthesis in maintaining homeostasis. The protection of the microbiota against oxidative stress was also an essential aspect of reducing microbial diversity.
Collapse
Affiliation(s)
- Florentina Gatea
- Centre of Bioanalysis, National Institute for Biological Sciences, 296 Spl. Independentei, 060031 Bucharest, Romania;
| | - Ionela Sârbu
- Department of Genetics, University of Bucharest, 36-46 Bd. M. Kogalniceanu, 5th District, 050107 Bucharest, Romania;
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agronomic Science and Veterinary Medicine, 59 Marasti blvd, 1 District, 011464 Bucharest, Romania
| |
Collapse
|
140
|
Zimmermann J, Kaleta C, Waschina S. gapseq: informed prediction of bacterial metabolic pathways and reconstruction of accurate metabolic models. Genome Biol 2021; 22:81. [PMID: 33691770 PMCID: PMC7949252 DOI: 10.1186/s13059-021-02295-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/10/2021] [Indexed: 12/21/2022] Open
Abstract
Genome-scale metabolic models of microorganisms are powerful frameworks to predict phenotypes from an organism's genotype. While manual reconstructions are laborious, automated reconstructions often fail to recapitulate known metabolic processes. Here we present gapseq ( https://github.com/jotech/gapseq ), a new tool to predict metabolic pathways and automatically reconstruct microbial metabolic models using a curated reaction database and a novel gap-filling algorithm. On the basis of scientific literature and experimental data for 14,931 bacterial phenotypes, we demonstrate that gapseq outperforms state-of-the-art tools in predicting enzyme activity, carbon source utilisation, fermentation products, and metabolic interactions within microbial communities.
Collapse
Affiliation(s)
- Johannes Zimmermann
- Christian-Albrechts-University Kiel, Institute of Experimental Medicine, Research Group Medical Systems Biology, Michaelis-Str. 5, Kiel, 24105 Germany
| | - Christoph Kaleta
- Christian-Albrechts-University Kiel, Institute of Experimental Medicine, Research Group Medical Systems Biology, Michaelis-Str. 5, Kiel, 24105 Germany
| | - Silvio Waschina
- Christian-Albrechts-University Kiel, Institute of Experimental Medicine, Research Group Medical Systems Biology, Michaelis-Str. 5, Kiel, 24105 Germany
- Christian-Albrechts-University Kiel, Institute of Human Nutrition and Food Science, Nutriinformatics, Heinrich-Hecht-Platz 10, Kiel, 24118 Germany
| |
Collapse
|
141
|
Marzano M, Fosso B, Piancone E, Defazio G, Pesole G, De Robertis M. Stem Cell Impairment at the Host-Microbiota Interface in Colorectal Cancer. Cancers (Basel) 2021; 13:996. [PMID: 33673612 PMCID: PMC7957811 DOI: 10.3390/cancers13050996] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) initiation is believed to result from the conversion of normal intestinal stem cells (ISCs) into cancer stem cells (CSCs), also known as tumor-initiating cells (TICs). Hence, CRC evolves through the multiple acquisition of well-established genetic and epigenetic alterations with an adenoma-carcinoma sequence progression. Unlike other stem cells elsewhere in the body, ISCs cohabit with the intestinal microbiota, which consists of a diverse community of microorganisms, including bacteria, fungi, and viruses. The gut microbiota communicates closely with ISCs and mounting evidence suggests that there is significant crosstalk between host and microbiota at the ISC niche level. Metagenomic analyses have demonstrated that the host-microbiota mutually beneficial symbiosis existing under physiologic conditions is lost during a state of pathological microbial imbalance due to the alteration of microbiota composition (dysbiosis) and/or the genetic susceptibility of the host. The complex interaction between CRC and microbiota is at the forefront of the current CRC research, and there is growing attention on a possible role of the gut microbiome in the pathogenesis of CRC through ISC niche impairment. Here we primarily review the most recent findings on the molecular mechanism underlying the complex interplay between gut microbiota and ISCs, revealing a possible key role of microbiota in the aberrant reprogramming of CSCs in the initiation of CRC. We also discuss recent advances in OMICS approaches and single-cell analyses to explore the relationship between gut microbiota and ISC/CSC niche biology leading to a desirable implementation of the current precision medicine approaches.
Collapse
Affiliation(s)
- Marinella Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy; (M.M.); (B.F.); (G.P.)
| | - Bruno Fosso
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy; (M.M.); (B.F.); (G.P.)
| | - Elisabetta Piancone
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| | - Giuseppe Defazio
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| | - Graziano Pesole
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy; (M.M.); (B.F.); (G.P.)
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| | - Mariangela De Robertis
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| |
Collapse
|
142
|
Bierlein M, Hedgespeth BA, Azcarate-Peril MA, Stauffer SH, Gookin JL. Dysbiosis of fecal microbiota in cats with naturally occurring and experimentally induced Tritrichomonas foetus infection. PLoS One 2021; 16:e0246957. [PMID: 33606740 PMCID: PMC7894905 DOI: 10.1371/journal.pone.0246957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
The protozoal pathogen Tritrichomonas foetus infects the colon of domestic cats and is a major cause of chronic colitis and diarrhea. Treatment failure is common, but antibiotics may improve clinical signs in a subset of cats, leading researchers to question involvement of the colonic microbiota in disease pathogenesis. Studies performed in women with venereal Trichomonas vaginalis infections have revealed that dysbiosis of host microbiota contributes to pathogenicity with similar findings also found in mice with intestinal Tritrichomonas musculis The aim of this study was to characterize differences in the fecal microbiota of cats with and without naturally occurring T. foetus infection and in a group of kittens prior to and after experimentally induced infection. Archived fecal DNA from cats undergoing testing for T. foetus infection (n = 89) and experimentally infected kittens (n = 4; at pre-, 2 weeks, and 9 weeks post-infection) were analyzed by sequencing of 16S rRNA genes. Amongst the naturally infected population, the genera Megamonas and Helicobacter were significantly increased in prevalence and abundance in cats testing positive for T. foetus infection. In the group of four experimentally infected kittens, fecal samples post-infection had significantly lower abundance of genus Dialister and Megamonas and greater abundance of the class Betaproteobacteria and family Succinivibrionaceae. We hypothesize that T. foetus promotes dysbiosis by competition for fermentable substrates used by these bacteria and that metabolic byproducts may contribute to the pathogenesis of colonic inflammation and diarrhea. Future studies are warranted for the measurement of fecal concentrations of microbial and protozoal metabolites in cats with T. foetus infection for the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Metzere Bierlein
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Barry A. Hedgespeth
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - M. Andrea Azcarate-Peril
- Division of Gastroenterology and Hepatology, Department of Medicine, UNC Microbiome Core, Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Stephen H. Stauffer
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jody L. Gookin
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| |
Collapse
|
143
|
Citraro R, Lembo F, De Caro C, Tallarico M, Coretti L, Iannone LF, Leo A, Palumbo D, Cuomo M, Buommino E, Nesci V, Marascio N, Iannone M, Quirino A, Russo R, Calignano A, Constanti A, Russo E, De Sarro G. First evidence of altered microbiota and intestinal damage and their link to absence epilepsy in a genetic animal model, the WAG/Rij rat. Epilepsia 2021; 62:529-541. [PMID: 33428780 DOI: 10.1111/epi.16813] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE A large number of studies have highlighted the important role of the gut microbiota in the pathophysiology of neurological disorders, suggesting that its manipulation might serve as a treatment strategy. We hypothesized that the gut microbiota participates in absence seizure development and maintenance in the WAG/Rij rat model and tested this hypothesis by evaluating potential gut microbiota and intestinal alterations in the model, as well as measuring the impact of microbiota manipulation using fecal microbiota transplantation (FMT). METHODS Initially, gut microbiota composition and intestinal histology of WAG/Rij rats (a well-recognized genetic model of absence epilepsy) were studied at 1, 4, and 8 months of age in comparison to nonepileptic Wistar rats. Subsequently, in a second set of experiments, at 6 months of age, untreated Wistar or WAG/Rij rats treated with ethosuximide (ETH) were used as gut microbiota donors for FMT in WAG/Rij rats, and electroencephalographic (EEG) recordings were obtained over 4 weeks. At the end of FMT, stool and gut samples were collected, absence seizures were measured on EEG recordings, and microbiota analysis and histopathological examinations were performed. RESULTS Gut microbiota analysis showed differences in beta diversity and specific phylotypes at all ages considered and significant variances in the Bacteroidetes/Firmicutes ratio between Wistar and WAG/Rij rats. FMT, from both Wistar and ETH-treated WAG/Rij donors to WAG/Rij rats, significantly decreased the number and duration of seizures. Histological results indicated that WAG/Rij rats were characterized by intestinal villi disruption and inflammatory infiltrates already at 1 month of age, before seizure occurrence; FMT partially restored intestinal morphology while also significantly modifying gut microbiota and concomitantly reducing absence seizures. SIGNIFICANCE Our results demonstrate for the first time that the gut microbiota is modified and contributes to seizure occurrence in a genetic animal model of absence epilepsy and that its manipulation may be a suitable therapeutic target for absence seizure management.
Collapse
Affiliation(s)
- Rita Citraro
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Francesca Lembo
- Department of Pharmacy, Federico II University of Naples, Naples, Italy.,Task Force on Microbiota Studies, Federico II University of Naples, Naples, Italy
| | - Carmen De Caro
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Martina Tallarico
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Lorena Coretti
- Department of Pharmacy, Federico II University of Naples, Naples, Italy.,Task Force on Microbiota Studies, Federico II University of Naples, Naples, Italy
| | - Luigi Francesco Iannone
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Antonio Leo
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Domenico Palumbo
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Mariella Cuomo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | | | - Valentina Nesci
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Nadia Marascio
- Division of Microbiology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Michelangelo Iannone
- National Council of Research, Institute of Neurological Science, Catanzaro, Italy
| | - Angela Quirino
- Division of Microbiology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Roberto Russo
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Andrew Constanti
- Department of Pharmacology, University College London School of Pharmacy, London, UK
| | - Emilio Russo
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giovambattista De Sarro
- System and Applied Pharmacology@University Magna Grecia (FAS@UMG) Research Center, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
144
|
Copeland JK, Chao G, Vanderhout S, Acton E, Wang PW, Benchimol EI, El-Sohemy A, Croitoru K, Gommerman JL, Guttman DS. The Impact of Migration on the Gut Metagenome of South Asian Canadians. Gut Microbes 2021; 13:1-29. [PMID: 33794735 PMCID: PMC8023248 DOI: 10.1080/19490976.2021.1902705] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023] Open
Abstract
South Asian (SA) Canadian immigrants have a higher risk of developing certain immune-mediated inflammatory diseases compared to non-migrant SAs. We sought to investigate the effect of migration on the gut metagenome and to identify microbiological associations between migration and conditions that may influence the development of immune-mediated inflammatory diseases. Metagenomic analysis of 58 first-generation (GEN1) SA immigrants and 38 unrelated Canadian born children-of-immigrants (GEN2) determined that the time lived in Canada was associated with continued changes in gut microbial communities. Migration of GEN1 to Canada early in life results in a gut community with similarities to GEN2 SA Canadians and non-SA North Americans. Conversely, GEN1 immigrants who arrived recently to Canada exhibited pronounced differences from GEN2, while displaying microbial similarities to a non-migrating SA cohort. Multivariate analysis identified that community composition was primarily influenced by high abundance taxa. Prevotella copri dominated in GEN1 and non-migrant SAs. Clostridia and functionally related Bacteroidia spp. replaced P. copri dominance over generations in Canada. Mutually exclusive Dialister species occurred at differing relative abundances over time and generations in Canada. This shift in species composition is accompanied by a change in genes associated with carbohydrate utilization and short-chain fatty acid production. Total energy derived from carbohydrates compared to protein consumption was significantly higher for GEN1 recent immigrants, which may influence the functional requirements of the gut community. This study demonstrates the associations between migration and the gut microbiome, which may be further associated with the altered risk of immune-mediated inflammatory diseases observed for SA Canadians.
Collapse
Affiliation(s)
- Julia K. Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
| | - Gary Chao
- Department of Immunology, University of Toronto, Toronto, CA, Canada
| | - Shelley Vanderhout
- Nutrigenomix, Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
| | - Erica Acton
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
| | - Pauline W. Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, CA, Canada
| | - Eric I. Benchimol
- Department of Pediatrics, and School of Epidemiology and Public Health, University of Ottawa, Ottawa, CA, Canada
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
| | - Ken Croitoru
- Department of Medicine, University of Toronto and Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, CA, Canada
| | | | - David S. Guttman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, CA, Canada
| | - the GEMINI Research Team
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, CA, Canada
- Department of Immunology, University of Toronto, Toronto, CA, Canada
- Nutrigenomix, Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, CA, Canada
- Department of Pediatrics, and School of Epidemiology and Public Health, University of Ottawa, Ottawa, CA, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, CA, Canada
- Department of Medicine, University of Toronto and Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, CA, Canada
| |
Collapse
|
145
|
Effects of rearing system and narasin on growth performance, gastrointestinal development, and gut microbiota of broilers. Poult Sci 2020; 100:100840. [PMID: 33531152 PMCID: PMC7936129 DOI: 10.1016/j.psj.2020.10.073] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 01/02/2023] Open
Abstract
This study was conducted to evaluate the effects of 3 rearing systems (FL: flooring litter rearing, MC: multilayer cage rearing, PN: plastic net rearing) with or without supplemental narasin on growth performance, gastrointestine development and health of broilers. A total of 2,400 one-day-old Ross 308 mixed-sex broilers (1:1 ratio of males and females) were used in a completely randomized design utilizing a 3 × 2 factorial arrangement of treatments, with 12 replicates per treatment. Each replicate for FL, MC, and PN consisted of 34 birds per floor pen, 30 birds per cage, and 36 birds per net pen, respectively, ensuring the same stocking density (12 birds/m2) across the 3 systems. Results showed that lower ADG (average daily gain), ADFI (average daily feed intake), and FCR (feed conversation ratio) observed in the MC group than those of the other 2 systems from 1 to 36 d of age (P < 0.05). Narasin inclusion in the diets decreased ADFI and FCR significantly (P < 0.05). Multilayer cage and PN rearing systems reduced the relative weight of the gizzard significantly (P < 0.05). Compared with FL, MC reduced the relative weight of the duodenum, jejunum, and ileum (P < 0.05). The mRNA expression levels of the ileal IL-1β and IFN-γ in FL were higher than those in PN and MC (P < 0.05). Narasin decreased the ileal mRNA expression of TNF-α (P < 0.05). Different rearing systems changed the ileal microflora structure of broilers. The FL system increased the ileal microbial diversity of broilers and the relative abundance of Actinobacteria. Narasin combined with MC increased the relative abundance of Proteobacteria. In conclusion, birds reared in PN had a higher body weight. The MC birds had poorer intestinal development and health condition, higher abundance of Proteobacteria, but better FCR. The FL rearing appeared to be propitious for gastrointestinal development and health. Narasin inclusion in the diets improved FCR and changed the relative abundance Proteobacteria of broilers.
Collapse
|
146
|
Rodriguez J, Olivares M, Delzenne NM. Implication of the Gut Microbiota in Metabolic Inflammation Associated with Nutritional Disorders and Obesity. Mol Nutr Food Res 2020; 65:e1900481. [PMID: 33111450 DOI: 10.1002/mnfr.201900481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/22/2020] [Indexed: 12/12/2022]
Abstract
SCOPE More than a decade ago, the concept of "metabolic endotoxemia" is elaborated on the fact that some bacterial components, classified as microbial associated membrane pathogens (MAMPs) can pass through the gut barrier and create a systemic low tone inflammation. METHODS AND RESULTS The translocation of lipopolysaccharides and its contribution to systemic inflammation are largely studied in murine models of obesity, allowing to unravel the molecular pathways involved in the process. Many different pathological contexts evoke the loss of gut barrier as an event contributing to inflammation and thereby driving metabolic and behavioral alterations. CONCLUSION This review describes the role of nutrition as a modulator of metabolic regulation and focuses on the contribution of the gut microbiota in the process of the production of a large diversity of bioactive metabolites. The two first sections of the review will be dedicated to the impact of nutritional disorders on both the gut microbiota composition and on metabolic inflammation. The last and more prominent section will describe the role of different nutrient-derived gut metabolites on the gut barrier integrity, metabolic inflammation, and peripheral tissue alterations during obesity or associated complications.
Collapse
Affiliation(s)
- Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Marta Olivares
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, 1200, Belgium
| |
Collapse
|
147
|
Nunes S, Viana SD, Preguiça I, Alves A, Fernandes R, Teodoro JS, Figueirinha A, Salgueiro L, Silva S, Jarak I, Carvalho RA, Cavadas C, Rolo AP, Palmeira CM, Pintado MM, Reis F. Blueberry Consumption Challenges Hepatic Mitochondrial Bioenergetics and Elicits Transcriptomics Reprogramming in Healthy Wistar Rats. Pharmaceutics 2020; 12:pharmaceutics12111094. [PMID: 33202669 PMCID: PMC7697217 DOI: 10.3390/pharmaceutics12111094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022] Open
Abstract
An emergent trend of blueberries’ (BB) “prophylactic” consumption, due to their phytochemicals’ richness and well-known health-promoting claims, is widely scaled-up. However, the benefits arising from BB indiscriminate intake remains puzzling based on incongruent preclinical and human data. To provide a more in-depth elucidation and support towards a healthier and safer consumption, we conducted a translation-minded experimental study in healthy Wistar rats that consumed BB in a juice form (25 g/kg body weight (BW)/day; 14 weeks’ protocol). Particular attention was paid to the physiological adaptations succeeding in the gut and liver tissues regarding the acknowledged BB-induced metabolic benefits. Systemically, BB boosted serum antioxidant activity and repressed the circulating levels of 3-hydroxybutyrate (3-HB) ketone bodies and 3-HB/acetoacetate ratio. Moreover, BB elicited increased fecal succinic acid levels without major changes on gut microbiota (GM) composition and gut ultra-structural organization. Remarkably, an accentuated hepatic mitochondrial bioenergetic challenge, ensuing metabolic transcriptomic reprogramming along with a concerted anti-inflammatory pre-conditioning, was clearly detected upon long-term consumption of BB phytochemicals. Altogether, the results disclosed herein portray a quiescent mitochondrial-related metabolomics and hint for a unified adaptive response to this nutritional challenge. The beneficial or noxious consequences arising from this dietary trend should be carefully interpreted and necessarily claims future research.
Collapse
Affiliation(s)
- Sara Nunes
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Sofia D. Viana
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Polytechnic Institute of Coimbra, ESTESC-Coimbra Health School, Pharmacy/Biomedical Laboratory Sciences, 3046-854 Coimbra, Portugal
| | - Inês Preguiça
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - André Alves
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Rosa Fernandes
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - João S. Teodoro
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (L.S.)
- LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (L.S.)
- CIEPQPF, Chemical Process Engineering and Forest Products Research Centre, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Sara Silva
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (S.S.); (M.M.P.)
| | - Ivana Jarak
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal;
| | - Rui A. Carvalho
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Associated Laboratory for Green Chemistry-Clean Technologies and Processes, REQUIMTE, Faculty of Sciences and Technology, University of Porto, 4050-313 Porto, Portugal
| | - Cláudia Cavadas
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (L.S.)
| | - Anabela P. Rolo
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carlos M. Palmeira
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Maria M. Pintado
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (S.S.); (M.M.P.)
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Correspondence: ; Tel.: +351-239-480-053
| |
Collapse
|
148
|
Mandal R, Cano R, Davis CD, Hayashi D, Jackson SA, Jones CM, Lampe JW, Latulippe ME, Lin NJ, Lippa KA, Piotrowski P, Da Silva SM, Swanson KS, Wishart DS. Workshop report: Toward the development of a human whole stool reference material for metabolomic and metagenomic gut microbiome measurements. Metabolomics 2020; 16:119. [PMID: 33164148 PMCID: PMC7649161 DOI: 10.1007/s11306-020-01744-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION To date, there has been little effort to develop standards for metabolome-based gut microbiome measurements despite the significant efforts toward standard development for DNA-based microbiome measurements. OBJECTIVES The National Institute of Standards and Technology (NIST), The BioCollective (TBC), and the North America Branch of the International Life Sciences Institute (ILSI North America) are collaborating to extend NIST's efforts to develop a Human Whole Stool Reference Material for the purpose of method harmonization and eventual quality control. METHODS The reference material will be rationally designed for adequate quality assurance and quality control (QA/QC) for underlying measurements in the study of the impact of diet and nutrition on functional aspects of the host gut microbiome and relationships of those functions to health. To identify which metabolites deserve priority in their value assignment, NIST, TBC, and ILSI North America jointly conducted a workshop on September 12, 2019 at the NIST campus in Gaithersburg, Maryland. The objective of the workshop was to identify metabolites for which evidence indicates relevance to health and disease and to decide on the appropriate course of action to develop a fit-for-purpose reference material. RESULTS This document represents the consensus opinions of workshop participants and co-authors of this manuscript, and provides additional supporting information. In addition to developing general criteria for metabolite selection and a preliminary list of proposed metabolites, this paper describes some of the strengths and limitations of this initiative given the current state of microbiome research. CONCLUSIONS Given the rapidly evolving nature of gut microbiome science and the current state of knowledge, an RM (as opposed to a CRM) measured for multiple metabolites is appropriate at this stage. As the science evolves, the RM can evolve to match the needs of the research community. Ultimately, the stool RM may exist in sequential versions. Beneficial to this evolution will be a clear line of communication between NIST and the stakeholder community to ensure alignment with current scientific understanding and community needs.
Collapse
Affiliation(s)
- Rupasri Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Raul Cano
- The BioCollective, LLC, 5650 N Washington St, Denver, CO, 80216, USA
| | - Cindy D Davis
- Office of Dietary Supplements, National Institutes of Health, Bethesda, MD, 20852, USA
| | | | - Scott A Jackson
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Christina M Jones
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Johanna W Lampe
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, M4-B802, PO Box 19024, Seattle, WA, 98109, USA
| | - Marie E Latulippe
- North American Branch of the International Life Sciences Institute (ILSI North America), 740 15th Street NW, Suite 600, Washington, DC, 20005, USA.
| | - Nancy J Lin
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Katrice A Lippa
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Paulina Piotrowski
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Sandra M Da Silva
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Kelly S Swanson
- University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - David S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| |
Collapse
|
149
|
Morishima S, Aoi W, Kawamura A, Kawase T, Takagi T, Naito Y, Tsukahara T, Inoue R. Intensive, prolonged exercise seemingly causes gut dysbiosis in female endurance runners. J Clin Biochem Nutr 2020; 68:253-258. [PMID: 34025029 PMCID: PMC8129978 DOI: 10.3164/jcbn.20-131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022] Open
Abstract
Intensive, prolonged exercise is known to induce gastrointestinal disorders such as diarrhea, with gut dysbiosis suggested as being one of the causatives. In the present study, we wanted to investigate the relationship between intensive exercise and the gut microbiota status. To that end, the microbiota, the moisture content and the bacterial metabolites (e.g., organic acids) of female endurance runners (n = 15) and those of non-athletic but healthy, age-matching female controls (n = 14) were compared. The analysis of the gut microbiota analysis showed that, unlike control subjects, female endurance runners had distinct microbiotas, with some bacteria found in higher abundances likely being involved in gut inflammation. The concentration of succinate, a gut bacterial metabolite regarded as undesirable when accumulated in the lumen, was significantly (p<0.05) higher in the female endurance runners. Faecalibacterium, that was significantly (p<0.05) abundant in female endurance runners, can produce succinate in certain environments and hence may contribute to succinate accumulation, at least partly. The present work suggested that the gut microbiotas of female endurance runners are seemingly dysbiotic when compared with those of control subjects. Further investigation of the mechanism by which intensive, prolonged exercise affects the gut microbiota is recommended.
Collapse
Affiliation(s)
- So Morishima
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan.,Laboratory of Animal Science, Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho, Sakyo-ku, Kyoto 606-8522, Japan
| | - Wataru Aoi
- Laboratory of Nutrition Science, Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho, Sakyo-ku, Kyoto 606-8522, Japan
| | - Aki Kawamura
- Sports Science Research Promotion Center, Nippon Sport Science University, 7-1-1 Fukasawa, Setagaya-ku, Tokyo 158-8508, Japan
| | - Takahiro Kawase
- Kyoto Institute of Nutrition & Pathology, Ujitawara, Kyoto 610-0231, Japan
| | - Tomohisa Takagi
- Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan.,Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan.,Laboratory of Animal Science, Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho, Sakyo-ku, Kyoto 606-8522, Japan
| |
Collapse
|
150
|
Schierová D, Březina J, Mrázek J, Fliegerová KO, Kvasnová S, Bajer L, Drastich P. Gut Microbiome Changes in Patients with Active Left-Sided Ulcerative Colitis after Fecal Microbiome Transplantation and Topical 5-aminosalicylic Acid Therapy. Cells 2020; 9:cells9102283. [PMID: 33066233 PMCID: PMC7602113 DOI: 10.3390/cells9102283] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease, and intestinal bacteria are implicated in the pathogenesis of this disorder. The administration of aminosalicylates (5-ASA) is a conventional treatment that targets the mucosa, while fecal microbial transplantation (FMT) is a novel treatment that directly targets the gut microbiota. The aim of this study was to identify changes in fecal bacterial composition after both types of treatments and evaluate clinical responses. Sixteen patients with active left-sided UC underwent enema treatment using 5-ASA (n = 8) or FMT (n = 8) with a stool from a single donor. Fecal microbiota were analyzed by 16S rDNA high-throughput sequencing, and clinical indices were used to assess the efficacy of treatments. 5-ASA therapy resulted in clinical remission in 50% (4/8) of patients, but no correlation with changes in fecal bacteria was observed. In FMT, remission was achieved in 37.5% (3/8) of patients and was associated with a significantly increased relative abundance of the families Lachnospiraceae, Ruminococcaceae, and Clostridiaceae of the phylum Firmicutes, and Bifidobacteriaceae and Coriobacteriaceae of the phylum Actinobacteria. At the genus level, Faecalibacterium, Blautia, Coriobacteria, Collinsela, Slackia, and Bifidobacterium were significantly more frequent in patients who reached clinical remission. However, the increased abundance of beneficial taxa was not a sufficient factor to achieve clinical improvement in all UC patients. Nevertheless, our preliminary results indicate that FMT as non-drug-using method is thought to be a promising treatment for UC patients.
Collapse
Affiliation(s)
- Dagmar Schierová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
- Correspondence: (D.S.); (J.M.); Tel.: +420-2-6709-0509 (D.S.); +420-2-6709-0506 (J.M.)
| | - Jan Březina
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| | - Jakub Mrázek
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
- Correspondence: (D.S.); (J.M.); Tel.: +420-2-6709-0509 (D.S.); +420-2-6709-0506 (J.M.)
| | - Kateřina Olša Fliegerová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
| | - Simona Kvasnová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
| | - Lukáš Bajer
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| | - Pavel Drastich
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| |
Collapse
|