101
|
Zhang X, Wu J, Zhou C, Tan Z, Jiao J. Spatial and temporal organization of jejunal microbiota in goats during animal development process. J Appl Microbiol 2020; 131:68-79. [PMID: 33300169 DOI: 10.1111/jam.14961] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/31/2022]
Abstract
AIMS This research aimed to investigate the temporal bacterial colonization relating to non-rumination, transition and rumination phases, together with the spatial organization of microbial community in the jejunal mucosa and digesta of goats. METHODS AND RESULTS This study explored the colonization programme of the jejunal microbiota by employing 16S rRNA amplicon sequencing. The colonization pattern of jejunal bacterial community exhibited an age- and gut region-dependent progression during animal development process. Approximately 268 bacterial signatures contributed to the discrimination between gut regions, with Lactobacillus, Ruminococcus, Eubacterium and Clostridium_sensu_stricto were enriched in the jejunal digesta, and Bacteroides and unclassified bacteria were enriched in the jejunal mucosa. Intriguingly, a shift from Lactobacillus to Butyrivibrio, Eubacterium and Ruminococcus after d 20 was observed for jejunal digesta. In mucosa, Bifidobacterium, Corynebacterium, Faecalibacterium and Roseburia increased with age (P < 0·05) while Arcobacter, Bacteroides and Porphyromonas peaked at d 10. CONCLUSIONS The jejunal bacterial community was settled after solid starter provision, which may mark the potential boundary of a timeframe for intervention in goats. The spatial heterogeneity highlighted the complicacy of ecological niches during manipulation of gut microbiota. SIGNIFICANCE AND IMPACT OF THE STUDY The present study extended the understanding of microbial programming and niche specific in the jejunum among different life stages and the basal cognition of persistent enhancement of nutrient utilization and decline of enteric diseases in ruminants.
Collapse
Affiliation(s)
- X Zhang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China.,University of the Chinese Academy of Sciences, Beijing, China
| | - J Wu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China.,University of the Chinese Academy of Sciences, Beijing, China
| | - C Zhou
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| | - Z Tan
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| | - J Jiao
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P. R. China
| |
Collapse
|
102
|
Schroeder BO, Birchenough GMH, Pradhan M, Nyström EEL, Henricsson M, Hansson GC, Bäckhed F. Obesity-associated microbiota contributes to mucus layer defects in genetically obese mice. J Biol Chem 2020; 295:15712-15726. [PMID: 32900852 PMCID: PMC7667970 DOI: 10.1074/jbc.ra120.015771] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
The intestinal mucus layer is a physical barrier separating the tremendous number of gut bacteria from the host epithelium. Defects in the mucus layer have been linked to metabolic diseases, but previous studies predominantly investigated mucus function during high-caloric/low-fiber dietary interventions, thus making it difficult to separate effects mediated directly through diet quality from potential obesity-dependent effects. As such, we decided to examine mucus function in mouse models with metabolic disease to distinguish these factors. Here we show that, in contrast to their lean littermates, genetically obese (ob/ob) mice have a defective inner colonic mucus layer that is characterized by increased penetrability and a reduced mucus growth rate. Exploiting the coprophagic behavior of mice, we next co-housed ob/ob and lean mice to investigate if the gut microbiota contributed to these phenotypes. Co-housing rescued the defect of the mucus growth rate, whereas mucus penetrability displayed an intermediate phenotype in both mouse groups. Of note, non-obese diabetic mice with high blood glucose levels displayed a healthy colonic mucus barrier, indicating that the mucus defect is obesity- rather than glucose-mediated. Thus, our data suggest that the gut microbiota community of obesity-prone mice may regulate obesity-associated defects in the colonic mucosal barrier, even in the presence of dietary fiber.
Collapse
Affiliation(s)
- Bjoern O Schroeder
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Laboratory for Molecular Infection Medicine Sweden (MIMS), Department of Molecular Biology, Umeå University, Umeå, Sweden.
| | - George M H Birchenough
- Department of Medical BiochemistryInstitute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Meenakshi Pradhan
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth E L Nyström
- Department of Medical BiochemistryInstitute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar C Hansson
- Department of Medical BiochemistryInstitute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
103
|
Rodríguez C, Romero E, Garrido-Sanchez L, Alcaín-Martínez G, Andrade RJ, Taminiau B, Daube G, García-Fuentes E. MICROBIOTA INSIGHTS IN CLOSTRIDIUM DIFFICILE INFECTION AND INFLAMMATORY BOWEL DISEASE. Gut Microbes 2020; 12:1725220. [PMID: 32129694 PMCID: PMC7524151 DOI: 10.1080/19490976.2020.1725220] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation that includes Crohn´s disease (CD) and ulcerative colitis (UC). Although the etiology is still unknown, some specific factors have been directly related to IBD, including genetic factors, abnormal intestinal immunity, and/or gut microbiota modifications. Recent findings highlight the primary role of the gut microbiota closely associated with a persistent inappropriate inflammatory response. This gut environment of dysbiosis in a susceptible IBD host can increasingly worsen and lead to colonization and infection with some opportunistic pathogens, especially Clostridium difficile. C. difficile is an intestinal pathogen considered the main cause of antibiotic-associated diarrhea and colitis and an important complication of IBD, which can trigger or worsen an IBD flare. Recent findings have highlighted the loss of bacterial cooperation in the gut ecosystem, as well as the pronounced intestinal dysbiosis, in patients suffering from IBD and concomitant C. difficile infection (CDI). The results of intestinal microbiota studies are still limited and often difficult to compare because of the variety of disease conditions. However, these data provide important clues regarding the main modifications and interrelations in the complicated gut ecosystem to better understand both diseases and to take advantage of the development of new therapeutic strategies. In this review, we analyze in depth the gut microbiota changes associated with both forms of IBD and CDI and their similarity with the dysbiosis that occurs in CDI. We also discuss the metabolic pathways that favor the proliferation or decrease in several important taxa directly related to the disease.
Collapse
Affiliation(s)
- C. Rodríguez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain,CONTACT C. Rodríguez Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, SpainUnidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Vitoria, Málaga, Spain
| | - E. Romero
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - L. Garrido-Sanchez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - G. Alcaín-Martínez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - RJ. Andrade
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain,Department of Medicine and Dermatology, Universidad de Málaga, Málaga, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Málaga, Spain
| | - B. Taminiau
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - G. Daube
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - E. García-Fuentes
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| |
Collapse
|
104
|
Wardill HR, da Silva Ferreira AR, Lichtenberg Cloo S, Havinga R, Harmsen HJM, Vermeij WP, Tissing WJE. Pre-therapy fasting slows epithelial turnover and modulates the microbiota but fails to mitigate methotrexate-induced gastrointestinal mucositis. Gut Microbes 2020; 12:1-9. [PMID: 32844722 PMCID: PMC7524354 DOI: 10.1080/19490976.2020.1809332] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Recent findings by Tang et al. (2020) show dietary restriction (30%, 2 weeks) prevents methotrexate-induced mortality by modulation of the microbiota, specifically the expansion of Lactobacillus. While fundamentally insightful, upscaling this schedule is a major obstacle to clinical uptake. Here, we evaluate a safe and clinically achievable schedule of pre-therapy fasting for 48 h on microbiota composition, body composition and intestinal proliferation, and assess its impact on the severity of methotrexate-induced gastrointestinal mucositis using a validated preclinical rat model. METHODS Age- and weight-matched male Wistar rats were treated with a sublethal dose of 45 mg/kg methotrexate with or without pre-therapy fasting. The impact of acute fasting on epithelial proliferation, body composition and the microbiota was assessed using plasma citrulline, Ki67 immunohistochemistry, miniSpec and 16S rRNA sequencing. The severity of gastrointestinal mucositis was evaluated using plasma citrulline and body weight. RESULTS Whilst pre-therapy fasting slowed epithelial proliferation and increased microbial diversity and richness, it also induced significant weight loss and was unable to attenuate the severity of mucositis in both age- and weight-matched groups. In contrast to Tang et al., we saw no expansion of Lactobacillus following acute fasting. CONCLUSIONS Our findings suggest that the beneficial effects of acute fasting are masked by the detrimental effects on body weight and composition and lacking influence on Lactobacillus. Future studies should consider alternative fasting schedules or aim to induce comparable microbial and mucosal manipulation without compromising body composition using clinically feasible methods of dietary or microbial intervention.
Collapse
Affiliation(s)
- H. R. Wardill
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia,Department of Pediatrics, University Medical Centre Groningen, The University of Groningen, Groningen, The Netherlands,CONTACT H. R. Wardill Adelaide Medical School, The University of Adelaide, AustraliaThis article has been republished with minor changes. These changes do not impact the academic content of the article
| | - A. R. da Silva Ferreira
- Department of Medical Microbiology, University Medical Centre Groningen, The University of Groningen, Groningen, The Netherlands
| | - S. Lichtenberg Cloo
- Department of Medical Microbiology, University Medical Centre Groningen, The University of Groningen, Groningen, The Netherlands
| | - R. Havinga
- Department of Pediatrics, University Medical Centre Groningen, The University of Groningen, Groningen, The Netherlands
| | - H. J. M. Harmsen
- Department of Medical Microbiology, University Medical Centre Groningen, The University of Groningen, Groningen, The Netherlands
| | - W. P. Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands,Oncode Institute, Utrecht, The Netherlands
| | - W. J. E. Tissing
- Department of Pediatrics, University Medical Centre Groningen, The University of Groningen, Groningen, The Netherlands,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
105
|
Gamage HKAH, Chong RWW, Bucio-Noble D, Kautto L, Hardikar AA, Ball MS, Molloy MP, Packer NH, Paulsen IT. Changes in dietary fiber intake in mice reveal associations between colonic mucin O-glycosylation and specific gut bacteria. Gut Microbes 2020; 12:1802209. [PMID: 32991816 PMCID: PMC7781582 DOI: 10.1080/19490976.2020.1802209] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The colonic mucus layer, comprised of highly O-glycosylated mucins, is vital to mediating host-gut microbiota interactions, yet the impact of dietary changes on colonic mucin O-glycosylation and its associations with the gut microbiota remains unexplored. Here, we used an array of omics techniques including glycomics to examine the effect of dietary fiber consumption on the gut microbiota, colonic mucin O-glycosylation and host physiology of high-fat diet-fed C57BL/6J mice. The high-fat diet group had significantly impaired glucose tolerance and altered liver proteome, gut microbiota composition, and short-chain fatty acid production compared to normal chow diet group. While dietary fiber inclusion did not reverse all high fat-induced modifications, it resulted in specific changes, including an increase in the relative abundance of bacterial families with known fiber digesters and a higher propionate concentration. Conversely, colonic mucin O-glycosylation remained similar between the normal chow and high-fat diet groups, while dietary fiber intervention resulted in major alterations in O-glycosylation. Correlation network analysis revealed previously undescribed associations between specific bacteria and mucin glycan structures. For example, the relative abundance of the bacterium Parabacteroides distasonis positively correlated with glycan structures containing one terminal fucose and correlated negatively with glycans containing two terminal fucose residues or with both an N-acetylneuraminic acid and a sulfate residue. This is the first comprehensive report of the impact of dietary fiber on the colonic mucin O-glycosylation and associations of these mucosal glycans with specific gut bacteria.
Collapse
Affiliation(s)
- Hasinika K. A. H. Gamage
- ARC Industrial Transformation Training Centre for Molecular Technologies in the Food Industry, Macquarie University, Sydney, Australia,Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Raymond W. W. Chong
- ARC Industrial Transformation Training Centre for Molecular Technologies in the Food Industry, Macquarie University, Sydney, Australia,Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Daniel Bucio-Noble
- ARC Industrial Transformation Training Centre for Molecular Technologies in the Food Industry, Macquarie University, Sydney, Australia,Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Liisa Kautto
- ARC Industrial Transformation Training Centre for Molecular Technologies in the Food Industry, Macquarie University, Sydney, Australia,Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Anandwardhan A. Hardikar
- Islet Biology and Diabetes, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | | | - Mark P. Molloy
- ARC Industrial Transformation Training Centre for Molecular Technologies in the Food Industry, Macquarie University, Sydney, Australia,Department of Molecular Sciences, Macquarie University, Sydney, Australia,Mark P. Molloy Bowel Cancer and Biomarker Research, Kolling Institute, The University of Sydney, Australia
| | - Nicolle H. Packer
- ARC Industrial Transformation Training Centre for Molecular Technologies in the Food Industry, Macquarie University, Sydney, Australia,Department of Molecular Sciences, Macquarie University, Sydney, Australia,Nicolle H. Packer
| | - Ian T. Paulsen
- ARC Industrial Transformation Training Centre for Molecular Technologies in the Food Industry, Macquarie University, Sydney, Australia,Department of Molecular Sciences, Macquarie University, Sydney, Australia,CONTACT Ian Paulsen Department of Molecular Sciences, Macquarie University, Australia
| |
Collapse
|
106
|
Nilsen M, Madelen Saunders C, Leena Angell I, Arntzen MØ, Lødrup Carlsen KC, Carlsen KH, Haugen G, Heldal Hagen L, Carlsen MH, Hedlin G, Monceyron Jonassen C, Nordlund B, Maria Rehbinder E, Skjerven HO, Snipen L, Cathrine Staff A, Vettukattil R, Rudi K. Butyrate Levels in the Transition from an Infant- to an Adult-Like Gut Microbiota Correlate with Bacterial Networks Associated with Eubacterium Rectale and Ruminococcus Gnavus. Genes (Basel) 2020; 11:genes11111245. [PMID: 33105702 PMCID: PMC7690385 DOI: 10.3390/genes11111245] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 01/14/2023] Open
Abstract
Relatively little is known about the ecological forces shaping the gut microbiota composition during infancy. Therefore, the objective of the present study was to identify the nutrient utilization- and short-chain fatty acid (SCFA) production potential of gut microbes in infants during the first year of life. Stool samples were obtained from mothers at 18 weeks of pregnancy and from infants at birth (first stool) at 3, 6, and 12-months of age from the general population-based PreventADALL cohort. We identified the taxonomic and SCFA composition in 100 mother-child pairs. The SCFA production and substrate utilization potential of gut microbes were observed by multiomics (shotgun sequencing and proteomics) on six infants. We found a four-fold increase in relative butyrate levels from 6 to 12 months of infant age. The increase was correlated to Eubacterium rectale and its bacterial network, and Faecalibacterium prausnitzii relative abundance, while low butyrate at 12 months was correlated to Ruminococcus gnavus and its associated network of bacteria. Both E. rectale and F. prausnitzii expressed enzymes needed for butyrate production and enzymes related to dietary fiber degradation, while R. gnavus expressed mucus-, fucose, and human milk oligosaccharides (HMO)-related degradation enzymes. Therefore, we believe that the presence of E. rectale, its network, and F. prausnitzii are key bacteria in the transition from an infant- to an adult-like gut microbiota with respect to butyrate production. Our results indicate that the transition from an infant- to an adult-like gut microbiota with respect to butyrate producing bacteria, occurs between 6 and 12 months of infant age. The bacteria associated with the increased butyrate ratio/levels were E. rectale and F. prausnitzii, which potentially utilize a variety of dietary fibers based on the glycoside hydrolases (GHs) expressed. R. gnavus with a negative association to butyrate potentially utilizes mucin, fucose, and HMO components. This knowledge could have future importance in understanding how microbial metabolites can impact infant health and development.
Collapse
Affiliation(s)
- Morten Nilsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1430 Ås, Norway; (I.L.A.); (M.Ø.A.); (L.H.H.); (C.M.J.); (L.S.); (K.R.)
- Correspondence: (M.N.); (C.M.S.)
| | - Carina Madelen Saunders
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, 0450 Oslo, Norway; (K.C.L.C.); (K.-H.C.); (H.O.S.); (R.V.)
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
- Correspondence: (M.N.); (C.M.S.)
| | - Inga Leena Angell
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1430 Ås, Norway; (I.L.A.); (M.Ø.A.); (L.H.H.); (C.M.J.); (L.S.); (K.R.)
| | - Magnus Ø. Arntzen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1430 Ås, Norway; (I.L.A.); (M.Ø.A.); (L.H.H.); (C.M.J.); (L.S.); (K.R.)
| | - Karin C. Lødrup Carlsen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, 0450 Oslo, Norway; (K.C.L.C.); (K.-H.C.); (H.O.S.); (R.V.)
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
| | - Kai-Håkon Carlsen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, 0450 Oslo, Norway; (K.C.L.C.); (K.-H.C.); (H.O.S.); (R.V.)
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
| | - Guttorm Haugen
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
- Division of Obstetrics and Gynaecology, Oslo University Hospital, 0450 Oslo, Norway
| | - Live Heldal Hagen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1430 Ås, Norway; (I.L.A.); (M.Ø.A.); (L.H.H.); (C.M.J.); (L.S.); (K.R.)
| | - Monica H. Carlsen
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0405 Oslo, Norway;
| | - Gunilla Hedlin
- Astrid Lindgren Children’s Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden; (G.H.); (B.N.)
- Department of Women’s and Children’s Health, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Christine Monceyron Jonassen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1430 Ås, Norway; (I.L.A.); (M.Ø.A.); (L.H.H.); (C.M.J.); (L.S.); (K.R.)
- Genetic Unit, Centre for Laboratory Medicine, Østfold Hospital Trust, 1714 Kalnes, Norway
| | - Björn Nordlund
- Astrid Lindgren Children’s Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden; (G.H.); (B.N.)
- Department of Women’s and Children’s Health, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Eva Maria Rehbinder
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
- Department of Dermatology, Oslo University Hospital, 0424 Oslo, Norway
| | - Håvard O. Skjerven
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, 0450 Oslo, Norway; (K.C.L.C.); (K.-H.C.); (H.O.S.); (R.V.)
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
| | - Lars Snipen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1430 Ås, Norway; (I.L.A.); (M.Ø.A.); (L.H.H.); (C.M.J.); (L.S.); (K.R.)
| | - Anne Cathrine Staff
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
- Division of Obstetrics and Gynaecology, Oslo University Hospital, 0450 Oslo, Norway
| | - Riyas Vettukattil
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, 0450 Oslo, Norway; (K.C.L.C.); (K.-H.C.); (H.O.S.); (R.V.)
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (G.H.); (E.M.R.); (A.C.S.)
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1430 Ås, Norway; (I.L.A.); (M.Ø.A.); (L.H.H.); (C.M.J.); (L.S.); (K.R.)
| |
Collapse
|
107
|
Bell A, Juge N. Mucosal glycan degradation of the host by the gut microbiota. Glycobiology 2020; 31:691-696. [PMID: 33043970 PMCID: PMC8252862 DOI: 10.1093/glycob/cwaa097] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/21/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota plays a major role in human health and an alteration in gut microbiota structure and function has been implicated in several diseases. In the colon, mucus covering the epithelium is critical to maintain a homeostatic relationship with the gut microbiota by harboring a microbial community at safe distance from the epithelium surface. The mucin glycans composing the mucus layer provide binding sites and a sustainable source of nutrients to the bacteria inhabiting the mucus niche. Access to these glycan chains requires a complement of glycoside hydrolases (GHs) produced by bacteria across the phyla constituting the human gut microbiota. Due to the increased recognition of the role of mucus-associated microbes in human health, how commensal bacteria breakdown and utilize host mucin glycans has become of increased interest and is reviewed here. This short review provides an overview of the strategies evolved by gut commensal bacteria to access this rich source of the nutrient with a focus on the GHs involved in mucin degradation.
Collapse
Affiliation(s)
- Andrew Bell
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Rosalind Franklin Road Norwich Research Park, Norwich NR4 7UQ, UK
| | - Nathalie Juge
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Rosalind Franklin Road Norwich Research Park, Norwich NR4 7UQ, UK
| |
Collapse
|
108
|
Mohammed A, Alghetaa HK, Zhou J, Chatterjee S, Nagarkatti P, Nagarkatti M. Protective effects of Δ 9 -tetrahydrocannabinol against enterotoxin-induced acute respiratory distress syndrome are mediated by modulation of microbiota. Br J Pharmacol 2020; 177:5078-5095. [PMID: 32754917 PMCID: PMC7436585 DOI: 10.1111/bph.15226] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose Staphylococcal enterotoxin‐B (SEB) is one of the most potent bacterial superantigens that exerts profound toxic effects by inducing a cytokine storm. Inhaled SEB can cause acute respiratory distress syndrome (ARDS), which is often fatal and with no effective treatments. Experimental Approach Efficacy of Δ9‐tetrahydrocannabinol (THC) was tested in a mouse model of SEB‐mediated ARDS, in which lung inflammation, alterations in gut/lung microbiota and production of short‐chain fatty acids (SCFAs) was measured. Gene dysregulation of lung epithelial cells was studied by transcriptome arrays. Faecal microbiota transplantation (FMT) was performed to confirm the role of microbiota in suppressing ARDS. Key Results While SEB triggered ARDS and 100% mortality in mice, THC protected the mice from fatality. Pyrosequencing analysis revealed that THC caused significant and similar alterations in microbiota in the lungs and gut of mice exposed to SEB. THC significantly increased the abundance of beneficial bacterial species, Ruminococcus gnavus, but decreased pathogenic microbiota, Akkermansia muciniphila. FMT confirmed that THC‐mediated reversal of microbial dysbiosis played crucial role in attenuation of SEB‐mediated ARDS. THC treatment caused an increase in SCFA, of which propionic acid was found to inhibit the inflammatory response. Transcriptome array showed that THC up‐regulated several genes like lysozyme1 and lysozyme2, β‐defensin‐2, claudin, zonula‐1, occludin‐1, Mucin2 and Muc5b while down‐regulating β‐defensin‐1. Conclusion and Implications The study demonstrates for the first time that THC attenuates SEB‐mediated ARDS and toxicity by altering the microbiota in the lungs and the gut as well as promoting antimicrobial and anti‐inflammatory pathways.
Collapse
Affiliation(s)
- Amira Mohammed
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Hasan K Alghetaa
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Juhua Zhou
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Saurabh Chatterjee
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| |
Collapse
|
109
|
Gren C, Spiegelhauer MR, Rotbain EC, Ehmsen BK, Kampmann P, Andersen LP. Ruminococcus gnavus bacteraemia in a patient with multiple haematological malignancies. Access Microbiol 2020; 1:e000048. [PMID: 32974553 PMCID: PMC7470407 DOI: 10.1099/acmi.0.000048] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 07/17/2019] [Indexed: 12/16/2022] Open
Abstract
We present a case of Ruminococcus gnavus sepsis in a woman suffering from multiple myeloma and myelodysplastic syndrome. R. gnavus, a Gram-positive coccus and a gut commensal, has been described in nine cases of infection in the literature, with most infections having occurred in patients with either gastrointestinal symptoms or prosthesis infections. In this case, R gnavus was identified by mass spectrometry, and showed susceptibility to penicillin, meropenem, tetracycline, metronidazole and clindamycin. The patient was successfully treated initially with intravenous piperacillin/tazobactam and metronidazole, and then switched to oral penicillin and metronidazole. The cause of infection is hypothesized to have been a shift in the gut microbiota towards an excess growth of R. gnavus caused by immunosuppression, and bacterial translocation across a vulnerable mucosal barrier due to prednisolone treatment and severe thrombocytopenia.
Collapse
Affiliation(s)
- Caroline Gren
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Peter Kampmann
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
110
|
Jung Y, Tagele SB, Son H, Ibal JC, Kerfahi D, Yun H, Lee B, Park CY, Kim ES, Kim SJ, Shin JH. Modulation of Gut Microbiota in Korean Navy Trainees following a Healthy Lifestyle Change. Microorganisms 2020; 8:microorganisms8091265. [PMID: 32825401 PMCID: PMC7569816 DOI: 10.3390/microorganisms8091265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/11/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Environmental factors can influence the composition of gut microbiota, but understanding the combined effect of lifestyle factors on adult gut microbiota is limited. Here, we investigated whether changes in the modifiable lifestyle factors, such as cigarette smoking, alcohol consumption, sleep duration, physical exercise, and body mass index affected the gut microbiota of Korean navy trainees. The navy trainees were instructed to stop smoking and alcohol consumption and follow a sleep schedule and physical exercise regime for eight weeks. For comparison, healthy Korean civilians, who had no significant change in lifestyles for eight weeks were included in this study. A total of 208 fecal samples were collected from navy trainees (n = 66) and civilians (n = 38) at baseline and week eight. Gut flora was assessed by sequencing the highly variable region of the 16S rRNA gene. The α-and β -diversity of gut flora of both the test and control groups were not significantly changed after eight weeks. However, there was a significant difference among individuals. Smoking had a significant impact in altering α-diversity. Our study showed that a healthy lifestyle, particularly cessation of smoking, even in short periods, can affect the gut microbiome by enhancing the abundance of beneficial taxa and reducing that of harmful taxa.
Collapse
Affiliation(s)
- YeonGyun Jung
- School of Applied Biosciences, Kyungpook National University, Daegu 41566, Korea; (Y.J.); (S.B.T.); (H.S.); (J.C.I.)
| | - Setu Bazie Tagele
- School of Applied Biosciences, Kyungpook National University, Daegu 41566, Korea; (Y.J.); (S.B.T.); (H.S.); (J.C.I.)
- Department of Applied Plant Sciences, University of Gondar, Gondar 196, Ethiopia
| | - HyunWoo Son
- School of Applied Biosciences, Kyungpook National University, Daegu 41566, Korea; (Y.J.); (S.B.T.); (H.S.); (J.C.I.)
| | - Jerald Conrad Ibal
- School of Applied Biosciences, Kyungpook National University, Daegu 41566, Korea; (Y.J.); (S.B.T.); (H.S.); (J.C.I.)
| | - Dorsaf Kerfahi
- Department of Biological Sciences, Keimyung University, Daegu 42601, Korea;
| | - Hyunju Yun
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (H.Y.); (B.L.); (C.Y.P.)
| | - Bora Lee
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (H.Y.); (B.L.); (C.Y.P.)
| | - Clara Yongjoo Park
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (H.Y.); (B.L.); (C.Y.P.)
| | - Eun Soo Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Sang-Jun Kim
- Department of Natural Sciences, Republic of Korea Naval Academy, Changwon 51702, Korea;
| | - Jae-Ho Shin
- School of Applied Biosciences, Kyungpook National University, Daegu 41566, Korea; (Y.J.); (S.B.T.); (H.S.); (J.C.I.)
- Correspondence: ; Tel.: +82-53-950-5716; Fax: +82-53-953-7233
| |
Collapse
|
111
|
Bell A, Severi E, Lee M, Monaco S, Latousakis D, Angulo J, Thomas GH, Naismith JH, Juge N. Uncovering a novel molecular mechanism for scavenging sialic acids in bacteria. J Biol Chem 2020; 295:13724-13736. [PMID: 32669363 PMCID: PMC7535918 DOI: 10.1074/jbc.ra120.014454] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
The human gut symbiont Ruminococcus gnavus scavenges host-derived N-acetylneuraminic acid (Neu5Ac) from mucins by converting it to 2,7-anhydro-Neu5Ac. We previously showed that 2,7-anhydro-Neu5Ac is transported into R. gnavus ATCC 29149 before being converted back to Neu5Ac for further metabolic processing. However, the molecular mechanism leading to the conversion of 2,7-anhydro-Neu5Ac to Neu5Ac remained elusive. Using 1D and 2D NMR, we elucidated the multistep enzymatic mechanism of the oxidoreductase (RgNanOx) that leads to the reversible conversion of 2,7-anhydro-Neu5Ac to Neu5Ac through formation of a 4-keto-2-deoxy-2,3-dehydro-N-acetylneuraminic acid intermediate and NAD+ regeneration. The crystal structure of RgNanOx in complex with the NAD+ cofactor showed a protein dimer with a Rossman fold. Guided by the RgNanOx structure, we identified catalytic residues by site-directed mutagenesis. Bioinformatics analyses revealed the presence of RgNanOx homologues across Gram-negative and Gram-positive bacterial species and co-occurrence with sialic acid transporters. We showed by electrospray ionization spray MS that the Escherichia coli homologue YjhC displayed activity against 2,7-anhydro-Neu5Ac and that E. coli could catabolize 2,7-anhydro-Neu5Ac. Differential scanning fluorimetry analyses confirmed the binding of YjhC to the substrates 2,7-anhydro-Neu5Ac and Neu5Ac, as well as to co-factors NAD and NADH. Finally, using E. coli mutants and complementation growth assays, we demonstrated that 2,7-anhydro-Neu5Ac catabolism in E. coli depended on YjhC and on the predicted sialic acid transporter YjhB. These results revealed the molecular mechanisms of 2,7-anhydro-Neu5Ac catabolism across bacterial species and a novel sialic acid transport and catabolism pathway in E. coli.
Collapse
Affiliation(s)
- Andrew Bell
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | | | - Micah Lee
- Division of Structural Biology, University of Oxford, Headington, Oxford, United Kingdom
| | - Serena Monaco
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Dimitrios Latousakis
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Jesus Angulo
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom; Departamento de Química Orgánica, Universidad de Sevilla, Sevilla, Spain; Instituto de Investigaciones Químicas (CSIC-US), Sevilla, Spain
| | - Gavin H Thomas
- Department of Biology, University of York, York, United Kingdom
| | - James H Naismith
- Division of Structural Biology, University of Oxford, Headington, Oxford, United Kingdom
| | - Nathalie Juge
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich, United Kingdom.
| |
Collapse
|
112
|
Levade I, Saber MM, Midani FS, Chowdhury F, Khan AI, Begum YA, Ryan ET, David LA, Calderwood SB, Harris JB, LaRocque RC, Qadri F, Shapiro BJ, Weil AA. Predicting Vibrio cholerae Infection and Disease Severity Using Metagenomics in a Prospective Cohort Study. J Infect Dis 2020; 223:342-351. [PMID: 32610345 PMCID: PMC7857355 DOI: 10.1093/infdis/jiaa358] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Susceptibility to Vibrio cholerae infection is affected by blood group, age, and preexisting immunity, but these factors only partially explain who becomes infected. A recent study used 16S ribosomal RNA amplicon sequencing to quantify the composition of the gut microbiome and identify predictive biomarkers of infection with limited taxonomic resolution. METHODS To achieve increased resolution of gut microbial factors associated with V. cholerae susceptibility and identify predictors of symptomatic disease, we applied deep shotgun metagenomic sequencing to a cohort of household contacts of patients with cholera. RESULTS Using machine learning, we resolved species, strains, gene families, and cellular pathways in the microbiome at the time of exposure to V. cholerae to identify markers that predict infection and symptoms. Use of metagenomic features improved the precision and accuracy of prediction relative to 16S sequencing. We also predicted disease severity, although with greater uncertainty than our infection prediction. Species within the genera Prevotella and Bifidobacterium predicted protection from infection, and genes involved in iron metabolism were also correlated with protection. CONCLUSION Our results highlight the power of metagenomics to predict disease outcomes and suggest specific species and genes for experimental testing to investigate mechanisms of microbiome-related protection from cholera.
Collapse
Affiliation(s)
- Inès Levade
- Department of Biological Sciences, University of Montreal, Montreal, Quebec, Canada
| | - Morteza M Saber
- Department of Biological Sciences, University of Montreal, Montreal, Quebec, Canada
| | - Firas S Midani
- Program in Computational Biology and Bioinformatics, Duke University, Durham, North Carolina, USA,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA,Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Fahima Chowdhury
- Center for Vaccine Sciences, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Ashraful I Khan
- Center for Vaccine Sciences, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Yasmin A Begum
- Center for Vaccine Sciences, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Edward T Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lawrence A David
- Program in Computational Biology and Bioinformatics, Duke University, Durham, North Carolina, USA,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA,Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA,Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Stephen B Calderwood
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA,Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason B Harris
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Regina C LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Firdausi Qadri
- Center for Vaccine Sciences, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - B Jesse Shapiro
- Department of Biological Sciences, University of Montreal, Montreal, Quebec, Canada,Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada,McGill Genome Centre, Montreal, Quebec, Canada,Correspondence: B. Jesse Shapiro, McGill University, Montreal, Quebec, Canada ()
| | - Ana A Weil
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| |
Collapse
|
113
|
Wu H, Rebello O, Crost EH, Owen CD, Walpole S, Bennati-Granier C, Ndeh D, Monaco S, Hicks T, Colvile A, Urbanowicz PA, Walsh MA, Angulo J, Spencer DIR, Juge N. Fucosidases from the human gut symbiont Ruminococcus gnavus. Cell Mol Life Sci 2020; 78:675-693. [PMID: 32333083 PMCID: PMC7872956 DOI: 10.1007/s00018-020-03514-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/11/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022]
Abstract
The availability and repartition of fucosylated glycans within the gastrointestinal tract contributes to the adaptation of gut bacteria species to ecological niches. To access this source of nutrients, gut bacteria encode α-l-fucosidases (fucosidases) which catalyze the hydrolysis of terminal α-l-fucosidic linkages. We determined the substrate and linkage specificities of fucosidases from the human gut symbiont Ruminococcus gnavus. Sequence similarity network identified strain-specific fucosidases in R. gnavus ATCC 29149 and E1 strains that were further validated enzymatically against a range of defined oligosaccharides and glycoconjugates. Using a combination of glycan microarrays, mass spectrometry, isothermal titration calorimetry, crystallographic and saturation transfer difference NMR approaches, we identified a fucosidase with the capacity to recognize sialic acid-terminated fucosylated glycans (sialyl Lewis X/A epitopes) and hydrolyze α1–3/4 fucosyl linkages in these substrates without the need to remove sialic acid. Molecular dynamics simulation and docking showed that 3′-Sialyl Lewis X (sLeX) could be accommodated within the binding site of the enzyme. This specificity may contribute to the adaptation of R. gnavus strains to the infant and adult gut and has potential applications in diagnostic glycomic assays for diabetes and certain cancers.
Collapse
Affiliation(s)
- Haiyang Wu
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Osmond Rebello
- Ludger Ltd, Culham Science Centre, Abingdon, OX14 3EB, UK
| | - Emmanuelle H Crost
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - C David Owen
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK.,Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Samuel Walpole
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Chloe Bennati-Granier
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Didier Ndeh
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Serena Monaco
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Thomas Hicks
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Anna Colvile
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK.,Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK.,The John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | | | - Martin A Walsh
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK.,Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Jesus Angulo
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.,Departamento de Química Orgánica, Universidad de Sevilla, C/ Prof. García González, 1, 41012, Sevilla, Spain.,Instituto de Investigaciones Químicas (CSIC-US), Avda. Américo Vespucio, 49, 41092, Sevilla, Spain
| | | | - Nathalie Juge
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK.
| |
Collapse
|
114
|
Shimokawa C, Kato T, Takeuchi T, Ohshima N, Furuki T, Ohtsu Y, Suzue K, Imai T, Obi S, Olia A, Izumi T, Sakurai M, Arakawa H, Ohno H, Hisaeda H. CD8 + regulatory T cells are critical in prevention of autoimmune-mediated diabetes. Nat Commun 2020; 11:1922. [PMID: 32321922 PMCID: PMC7176710 DOI: 10.1038/s41467-020-15857-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 03/31/2020] [Indexed: 02/08/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which insulin-producing pancreatic β-cells are destroyed. Intestinal helminths can cause asymptomatic chronic and immunosuppressive infections and suppress disease in rodent models of T1D. However, the underlying regulatory mechanisms for this protection are unclear. Here, we report that CD8+ regulatory T (Treg) cells prevent the onset of streptozotocin -induced diabetes by a rodent intestinal nematode. Trehalose derived from nematodes affects the intestinal microbiota and increases the abundance of Ruminococcus spp., resulting in the induction of CD8+ Treg cells. Furthermore, trehalose has therapeutic effects on both streptozotocin-induced diabetes and in the NOD mouse model of T1D. In addition, compared with healthy volunteers, patients with T1D have fewer CD8+ Treg cells, and the abundance of intestinal Ruminococcus positively correlates with the number of CD8+ Treg cells in humans. Helminth infections are associated with a reduction in inflammatory pathology in rodent models of type 1 diabetes. Here, the authors show patient data and that trehalose (produced by H. polygyrus) can alter the microbiome of mice, inducing regulatory CD8+ T cells and reducing susceptibility to autoimmune diabetes.
Collapse
Affiliation(s)
- Chikako Shimokawa
- Department of Parasitology, National Institute of Infectious Disease, Tokyo, 162-8640, Japan. .,Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan. .,Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.
| | - Tamotsu Kato
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Immunobiolgy Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Tadashi Takeuchi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Grauduate School of Medicine, Keio University, Tokyo, 160-8582, Japan
| | - Noriyasu Ohshima
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Takao Furuki
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8502, Japan
| | - Yoshiaki Ohtsu
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Kazutomo Suzue
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Takashi Imai
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Seiji Obi
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Alex Olia
- Department of Parasitology, National Institute of Infectious Disease, Tokyo, 162-8640, Japan.,Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Takashi Izumi
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Minoru Sakurai
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8502, Japan
| | - Hirokazu Arakawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan. .,Immunobiolgy Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan. .,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Ebina, 243-0435, Japan.
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Disease, Tokyo, 162-8640, Japan. .,Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan.
| |
Collapse
|
115
|
Vacca M, Celano G, Calabrese FM, Portincasa P, Gobbetti M, De Angelis M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms 2020; 8:E573. [PMID: 32326636 PMCID: PMC7232163 DOI: 10.3390/microorganisms8040573] [Citation(s) in RCA: 873] [Impact Index Per Article: 174.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
The complex polymicrobial composition of human gut microbiota plays a key role in health and disease. Lachnospiraceae belong to the core of gut microbiota, colonizing the intestinal lumen from birth and increasing, in terms of species richness and their relative abundances during the host's life. Although, members of Lachnospiraceae are among the main producers of short-chain fatty acids, different taxa of Lachnospiraceae are also associated with different intra- and extraintestinal diseases. Their impact on the host physiology is often inconsistent across different studies. Here, we discuss changes in Lachnospiraceae abundances according to health and disease. With the aim of harnessing Lachnospiraceae to promote human health, we also analyze how nutrients from the host diet can influence their growth and how their metabolites can, in turn, influence host physiology.
Collapse
Affiliation(s)
- Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70121 Bari, Italy
| | - Marco Gobbetti
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| |
Collapse
|
116
|
Jeffery IB, Das A, O'Herlihy E, Coughlan S, Cisek K, Moore M, Bradley F, Carty T, Pradhan M, Dwibedi C, Shanahan F, O'Toole PW. Differences in Fecal Microbiomes and Metabolomes of People With vs Without Irritable Bowel Syndrome and Bile Acid Malabsorption. Gastroenterology 2020; 158:1016-1028.e8. [PMID: 31843589 DOI: 10.1053/j.gastro.2019.11.301] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 11/16/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Irritable bowel syndrome (IBS) is a heterogeneous disorder, but diagnoses and determination of subtypes are made based on symptoms. We profiled the fecal microbiomes of patients with and without IBS to identify biomarkers of this disorder. METHODS We collected fecal and urine samples from 80 patients with IBS (Rome IV criteria; 16-70 years old) and 65 matched individuals without IBS (control individuals), along with anthropometric, medical, and dietary information. Shotgun and 16S ribosomal RNA amplicon sequencing were performed on feces, whereas urine and fecal metabolites were analyzed by gas chromatography and liquid chromatography-mass spectrometry. Co-occurrence networks were generated based on significant Spearman correlations between data. Bile acid malabsorption (BAM) was identified in patients with diarrhea by retention of radiolabeled selenium-75 homocholic acid taurine. RESULTS Patients with IBS had significant differences in network connections between diet and fecal microbiomes compared with control individuals; these were accompanied by differences in fecal metabolomes. We did not find significant differences in fecal microbiota composition among patients with different IBS symptom subtypes. Fecal metabolome profiles could discriminate patients with IBS from control individuals. Urine metabolomes also differed significantly between patients with IBS and control individuals, but most discriminatory metabolites were related to diet or medications. Fecal metabolomes, but not microbiomes, could distinguish patients with IBS with vs those without BAM. CONCLUSIONS Despite the heterogeneity of IBS, patients have significant differences in urine and fecal metabolomes and fecal microbiome vs control individuals, independent of symptom-based subtypes of IBS. Fecal metabolome analysis can be used to distinguish patients with IBS with vs those without BAM. These findings might be used for developing microbe-based treatments for these disorders.
Collapse
Affiliation(s)
- Ian B Jeffery
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland
| | - Anubhav Das
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland
| | - Eileen O'Herlihy
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland
| | - Simone Coughlan
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland
| | - Katryna Cisek
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland
| | - Michael Moore
- Department of Radiology, Cork University Hospital, Cork, Ireland
| | - Fintan Bradley
- Medical Physics Department, Cork University Hospital, Cork, Ireland
| | - Tom Carty
- Medical Physics Department, Cork University Hospital, Cork, Ireland
| | - Meenakshi Pradhan
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland
| | - Chinmay Dwibedi
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland
| | - Fergus Shanahan
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Paul W O'Toole
- 4D pharma Cork Limited, Cavanagh Pharmacy Building, University College Cork, National University of Ireland, Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland; School of Microbiology, University College Cork, Ireland.
| |
Collapse
|
117
|
Zhang L, Song P, Zhang X, Metea C, Schleisman M, Karstens L, Leung E, Zhang J, Xu Q, Liu Y, Asquith M, Chu CQ. Alpha-Glucosidase Inhibitors Alter Gut Microbiota and Ameliorate Collagen-Induced Arthritis. Front Pharmacol 2020; 10:1684. [PMID: 32116681 PMCID: PMC7010955 DOI: 10.3389/fphar.2019.01684] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/24/2019] [Indexed: 02/05/2023] Open
Abstract
Acarose is an anti-diabetic drug and exhibits anti-arthritic effects. We hypothesized that acarbose influences the gut microbiota to affect the course of arthritis and tested this hypothesis in a collagen-induced arthritis (CIA) murine model. Acarbose in drinking water was administered via gastric gavage started prior to or at the time of CIA induction. Gut microbiota were evaluated with 16S rRNA gene sequencing from fecal pellets collected prior to arthritis induction, during onset of arthritis, and after treatment. Immune response was evaluated by measuring changes in T helper-17 (Th17) and T regulatory (Treg) cells in the spleen and intestine, as well as serum cytokine levels. Before induction of CIA, acarbose significantly reduced the incidence of arthritis and attenuated clinical severity of arthritis. The frequency of Th17 cells was significantly decreased in the intestinal lamina propria in acarbose treated mice. Mice that were treated with acarbose showed significantly increased CD4+CD25+Foxp3+ Treg cells with elevation of Helios and CCR6. A remarkable alteration in microbial community was observed in acarbose treated mice. Bacterial diversity and richness in mice with arthritis were significantly lower than those in acarbose treated groups. The frequency of Firmicutes was significantly reduced after arthritis onset but was restored after treatment with acarbose. The frequency of Lactobacillus, Anaeroplasma, Adlercreutzia, RF39 and Corynebacterium was significantly higher in control groups than in acarbose treated, while Oscillospira, Desulfovibrio and Ruminococcus enriched in acarbose treated group. Miglitol, another α-glucosidase inhibitor showed a similar but less potent anti-arthritic effect to that of acarbose. These data demonstrate that acarbose alleviated CIA through regulation of Th17/Treg cells in the intestinal mucosal immunity, which may have resulted from the impact of acarbose on gut microbial community. Inexpensive antidiabetic drugs with an excellent safety profile are potentially useful for managing rheumatoid arthritis.
Collapse
Affiliation(s)
- Lingshu Zhang
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, China
- Section of Rheumatology, VA Portland Health Care System, Portland, OR, United States
| | - Pingfang Song
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
| | - Xiaowei Zhang
- Section of Rheumatology, VA Portland Health Care System, Portland, OR, United States
| | - Christina Metea
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Matthew Schleisman
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Lisa Karstens
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, United States
| | - Eric Leung
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, United States
| | - Jun Zhang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Qiang Xu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
- Department of Rheumatology, The First Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Liu
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, China
| | - Mark Asquith
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
- Section of Rheumatology, VA Portland Health Care System, Portland, OR, United States
| |
Collapse
|
118
|
Journey EK, Ortega-Santos CP, Bruening M, Whisner CM. Changes in Weight Status and the Intestinal Microbiota Among College Freshman, Aged 18 Years. J Adolesc Health 2020; 66:166-171. [PMID: 31477511 PMCID: PMC6980309 DOI: 10.1016/j.jadohealth.2019.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/10/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
Abstract
PURPOSE The transition to college is a vulnerable period for weight gain and the onset of obesity. Gut microbes differ in obese compared with lean individuals, but gut microbiota in adolescent-aged college freshmen during a known period of weight gain have never been studied. This pre-post observational pilot study assessed associations between intestinal microbiota changes and weight-related outcomes in healthy adolescent college freshmen living in on-campus dormitories at Arizona State University (n = 39). METHODS We measured anthropometrics (waist circumference [WC], height, weight, and body mass index [BMI]) and collected fecal samples at the beginning and end of the 2015-2016 academic year. Fold changes in species-level microbes across time were measured by quantitative real-time polymerase chain reaction and used in correlation and multivariate regression analyses. RESULTS A total of 24 female and 15 male adolescents (aged 18.54 ± .67 years) participated in this study. Over the academic year, BMI and WC increased by .97±1.28 kg/m2 and 2.64±4.90 cm, respectively. Correlation analyses indicated a significant negative association between Akkermansia muciniphila and both percentage WC change and percentage BMI change (r = -.66, p < .01; and r = -.33, p = .04, respectively). Multivariate regression analysis controlling for sociodemographics showed a significant association between A. muciniphila and percentage WC change, but not percentage BMI change (R2 = .53, p < .01; and R2 = .24, p = .15, respectively). CONCLUSIONS As this was the first study in a university-based adolescent population to show a relationship between A. muciniphila and weight-related outcomes, further research is needed to explore these findings.
Collapse
Affiliation(s)
| | | | - Meg Bruening
- Arizona State University, School of Nutrition and Health Promotion
| | - Corrie M. Whisner
- Arizona State University, School of Nutrition and Health Promotion;,Correspondence: ; Tel.: +1 602-496-3348; Fax: +1 602-827-2253
| |
Collapse
|
119
|
Zhou ZY, Xu X, Zhou Y. [Research progress on carbohydrate active enzymes of human microbiome]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:666-670. [PMID: 31875448 DOI: 10.7518/hxkq.2019.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A massive variety of microorganisms live in and on the human body, especially at oral, skin, vaginal, gastroin-testinal, and respiratory sites. The complicated metabolic activities of microorganisms assist human digestive function and participate in a series of physiological and pathogenetic processes. Carbohydrate-active enzymes (CAZymes) are a series of enzymes that function in degradation, modification, and formation of glycoside bonds. Microbes regulate the physiological and pathogenetic processes of human body by producing various CAZymes to degrade and modify complex carbohydrates and generate signal molecules for further utilization in human cells. Here, we reviewed the mechanisms of complex carbohy-drate metabolism and related microbial CAZymes, especially in digestive tract and oral cavity. We also summarized the rela-tionship between microbial CAZymes and human health, and proposed potential applications.
Collapse
Affiliation(s)
- Zhi-Yan Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuan Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
120
|
Lefever S, Van Den Bossche D, Van Moerkercke W, D’Hondt M, Alegret Pampols MDC, Struyve M, De Bel A, Boudewijns M. Ruminococcus gnavus bacteremia, an uncommon presentation of a common member of the human gut microbiota: case report and literature review. Acta Clin Belg 2019; 74:435-438. [PMID: 31658898 DOI: 10.1080/17843286.2018.1541045] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Case report: We present a case of a 66-year-old female diagnosed with R. gnavus bacteremia associated with fecal peritonits secondary to small-bowel herniation and perforation. Identification as R. gnavus was delayed because of absence of this species in the MALDI-TOF MS database (Vitek MS, bioMérieux). Identification was provided by 16S rRNA gene sequencing. Review: R. gnavus, a Gram-positive, strictly anaerobic bacterium, is a member of the human gut microbiota. Dysbiosis in the gut microbiota, with increased amounts of R. gnavus, has been described in inflammatory bowel disease. R. gnavus has only been reported occasionally as the cause of infections. Hence the potential pathogenicity is not yet fully recognized, and data regarding the antimicrobial susceptibility profile are rare. Identification of anaerobic bacteria such as R. gnavus is greatly accelerated as a result of the introduction of MALDI-TOF MS. However, as illustrated in this case report, an extensive and up-to-date MALDI-TOF MS database is necessary for providing an accurate identification.
Collapse
Affiliation(s)
- Stefanie Lefever
- Department of Microbiology, AZ Groeninge, Kortrijk, Belgium
- Department of Laboratory Medicine, University Hospital Leuven and KU Leuven, Leuven, Belgium
| | - Dorien Van Den Bossche
- Department of Microbiology, AZ Groeninge, Kortrijk, Belgium
- Department of Laboratory Medicine, University Hospital Leuven and KU Leuven, Leuven, Belgium
| | - Wouter Van Moerkercke
- Department of Gastroenterology, AZ Groeninge, Kortrijk, Belgium
- Department of Gastroenterology, University Hospital Leuven, Leuven, Belgium
| | - Mathieu D’Hondt
- Department of Abdominal Surgery, AZ Groeninge, Kortrijk, Belgium
| | | | - Mathieu Struyve
- Department of Gastroenterology, University Hospital Leuven, Leuven, Belgium
- Department of Gastroenterology, Hospital Oost-Limburg (ZOL), Genk, Belgium
| | | | | |
Collapse
|
121
|
Tang KY, Wang ZW, Wan QH, Fang SG. Metagenomics Reveals Seasonal Functional Adaptation of the Gut Microbiome to Host Feeding and Fasting in the Chinese Alligator. Front Microbiol 2019; 10:2409. [PMID: 31708889 PMCID: PMC6824212 DOI: 10.3389/fmicb.2019.02409] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/07/2019] [Indexed: 12/20/2022] Open
Abstract
As a natural hibernator, the Chinese alligator (Alligator sinensis) is an ideal and intriguing model to investigate changes in microbial community structure and function caused by hibernation. In this study, we used 16S rRNA profiling and metagenomic analysis to compare the composition, diversity, and functional capacity in the gut microbiome of hibernating vs. active Chinese alligators. Our results show that gut microbial communities undergo seasonal restructuring in response to seasonal cycles of feeding and fasting in the Chinese alligator, but this animal harbors a core gut microbial community primarily dominated by Proteobacteria, Fusobacteria, Bacteroidetes, and Firmicutes across the gut regions. During hibernation, there is an increase in the abundance of bacterial taxa (e.g., the genus Bacteroides) that can degrade host mucin glycans, which allows adaptation to winter fasting. This is accompanied by the enrichment of mucin oligosaccharide-degrading enzyme and carbohydrate-active enzyme families. In contrast, during the active phase (feeding), active Chinese alligators exhibit a carnivore gut microbiome dominated by Fusobacteria, and there is an increase in the relative abundance of bacteria (e.g., Cetobacterium somerae) with known proteolytic and amino acids-fermentating functions that improve host protein-rich food digestion efficiency. In addition, seasonal variations in the expression of β-defensins play a protective role in intestinal immunity. These findings provide insights into the functional adaptations of host-gut microbe symbioses to seasonal dietary shifts to maintain gut homeostasis and health, especially in extreme physiological states.
Collapse
Affiliation(s)
- Ke-Yi Tang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, State Conservation Centre for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Zhen-Wei Wang
- Changxing Yinjiabian Chinese Alligator Nature Reserve, Changxing, China
| | - Qiu-Hong Wan
- MOE Key Laboratory of Biosystems Homeostasis & Protection, State Conservation Centre for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sheng-Guo Fang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, State Conservation Centre for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
122
|
Stothart MR, Palme R, Newman AEM. It's what's on the inside that counts: stress physiology and the bacterial microbiome of a wild urban mammal. Proc Biol Sci 2019; 286:20192111. [PMID: 31640519 DOI: 10.1098/rspb.2019.2111] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The microbiome's capacity to shape the host phenotype and its mutability underlie theorization that the microbiome might facilitate host acclimation to rapid environmental change. However, when environmental change occurs, it is unclear whether resultant microbiome restructuring is proximately driven by this changing external environment or by the host's physiological response to this change. We leveraged urbanization to compare the ability of host environment (urban or forest) versus multi-scale biological measures of host hypothalamic-pituitary-adrenal (HPA) axis physiology (neutrophil : lymphocyte ratio, faecal glucocorticoid metabolites, hair cortisol) to explain variation in the eastern grey squirrel (Sciurus carolinensis) faecal microbiome. Urban and forest squirrels differed across all three of the interpretations of HPA axis activity we measured. Direct consideration of these physiological measures better explained greater phylogenetic turnover between squirrels than environment. This pattern was strongly driven by trade-offs between bacteria which specialize on metabolizing digesta versus host-derived nutrient sources. Drawing on ecological theory to explain patterns in intestinal bacterial communities, we conclude that although environmental change can affect the microbiome, it might primarily do so indirectly by altering host physiology. We demonstrate that the inclusion and careful consideration of dynamic, rather than fixed (e.g. sex), dimensions of host physiology are essential for the study of host-microbe symbioses at the micro-evolutionary scale.
Collapse
Affiliation(s)
- Mason R Stothart
- Department of Integrative Biology, College of Biological Sciences, University of Guelph, Guelph, Canada N1G 2W1.,Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada T2N 4Z6
| | - Rupert Palme
- Department of Biomedical Sciences/Unit of Physiology, Pathophysiology and Experimental Endocrinology, University of Veterinary Medicine Vienna, Vienna 1210, Austria
| | - Amy E M Newman
- Department of Integrative Biology, College of Biological Sciences, University of Guelph, Guelph, Canada N1G 2W1
| |
Collapse
|
123
|
Elucidation of a sialic acid metabolism pathway in mucus-foraging Ruminococcus gnavus unravels mechanisms of bacterial adaptation to the gut. Nat Microbiol 2019; 4:2393-2404. [PMID: 31636419 PMCID: PMC6881182 DOI: 10.1038/s41564-019-0590-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 09/12/2019] [Indexed: 12/26/2022]
Abstract
Sialic acid (Neu5Ac) is commonly found in terminal location of colonic mucins glycans where it is a much-coveted nutrient for gut bacteria including Ruminococcus gnavus. R. gnavus is part of the healthy gut microbiota in humans but shows a disproportionate representation in diseases. There is therefore a need in understanding the molecular mechanisms underpinning its adaptation to the gut. Previous in vitro work demonstrated that R. gnavus mucin glycan-foraging strategy is strain-dependent and associated with the expression of an intramolecular trans-sialidase releasing 2,7-anhydro-Neu5Ac instead of Neu5Ac from mucins. Here, we have unravelled the metabolism pathway of 2,7-anhydro-Neu5Ac in R. gnavus which is underpinned by the exquisite specificity of the sialic transporter for 2,7-anhydro-Neu5Ac, and by the action of an oxidoreductase converting 2,7-anhydro-Neu5Ac into Neu5Ac which then becomes substrate of a Neu5Ac-specific aldolase. Having generated a R. gnavus nan cluster deletion mutant that lost the ability to grow on sialylated substrates, we showed that in gnotobiotic mice colonised with R. gnavus wild-type and mutant strains, the fitness of the nan mutant was significantly impaired with a reduced ability to colonise the mucus layer. Overall, our study revealed a unique sialic acid pathway in bacteria, with significant implications for the spatial adaptation of mucin-foraging gut symbionts in health and disease.
Collapse
|
124
|
Fish oil supplementation to a high-fat diet improves both intestinal health and the systemic obese phenotype. J Nutr Biochem 2019; 72:108216. [DOI: 10.1016/j.jnutbio.2019.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/28/2019] [Accepted: 07/19/2019] [Indexed: 12/25/2022]
|
125
|
Tanaka M, Sanefuji M, Morokuma S, Yoden M, Momoda R, Sonomoto K, Ogawa M, Kato K, Nakayama J. The association between gut microbiota development and maturation of intestinal bile acid metabolism in the first 3 y of healthy Japanese infants. Gut Microbes 2019; 11:205-216. [PMID: 31550982 PMCID: PMC7053967 DOI: 10.1080/19490976.2019.1650997] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The gut microbial community greatly changes in early life, influencing infant health and subsequent host physiology, notably through its collective metabolism, including host-microbiota interplay of bile acid (BA) metabolism. However, little is known regarding how the development of the intestinal microbial community is associated with maturation of intestinal BA metabolism. To address this, we monitored the succession of gut bacterial community and its association with fecal BA profile in the first 3 y of ten healthy Japanese infants. The BA profiles were classified into four types, defined by high content of conjugated primary BA (Con type), unconjugated primary BA (chenodeoxycholic acid and cholic acid) (Pri type), ursodeoxycholic acid (Urs type), and deoxycholic and lithocholic acid (Sec type). Most subjects begun with Con type or Pri type profiles during lactation and eventually transited to Sec type through Urs type after the start of solid food intake. Con type and Pri type were associated with Enterobacteriaceae-dominant microbiota corresponding to the neonatal type or Bifidobacterium-dominant microbiota corresponding to lactation type, respectively. Urs type subjects were strongly associated with Ruminococcus gnavus colonization, mostly occurring between Pri type and Sec type. Sec type was associated with adult-type complex microbiota dominated by a variety of Firmicutes and Bacteroidetes species. Addressing the link of the common developmental passage of intestinal BA metabolism with infant's health and subsequent host physiology requires further study.
Collapse
Affiliation(s)
- Masaru Tanaka
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Masafumi Sanefuji
- Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiichi Morokuma
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Misako Yoden
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Rie Momoda
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Kenji Sonomoto
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Masanobu Ogawa
- Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiyoko Kato
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jiro Nakayama
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan,CONTACT Jiro Nakayama Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| |
Collapse
|
126
|
Jarett JK, Carlson A, Rossoni Serao M, Strickland J, Serfilippi L, Ganz HH. Diets with and without edible cricket support a similar level of diversity in the gut microbiome of dogs. PeerJ 2019; 7:e7661. [PMID: 31565574 PMCID: PMC6743483 DOI: 10.7717/peerj.7661] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome plays an important role in the health of dogs. Both beneficial microbes and overall diversity can be modulated by diet. Fermentable sources of fiber in particular often increase the abundance of beneficial microbes. Banded crickets (Gryllodes sigillatus) contain the fermentable polysaccharides chitin and chitosan. In addition, crickets are an environmentally sustainable protein source. Considering crickets as a potential source of both novel protein and novel fiber for dogs, four diets ranging from 0% to 24% cricket content were fed to determine their effects on healthy dogs’ (n = 32) gut microbiomes. Fecal samples were collected serially at 0, 14, and 29 days, and processed using high-throughput sequencing of 16S rRNA gene PCR amplicons. Microbiomes were generally very similar across all diets at both the phylum and genus level, and alpha and beta diversities did not differ between the various diets at 29 days. A total of 12 ASVs (amplicon sequence variants) from nine genera significantly changed in abundance following the addition of cricket, often in a dose-response fashion with increasing amounts of cricket. A net increase was observed in Catenibacterium, Lachnospiraceae [Ruminococcus], and Faecalitalea, whereas Bacteroides, Faecalibacterium, Lachnospiracaeae NK4A136 group and others decreased in abundance. Similar changes in Catenibacterium and Bacteroides have been associated with gut health benefits in other studies. However, the total magnitude of all changes was small and only a few specific taxa changed in abundance. Overall, we found that diets containing cricket supported the same level of gut microbiome diversity as a standard healthy balanced diet. These results support crickets as a potential healthy, novel food ingredient for dogs.
Collapse
|
127
|
Monk JM, Wu W, Lepp D, Wellings HR, Hutchinson AL, Liddle DM, Graf D, Pauls KP, Robinson LE, Power KA. Navy bean supplemented high-fat diet improves intestinal health, epithelial barrier integrity and critical aspects of the obese inflammatory phenotype. J Nutr Biochem 2019; 70:91-104. [DOI: 10.1016/j.jnutbio.2019.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/03/2019] [Accepted: 04/25/2019] [Indexed: 02/07/2023]
|
128
|
Van Herreweghen F, De Paepe K, Roume H, Kerckhof FM, Van de Wiele T. Mucin degradation niche as a driver of microbiome composition and Akkermansia muciniphila abundance in a dynamic gut model is donor independent. FEMS Microbiol Ecol 2019; 94:5101425. [PMID: 30239657 DOI: 10.1093/femsec/fiy186] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 09/14/2018] [Indexed: 12/21/2022] Open
Abstract
Akkermansia muciniphila, an abundant mucin degrading intestinal bacterium, has been correlated with human health in various studies. The in vitro SHIME model was used to reach a mechanistic understanding of A. muciniphila's colonization preferences and its response to environmental parameters such as colon pH and mucins. These insight can help to identify the optimal conditions for successful in vivo application. After a period of mucin deprivation, we found that mucin supplementation resulted in significantly different microbial communities, with more Akkermansia, Bacteroides and Ruminococcus. Mucin treatment accounted for 26% of the observed variation in the microbial community at OTU level (P = 0.001), whereas the donor effect was limited (8%) (P = 0.035), indicating mucins to constitute an important ecological niche shaping the microbiota composition. The effect of colonic pH had a less profound impact on the microbiome with both pH and donor origin explaining around 10% of the variability in the dataset. Yet, higher simulated colonic pH had a positive impact on Akkermansia abundance while short chain fatty acid analysis displayed a preference for propionate production with higher colonic pH. Our results show that mucins as nutritional resource are a more important modulator of the gut microbiome than colon pH as environmental factor.
Collapse
Affiliation(s)
| | - Kim De Paepe
- Center for Microbial Ecology and Technology (CMET), Ghent University, Ghent, Belgium
| | - Hugo Roume
- MetaGenoPolis Unit,National Institute for Agricultural Research (INRA), Allée de Vilvert, 78352, Jouy-en-Josas, Île-de-France, France
| | | | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Ghent University, Ghent, Belgium
| |
Collapse
|
129
|
Singh RP. Glycan utilisation system in Bacteroides and Bifidobacteria and their roles in gut stability and health. Appl Microbiol Biotechnol 2019; 103:7287-7315. [PMID: 31332487 DOI: 10.1007/s00253-019-10012-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
Gut residential hundred trillion microbial cells are indispensable for maintaining gut homeostasis and impact on host physiology, development and immune systems. Many of them have displayed excellence in utilising dietary- and host-derived complex glycans and are producing useful postbiotics including short-chain fatty acids to primarily fuel different organs of the host. Therefore, employing individual microbiota is nowadays becoming a propitious target in biomedical for improving gut dysbiosis conditions of the host. Among other gut microbial communities, Bacteroides and Bifidobacteria are coevolved to utilise diverse ranges of diet- and host-derived glycans through harmonising distinct glycan utilisation systems. These gut symbionts frequently share digested oligosaccharides, carbohydrate-active enzymes and fermentable intermediate molecules for sustaining gut microbial symbiosis and improving fitness of own or other communities. Genomics approaches have provided unprecedented insights into these functions, but their precise mechanisms of action have poorly known. Sympathetic glycan-utilising strategy of each gut commensal will provide overview of mechanistic dynamic nature of the gut environment and will then assist in applying aptly personalised nutritional therapy. Thus, the review critically summarises cutting edge understanding of major plant- and host-derived glycan-utilising systems of Bacteroides and Bifidobacteria. Their evolutionary adaptation to gut environment and roles of postbiotics in human health are also highlighted.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS, Nagar, Punjab, 140306, India.
| |
Collapse
|
130
|
Jakobsen LMA, Sundekilde UK, Andersen HJ, Nielsen DS, Bertram HC. Lactose and Bovine Milk Oligosaccharides Synergistically Stimulate B. longum subsp. longum Growth in a Simplified Model of the Infant Gut Microbiome. J Proteome Res 2019; 18:3086-3098. [PMID: 31264424 DOI: 10.1021/acs.jproteome.9b00211] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Increasing awareness of the importance of a healthy Bifidobacterium-rich microbiome has led to a need for more knowledge on how different prebiotic carbohydrates specifically impact the infant microbiome, especially as a community instead of single bacterial targets. In this study, we combined proton nuclear magnetic resonance (1H NMR) metabolomics and molecular biology methods for quantification of bacteria to compare the prebiotic effect of bovine milk oligosaccharides (BMO) and synthetic galacto oligosaccharides (GOS) using mono- and cocultures of eight major bacteria related to a healthy infant microbiome. The results revealed that BMO treatments supported growth of Bifidobacterium longum subsp. longum and Parabacteroides distasonis, while at the same time growth of Clostridium perfringens and Escherichia coli was inhibited. In addition, there was a synergistic effect of combining lactose and BMO in regards to reducing C. perfringens, maintaining stable numbers of P. distasonis and simultaneously increasing numbers of the beneficial B. longum subsp. longum. These results indicate that the oligosaccharide composition plays a vital role in shaping the developing microbiota.
Collapse
Affiliation(s)
- Louise M A Jakobsen
- Department of Food Science , Aarhus University , Kirstinebjergvej 10 , Årslev 5792 , Denmark
| | - Ulrik K Sundekilde
- Department of Food Science , Aarhus University , Kirstinebjergvej 10 , Årslev 5792 , Denmark
| | - Henrik J Andersen
- Arla Food Ingredients Group P/S , Sønderhøj 10 , Viby J 8260 , Denmark
| | - Dennis S Nielsen
- Department of Food Science , University of Copenhagen , Rolighedsvej 30 , Frederiksberg C 1958 , Denmark
| | - Hanne C Bertram
- Department of Food Science , Aarhus University , Kirstinebjergvej 10 , Årslev 5792 , Denmark
| |
Collapse
|
131
|
McKeen S, Young W, Fraser K, Roy NC, McNabb WC. Glycan Utilisation and Function in the Microbiome of Weaning Infants. Microorganisms 2019; 7:microorganisms7070190. [PMID: 31277402 PMCID: PMC6681113 DOI: 10.3390/microorganisms7070190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Glycans are present exogenously in the diet, expressed and secreted endogenously by host cells, and produced by microbes. All of these processes result in them being available to the gut microbiome, firmly placing glycans at the interface of diet–microbe–host interactions. The most dramatic shift in dietary sources of glycans occurs during the transition from the milk-based neonatal diet to the diverse omnivorous adult diet, and this has profound effects on the composition of the gut microbiome, gene expression by microbes and host cells, mucin composition, and immune development from innate towards adaptive responses. Understanding the glycan-mediated interactions occurring during this transitional window may inform dietary recommendations to support gut and immune development during a vulnerable age. This review aims to summarise the current state of knowledge on dietary glycan mediated changes that may occur in the infant gut microbiome and immune system during weaning.
Collapse
Affiliation(s)
- Starin McKeen
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Wayne Young
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Karl Fraser
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Nicole C Roy
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand.
| |
Collapse
|
132
|
Azzouz D, Omarbekova A, Heguy A, Schwudke D, Gisch N, Rovin BH, Caricchio R, Buyon JP, Alekseyenko AV, Silverman GJ. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal. Ann Rheum Dis 2019; 78:947-956. [PMID: 30782585 PMCID: PMC6585303 DOI: 10.1136/annrheumdis-2018-214856] [Citation(s) in RCA: 265] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND/PURPOSE To search for a transmissible agent involved in lupus pathogenesis, we investigated the faecal microbiota of patients with systemic lupus erythematosus (SLE) for candidate pathobiont(s) and evaluated them for special relationships with host immunity. METHODS In a cross-sectional discovery cohort, matched blood and faecal samples from 61 female patients with SLE were obtained. Faecal 16 S rRNA analyses were performed, and sera profiled for antibacterial and autoantibody responses, with findings validated in two independent lupus cohorts. RESULTS Compared with controls, the microbiome in patients with SLE showed decreased species richness diversity, with reductions in taxonomic complexity most pronounced in those with high SLE disease activity index (SLEDAI). Notably, patients with SLE had an overall 5-fold greater representation of Ruminococcus gnavus (RG) of the Lachnospiraceae family, and individual communities also displayed reciprocal contractions of a species with putative protective properties. Gut RG abundance correlated with serum antibodies to only 1/8 RG strains tested. Anti-RG antibodies correlated directly with SLEDAI score and antinative DNA levels, but inversely with C3 and C4. These antibodies were primarily against antigen(s) in an RG strain-restricted pool of cell wall lipoglycans. Novel structural features of these purified lipoglycans were characterised by mass spectrometry and NMR. Highest levels of serum anti-RG strain-restricted antibodies were detected in those with active nephritis (including Class III and IV) in the discovery cohort, with findings validated in two independent cohorts. CONCLUSION These findings suggest a novel paradigm in which specific strains of a gut commensal may contribute to the immune pathogenesis of lupus nephritis.
Collapse
Affiliation(s)
- Doua Azzouz
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Aidana Omarbekova
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Adriana Heguy
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Dominik Schwudke
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Borstel, Germany
| | - Nicolas Gisch
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Borstel, Germany
| | | | | | - Jill P Buyon
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Alexander V Alekseyenko
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gregg J Silverman
- Department of Medicine, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
133
|
Wang J, Feng W, Zhang S, Chen L, Sheng Y, Tang F, He J, Xu X, Ao H, Peng C. Ameliorative effect of Atractylodes macrocephala essential oil combined with Panax ginseng total saponins on 5-fluorouracil induced diarrhea is associated with gut microbial modulation. JOURNAL OF ETHNOPHARMACOLOGY 2019; 238:111887. [PMID: 31004726 DOI: 10.1016/j.jep.2019.111887] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 03/21/2019] [Accepted: 04/12/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) holds that deficiency of spleen-Qi is the major pathogenesis of chemotherapy-induced diarrhea (CID). Herb pair of Atractylodes macrocephala Koidz. (AM) and Panax ginseng C. A. Mey. (PG) has good effects of supplementing Qi and strengthening spleen. AIM OF THE STUDY To investigate therapeutic effects and mechanism of Atractylodes macrocephala essential oil (AMO) and Panax ginseng total saponins (PGS) alone and in combination (AP) on 5-fluorouracil (5-FU) chemotherapy induced diarrhea in mice. MATERIALS AND METHODS The mice were administered with AMO, PGS and AP respectively for 11 days, and intraperitoneally injected with 5-FU for 6 days since the 3rd day of the experiment. During the experiment, the body weights and diarrhea scores of mice were recorded daily. Thymus and spleen indexes were calculated after sacrifice of the mice. Pathological changes in ileum and colonic tissues were examined by hematoxylin-eosin (HE) staining. And the content levels of intestinal inflammatory cytokines were measured by enzyme-linked immmunosorbent assays (ELISA). 16S rDNA Amplicon Sequencing was used to analyze and interpret the gut microbiota of fecal samples. RESULTS AP significantly inhibited body weights loss, diarrhea, reductions of thymus and spleen indexes, and pathological changes of ileums and colons induced by 5-FU. Neither AMO nor PGS alone significantly improved above-mentioned abnormalities. Besides, AP could significantly suppressed the 5-FU-mediated increases of the intestinal inflammatory cytokines (TNF-α, IFN-γ, IL-6, IL-1β and IL-17), while AMO or PGS only inhibited some of them after 5-FU chemotherapy. Gut microbiota analysis indicated that 5-FU induced overall structural changes of gut microbiota were reversed after AP treatment. Additionally, AP significantly modulated the abundances of different phyla similar to normal values, and restored the ratios of Firmicutes/Bacteroidetes (F/B). At genus level, AP treatment dramatically decreased potential pathogens like Bacteroides, Ruminococcus, Anaerotruncus and Desulfovibrio. AP also antagonized the abnormal effects of AMO and PGS alone on certain genera like Blautia, Parabacteroides and Lactobacillus. Neither AMO nor PGS alone inhibited changes of gut microbial structure caused by 5-FU. CONCLUSIONS AP, combination of AMO and PGS, not AMO or PGS alone, significantly ameliorated diarrhea, inhibited intestinal pathology, and modulated gut microbial structure in 5-FU induced mice. AP also antagonized abnormal effects of AMO or PGS on certain genera. The results illustrated that gut microbiota was involved in the combined effects of AP on 5-FU induced diarrhea.
Collapse
Affiliation(s)
- Jing Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Wuwen Feng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Shiyang Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Lu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yongcheng Sheng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Fei Tang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Junlin He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xin Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
134
|
Richards P, Fothergill J, Bernardeau M, Wigley P. Development of the Caecal Microbiota in Three Broiler Breeds. Front Vet Sci 2019; 6:201. [PMID: 31294039 PMCID: PMC6603203 DOI: 10.3389/fvets.2019.00201] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022] Open
Abstract
The development of the caecal microbiota plays a role in the metabolism and immune competence of chickens. A detailed understanding of normal succession in the caecal microbiota can inform the use of probiotics and other interventions to optimize the caecal microbiota. The development of the microbiota in caecal mucus and lumen samples from three breeds of broiler chicken (Cobb 500, n = 36; Hubbard JA87, n = 38; and Ross 308, n = 36) was observed between 0 and 42 days post hatch. Chicks were housed in the same room of a climate-controlled, biosecure chicken housing unit. Between 0 and 14 days post hatch, chicks were kept in brooder pens ensuring a mixture of breeds in each brooder. From 22 days post hatch, chicks were removed from the brooders and kept in the same room. DNA was extracted from a pooled sample of caecal mucus and luminal contents from five birds of each breed at 0, 3, 7, 14, 21, 28, and 42 days post hatch. High-throughput Illumina sequencing was performed for the V4 hypervariable region of the 16S rRNA gene. The early caecal microbiota was characterized by poor diversity and dominance by one or two bacterial species. Early colonizers of the caecum included Bifidobacteriaceae, Lachnospiraceae, Bacteroidaceae and Burkholderiaceae with some amplicon sequence variants (ASVs) assigned to Ruminococcaceae. Later colonizers of the caecal microbiota were most apparent from 14 d.p.h and included Ruminococcaceae, Clostridiales vadin BB60 group, Christensenellaceae and Bacillaceae. The caecal microbiota continued to change until 42 d.p.h when the microbiota was characterized by a high abundance of Bacteroidaceae, Lachnospiraceae and Ruminococcaceae. The lumen microbiota was significantly different to the mucus with some ASVs assigned to Lachnospiraceae, Ruminococcaceae, Christensenellaceae and Bacillaceae showing increased abundance in the mucus. ASVs assigned to Bacteroidaceae, Lactobacillaceae and Burkholderiaceae showed a preference for the lumen. Analysis of five caecal mucus samples from each breed at 42 days post hatch showed differences in microbiota composition between Ross and Cobb as well as between Ross and Hubbard. Since performance data was not collected no functional inferences as to the significance of this finding can be made.
Collapse
Affiliation(s)
- Peter Richards
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Jo Fothergill
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Marion Bernardeau
- DuPont Industrial Biosciences, Genencor International BV, Leiden, Netherlands
| | - Paul Wigley
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
135
|
Systems Pharmacology and Microbiome Dissection of Shen Ling Bai Zhu San Reveal Multiscale Treatment Strategy for IBD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8194804. [PMID: 31341536 PMCID: PMC6612409 DOI: 10.1155/2019/8194804] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/19/2019] [Accepted: 05/30/2019] [Indexed: 02/08/2023]
Abstract
Generally, inflammatory bowel disease (IBD) can be caused by psychology, genes, environment, and gut microbiota. Therefore, IBD therapy should be improved to utilize multiple strategies. Shen Ling Bai Zhu San (SLBZS) adheres to the aim of combating complex diseases from an integrative and holistic perspective, which is effective for IBD therapy. Herein, a systems pharmacology and microbiota approach was developed for these molecular mechanisms exemplified by SLBZS. First, by systematic absorption-distribution-metabolism-excretion (ADME) analysis, potential active compounds and their corresponding direct targets were retrieved. Then, the network relationships among the active compounds, targets, and disease were built to deduce the pharmacological actions of the drug. Finally, an “IBD pathway” consisting of several regulatory modules was proposed to dissect the therapeutic effects of SLBZS. In addition, the effects of SLBZS on gut microbiota were evaluated through analysis of the V3-V4 region and multivariate statistical methods. SLBZS significantly shifted the gut microbiota structure in a rat model. Taken together, we found that SLBZS has multidimensionality in the regulation of IBD-related physiological processes, which provides new sights into herbal medicine for the treatment of IBD.
Collapse
|
136
|
Grabinger T, Glaus Garzon JF, Hausmann M, Geirnaert A, Lacroix C, Hennet T. Alleviation of Intestinal Inflammation by Oral Supplementation With 2-Fucosyllactose in Mice. Front Microbiol 2019; 10:1385. [PMID: 31275292 PMCID: PMC6593069 DOI: 10.3389/fmicb.2019.01385] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022] Open
Abstract
Milk oligosaccharides exert a prebiotic action that contributes to the development of the infant gut microbiota during lactation. Given that milk oligosaccharides remain intact after passage through stomach and small intestine, they can potentially influence the composition of the gut microbiota when ingested as dietary supplements after weaning. To address the regulatory effects of specific oligosaccharides in colitis linked to the microbiota composition, we have supplemented interleukin-10 null (Il10 -/-) mice with four fucosylated and sialylated oligosaccharides. We found that oral supplementation with 2-fucosyllactose significantly decreased the severity of colitis as displayed by reduced inflammatory marker expression, histological and diarrhea scores, an increased epithelial integrity and less pronounced colon shortening. Oral supplementation with 2-fucosyllactose led to a marked expansion of the commensal Ruminococcus gnavus, which was accompanied by an enhanced cecal concentration of propionate. Decreased activation of immune cells by R. gnavus was confirmed by reconstitution of antibiotic-treated Il10 -/- mice and by stimulation of dendritic cells in vitro. This study demonstrates that post-weaning administration of specific oligosaccharides can shift the composition of the gut microbiota to lessen chronic inflammation as observed in Il10 -/- mice. The expansion of R. gnavus sets a positive microbial environment at the cost of pro-inflammatory Gram-negative bacteria, thereby lowering intestinal inflammation.
Collapse
Affiliation(s)
- Thomas Grabinger
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich - University of Zurich, Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
137
|
Engevik MA, Luk B, Chang-Graham AL, Hall A, Herrmann B, Ruan W, Endres BT, Shi Z, Garey KW, Hyser JM, Versalovic J. Bifidobacterium dentium Fortifies the Intestinal Mucus Layer via Autophagy and Calcium Signaling Pathways. mBio 2019; 10:e01087-19. [PMID: 31213556 PMCID: PMC6581858 DOI: 10.1128/mbio.01087-19] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/11/2019] [Indexed: 02/07/2023] Open
Abstract
Much remains unknown about how the intestinal microbiome interfaces with the protective intestinal mucus layer. Bifidobacterium species colonize the intestinal mucus layer and can modulate mucus production by goblet cells. However, select Bifidobacterium strains can also degrade protective glycans on mucin proteins. We hypothesized that the human-derived species Bifidobacterium dentium would increase intestinal mucus synthesis and expulsion, without extensive degradation of mucin glycans. In silico data revealed that B. dentium lacked the enzymes necessary to extensively degrade mucin glycans. This finding was confirmed by demonstrating that B. dentium could not use naive mucin glycans as primary carbon sources in vitro To examine B. dentium mucus modulation in vivo, Swiss Webster germfree mice were monoassociated with live or heat-killed B. dentium Live B. dentium-monoassociated mice exhibited increased colonic expression of goblet cell markers Krüppel-like factor 4 (Klf4), Trefoil factor 3 (Tff3), Relm-β, Muc2, and several glycosyltransferases compared to both heat-killed B. dentium and germfree counterparts. Likewise, live B. dentium-monoassociated colon had increased acidic mucin-filled goblet cells, as denoted by Periodic Acid-Schiff-Alcian Blue (PAS-AB) staining and MUC2 immunostaining. In vitro, B. dentium-secreted products, including acetate, were able to increase MUC2 levels in T84 cells. We also identified that B. dentium-secreted products, such as γ-aminobutyric acid (GABA), stimulated autophagy-mediated calcium signaling and MUC2 release. This work illustrates that B. dentium is capable of enhancing the intestinal mucus layer and goblet cell function via upregulation of gene expression and autophagy signaling pathways, with a net increase in mucin production.IMPORTANCE Microbe-host interactions in the intestine occur along the mucus-covered epithelium. In the gastrointestinal tract, mucus is composed of glycan-covered proteins, or mucins, which are secreted by goblet cells to form a protective gel-like structure above the epithelium. Low levels of mucin or alterations in mucin glycans are associated with inflammation and colitis in mice and humans. Although current literature links microbes to the modulation of goblet cells and mucins, the molecular pathways involved are not yet fully understood. Using a combination of gnotobiotic mice and mucus-secreting cell lines, we have identified a human-derived microbe, Bifidobacterium dentium, which adheres to intestinal mucus and secretes metabolites that upregulate the major mucin MUC2 and modulate goblet cell function. Unlike other Bifidobacterium species, B. dentium does not extensively degrade mucin glycans and cannot grow on mucin alone. This work points to the potential of using B. dentium and similar mucin-friendly microbes as therapeutic agents for intestinal disorders with disruptions in the mucus barrier.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Berkley Luk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Alexandra L Chang-Graham
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Anne Hall
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Beatrice Herrmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Wenly Ruan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Bradley T Endres
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Kevin W Garey
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
138
|
Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn's disease, produces an inflammatory polysaccharide. Proc Natl Acad Sci U S A 2019; 116:12672-12677. [PMID: 31182571 PMCID: PMC6601261 DOI: 10.1073/pnas.1904099116] [Citation(s) in RCA: 472] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The bacteria that live within the human gut play crucial roles in regulating our primary metabolism, protecting us from pathogens, and developing our immune system. Imbalances in bacterial community structure have been implicated in many diseases, such as Crohn’s disease, an inflammatory bowel disease. We found and characterized an inflammatory polysaccharide produced by the gut bacterium Ruminococcus gnavus, populations of which bloom during flares of symptoms in patients with Crohn’s disease. This molecule induces the production of inflammatory cytokines like TNFα by dendritic cells and may contribute to the association between R. gnavus and Crohn’s disease. This work establishes a plausible molecular mechanism that may explain the association between a member of the gut microbiome and an inflammatory disease. A substantial and increasing number of human diseases are associated with changes in the gut microbiota, and discovering the molecules and mechanisms underlying these associations represents a major research goal. Multiple studies associate Ruminococcus gnavus, a prevalent gut microbe, with Crohn’s disease, a major type of inflammatory bowel disease. We have found that R. gnavus synthesizes and secretes a complex glucorhamnan polysaccharide with a rhamnose backbone and glucose sidechains. Chemical and spectroscopic studies indicated that the glucorhamnan was largely a repeating unit of five sugars with a linear backbone formed from three rhamnose units and a short sidechain composed of two glucose units. The rhamnose backbone is made from 1,2- and 1,3-linked rhamnose units, and the sidechain has a terminal glucose linked to a 1,6-glucose. This glucorhamnan potently induces inflammatory cytokine (TNFα) secretion by dendritic cells, and TNFα secretion is dependent on toll-like receptor 4 (TLR4). We also identify a putative biosynthetic gene cluster for this molecule, which has the four biosynthetic genes needed to convert glucose to rhamnose and the five glycosyl transferases needed to build the repeating pentasaccharide unit of the inflammatory glucorhamnan.
Collapse
|
139
|
Reese AT, Carmody RN. Thinking Outside the Cereal Box: Noncarbohydrate Routes for Dietary Manipulation of the Gut Microbiota. Appl Environ Microbiol 2019; 85:e02246-18. [PMID: 30504210 PMCID: PMC6498178 DOI: 10.1128/aem.02246-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota is a diverse and dynamic ecological community that is increasingly recognized to play important roles in host metabolic, immunological, and behavioral functioning. As such, identifying new routes for manipulating the microbiota may provide valuable additional methods for improving host health. Dietary manipulations and prebiotic supplementation are active targets of research for altering the microbiota, but to date, this work has disproportionately focused on carbohydrates. However, many other resources can limit or shape microbial growth. Here, we provide a brief overview of the resource landscape in the mammalian gut and review relevant literature documenting associations between noncarbohydrate nutrients and the composition of the gut microbiota. To spur future work and accelerate translational applications, we propose that researchers take new approaches for studying the effects of diet on gut microbial communities, including more-careful consideration of media for in vitro experiments, measurement of absolute as well as relative abundances, concerted efforts to articulate how physiology may differ between humans and the animal models used in translational studies, and leveraging natural variation for additional insights. Finally, we close with a discussion of how to determine when or where to employ these potential dietary levers for manipulating the microbiota.
Collapse
Affiliation(s)
- Aspen T Reese
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
- Society of Fellows, Harvard University, Cambridge, Massachusetts, USA
| | - Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
140
|
Tauzin AS, Bruel L, Laville E, Nicoletti C, Navarro D, Henrissat B, Perrier J, Potocki-Veronese G, Giardina T, Lafond M. Sucrose 6 F-phosphate phosphorylase: a novel insight in the human gut microbiome. Microb Genom 2019; 5. [PMID: 30913025 PMCID: PMC6521584 DOI: 10.1099/mgen.0.000253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The human gut microbiome plays an essential role in maintaining human health including in degradation of dietary fibres and carbohydrates further used as nutrients by both the host and the gut bacteria. Previously, we identified a polysaccharide utilization loci (PUL) involved in sucrose and raffinose family oligosaccharide (RFO) metabolism from one of the most common Firmicutes present in individuals, Ruminococcus gnavus E1. One of the enzymes encoded by this PUL was annotated as a putative sucrose phosphate phosphorylase (RgSPP). In the present study, we have in-depth characterized the heterologously expressed RgSPP as sucrose 6F-phosphate phosphorylase (SPP), expanding our knowledge of the glycoside hydrolase GH13_18 subfamily. Specifically, the enzymatic characterization showed a selective activity on sucrose 6F-phosphate (S6FP) acting both in phosphorolysis releasing alpha-d-glucose-1-phosphate (G1P) and alpha-d-fructose-6-phosphate (F6P), and in reverse phosphorolysis from G1P and F6P to S6FP. Interestingly, such a SPP activity had never been observed in gut bacteria before. In addition, a phylogenetic and synteny analysis showed a clustering and a strictly conserved PUL organization specific to gut bacteria. However, a wide prevalence and abundance study with a human metagenomic library showed a correlation between SPP activity and the geographical origin of the individuals and, thus, most likely linked to diet. Rgspp gene overexpression has been observed in mice fed with a high-fat diet suggesting, as observed for humans, that intestine lipid and carbohydrate microbial metabolisms are intertwined. Finally, based on the genomic environment analysis, in vitro and in vivo studies, results provide new insights into the gut microbiota catabolism of sucrose, RFOs and S6FP.
Collapse
Affiliation(s)
- Alexandra S Tauzin
- 1Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France.,2LISBP, CNRS, INRA, INSAT, Université de Toulouse, F-31400 Toulouse, France.,‡Present address: LISBP, CNRS, INRA, INSAT, Université de Toulouse, F-31400 Toulouse, France
| | - Laetitia Bruel
- 1Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Elisabeth Laville
- 2LISBP, CNRS, INRA, INSAT, Université de Toulouse, F-31400 Toulouse, France
| | | | - David Navarro
- 3INRA, Aix-Marseille Université, UMR1163, Biodiversité et Biotechnologie Fongiques, PolyTech, F-13009, Marseille, France
| | - Bernard Henrissat
- 4Architecture et Fonction des Macromolécules Biologiques, CNRS, Aix-Marseille Université, F-13288 Marseille, France.,5Department of Biological Sciences, King Abdulaziz University, 23218 Jeddah, Saudi Arabia
| | - Josette Perrier
- 1Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France
| | | | - Thierry Giardina
- 1Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Mickael Lafond
- 1Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France
| |
Collapse
|
141
|
Schroeder BO. Fight them or feed them: how the intestinal mucus layer manages the gut microbiota. Gastroenterol Rep (Oxf) 2019; 7:3-12. [PMID: 30792861 PMCID: PMC6375348 DOI: 10.1093/gastro/goy052] [Citation(s) in RCA: 290] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
The intestinal tract is inhabited by a tremendous number of microorganisms, termed the gut microbiota. These microorganisms live in a mutualistic relationship with their host and assist in the degradation of complex carbohydrates. Although the gut microbiota is generally considered beneficial, the vast number of microbial cells also form a permanent threat to the host. Thus, the intestinal epithelium is covered with a dense layer of mucus to prevent translocation of the gut microbiota into underlying tissues. Intestinal mucus is an organized glycoprotein network with a host-specific glycan structure. While the mucus layer has long been considered a passive, host-designed barrier, recent studies showed that maturation and function of the mucus layer are strongly influenced by the gut microbiota. In return, the glycan repertoire of mucins can select for distinct mucosa-associated bacteria that are able to bind or degrade specific mucin glycans as a nutrient source. Because the intestinal mucus layer is at the crucial interface between host and microbes, its breakdown leads to gut bacterial encroachment that can eventually cause inflammation and infection. Accordingly, a dysfunctional mucus layer has been observed in colitis in mice and humans. Moreover, the increased consumption of a low-fiber Western-style diet in our modern society has recently been demonstrated to cause bacteria-mediated defects of the intestinal mucus layer. Here, I will review current knowledge on the interaction between gut bacteria and the intestinal mucus layer in health and disease. Understanding the molecular details of this host–microbe interaction may contribute to the development of novel treatment options for diseases involving a dysfunctional mucus layer, such as ulcerative colitis.
Collapse
Affiliation(s)
- Bjoern O Schroeder
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Bruna Stråket 16, University of Gothenburg, SE 413 45 Gothenburg, Sweden
| |
Collapse
|
142
|
Ndeh D, Gilbert HJ. Biochemistry of complex glycan depolymerisation by the human gut microbiota. FEMS Microbiol Rev 2018; 42:146-164. [PMID: 29325042 DOI: 10.1093/femsre/fuy002] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/06/2018] [Indexed: 12/21/2022] Open
Abstract
The human gut microbiota (HGM) makes an important contribution to health and disease. It is a complex microbial community of trillions of microbes with a majority of its members represented within two phyla, the Bacteroidetes and Firmicutes, although it also contains species of Actinobacteria and Proteobacteria. Reflecting its importance, the HGM is sometimes referred to as an 'organ' as it performs functions analogous to systemic tissues within the human host. The major nutrients available to the HGM are host and dietary complex carbohydrates. To utilise these nutrient sources, the HGM has developed elaborate, variable and sophisticated systems for the sensing, capture and utilisation of these glycans. Understanding nutrient acquisition by the HGM can thus provide mechanistic insights into the dynamics of this ecosystem, and how it impacts human health. Dietary nutrient sources include a wide variety of simple and complex plant and animal-derived glycans most of which are not degraded by enzymes in the digestive tract of the host. Here we review how various adaptive mechanisms that operate across the major phyla of the HGM contribute to glycan utilisation, focusing on the most complex carbohydrates presented to this ecosystem.
Collapse
Affiliation(s)
- Didier Ndeh
- Institute for Cell and Molecular Biosciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Harry J Gilbert
- Institute for Cell and Molecular Biosciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
143
|
Crost EH, Le Gall G, Laverde-Gomez JA, Mukhopadhya I, Flint HJ, Juge N. Mechanistic Insights Into the Cross-Feeding of Ruminococcus gnavus and Ruminococcus bromii on Host and Dietary Carbohydrates. Front Microbiol 2018; 9:2558. [PMID: 30455672 PMCID: PMC6231298 DOI: 10.3389/fmicb.2018.02558] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022] Open
Abstract
Dietary and host glycans shape the composition of the human gut microbiota with keystone carbohydrate-degrading species playing a critical role in maintaining the structure and function of gut microbial communities. Here, we focused on two major human gut symbionts, the mucin-degrader Ruminococcus gnavus ATCC 29149, and R. bromii L2-63, a keystone species for the degradation of resistant starch (RS) in human colon. Using anaerobic individual and co-cultures of R. bromii and R. gnavus grown on mucin or starch as sole carbon source, we showed that starch degradation by R. bromii supported the growth of R. gnavus whereas R. bromii did not benefit from mucin degradation by R. gnavus. Further we analyzed the growth (quantitative PCR), metabolite production (1H NMR analysis), and bacterial transcriptional response (RNA-Seq) of R. bromii cultured with RS or soluble starch (SS) in the presence or absence of R. gnavus. In co-culture fermentations on starch, 1H NMR analysis showed that R. gnavus benefits from transient glucose and malto-oligosaccharides released by R. bromii upon starch degradation, producing acetate, formate, and lactate as main fermentation end-products. Differential expression analysis (DESeq 2) on starch (SS and RS) showed that the presence of R. bromii induced changes in R. gnavus transcriptional response of genes encoding several maltose transporters and enzymes involved in its metabolism such as maltose phosphorylase, in line with the ability of R. gnavus to utilize R. bromii starch degradation products. In the RS co-culture, R. bromii showed a significant increase in the induction of tryptophan (Trp) biosynthesis genes and a decrease of vitamin B12 (VitB12)-dependent methionine biosynthesis as compared to the mono-culture, suggesting that Trp and VitB12 availability become limited in the presence of R. gnavus. Together this study showed a direct competition between R. bromii and R. gnavus on RS, suggesting that in vivo, the R. gnavus population inhabiting the mucus niche may be modulated by the supply of non-digestible carbohydrates reaching the colon such as RS.
Collapse
Affiliation(s)
- Emmanuelle H Crost
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich, United Kingdom
| | - Gwenaelle Le Gall
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich, United Kingdom
| | - Jenny A Laverde-Gomez
- Gut Health Group, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Indrani Mukhopadhya
- Gut Health Group, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Harry J Flint
- Gut Health Group, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich Research Park, Norwich, United Kingdom
| |
Collapse
|
144
|
Direct-fed microbial supplementation influences the bacteria community composition of the gastrointestinal tract of pre- and post-weaned calves. Sci Rep 2018; 8:14147. [PMID: 30237565 PMCID: PMC6148029 DOI: 10.1038/s41598-018-32375-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023] Open
Abstract
This study investigated the effect of supplementing the diet of calves with two direct fed microbials (DFMs) (Saccharomyces cerevisiae boulardii CNCM I-1079 (SCB) and Lactobacillus acidophilus BT1386 (LA)), and an antibiotic growth promoter (ATB). Thirty-two dairy calves were fed a control diet (CTL) supplemented with SCB or LA or ATB for 96 days. On day 33 (pre-weaning, n = 16) and day 96 (post-weaning, n = 16), digesta from the rumen, ileum, and colon, and mucosa from the ileum and colon were collected. The bacterial diversity and composition of the gastrointestinal tract (GIT) of pre- and post-weaned calves were characterized by sequencing the V3-V4 region of the bacterial 16S rRNA gene. The DFMs had significant impact on bacteria community structure with most changes associated with treatment occurring in the pre-weaning period and mostly in the ileum but less impact on bacteria diversity. Both SCB and LA significantly reduced the potential pathogenic bacteria genera, Streptococcus and Tyzzerella_4 (FDR ≤ 8.49E-06) and increased the beneficial bacteria, Fibrobacter (FDR ≤ 5.55E-04) compared to control. Other potential beneficial bacteria, including Rumminococcaceae UCG 005, Roseburia and Olsenella, were only increased (FDR ≤ 1.30E-02) by SCB treatment compared to control. Furthermore, the pathogenic bacterium, Peptoclostridium, was reduced (FDR = 1.58E-02) by SCB only while LA reduced (FDR = 1.74E-05) Ruminococcus_2. Functional prediction analysis suggested that both DFMs impacted (p < 0.05) pathways such as cell cycle, bile secretion, proteasome, cAMP signaling pathway, thyroid hormone synthesis pathway and dopaminergic synapse pathway. Compared to the DFMs, ATB had similar impact on bacterial diversity in all GIT sites but greater impact on the bacterial composition of the ileum. Overall, this study provides an insight on the bacteria genera impacted by DFMs and the potential mechanisms by which DFMs affect the GIT microbiota and may therefore facilitate development of DFMs as alternatives to ATB use in dairy calf management.
Collapse
|
145
|
Latorre JD, Adhikari B, Park SH, Teague KD, Graham LE, Mahaffey BD, Baxter MFA, Hernandez-Velasco X, Kwon YM, Ricke SC, Bielke LR, Hargis BM, Tellez G. Evaluation of the Epithelial Barrier Function and Ileal Microbiome in an Established Necrotic Enteritis Challenge Model in Broiler Chickens. Front Vet Sci 2018; 5:199. [PMID: 30186844 PMCID: PMC6110846 DOI: 10.3389/fvets.2018.00199] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022] Open
Abstract
Necrotic enteritis (NE) is a recognized multifactorial disease that cost annually to the poultry industry around $2 billion. However, diverse aspects related to its presentation are not completely understood, requiring further studies using known induction experimental models. Therefore, the purpose of this study was to measure the changes occurring in performance, intestinal integrity and ileal microbiome using a previously established NE-challenge model. Chickens were assigned to a negative control group (NC) or a positive control group (PC). In the PC, broilers were orally gavaged with Salmonella Typhimurium (ST) (1 × 107 cfu/chick) at day 1, Eimeria maxima (EM) (2.5 × 104 oocyst/chick) at day 18 and Clostridium perfringens (CP) (1 × 108 cfu/chick/day) at 23-24 days of age. Weekly, body weight (BW), body weight gain (BWG), feed intake (FI) and feed conversion ratio (FCR) were evaluated. Morbidity and mortality were determined throughout the study, and NE lesion scores were recorded at day 25. Additionally, blood and liver samples were collected to measure gut permeability as determined by levels of serum fluorescein isothiocyanate-dextran (FITC-d) and bacterial translocation (BT). Ileal contents were processed for 16S rRNA gene-based microbiome analysis. Performance parameters and intestinal permeability measurements were negatively impacted in the PC resulting in elevated serum FITC-d and BT with a -6.4% difference in BWG. The NE lesion score in PC (1.97 vs. 0.00) was significantly higher in comparison to NC, although there was no difference in mortality. The microbiome analysis showed a dramatic shift of ileal microbiomes in PC groups as compared to NC (ANOSIM: R = 0.76, P = 0.001). The shift was characterized by reduced abundance of the phylum Actinobacteria (P < 0.01), and increased abundance of the genera Butyrivibrio, Lactobacillus, Prevotella and Ruminococcus in PC compared to NC (P < 0.05). Expectedly, Clostridium was found higher in PC (2.98 ± 0.71%) as compared to NC (1.84 ± 0.36%), yet the difference was not significant. In conclusion, results of the present study showed the different intestinal epithelial and microbiological alterations occurring in an established NE-challenge model that considers paratyphoid Salmonella infections in young chicks as an important predisposing factor for presentation of NE.
Collapse
Affiliation(s)
- Juan D. Latorre
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Bishnu Adhikari
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Si H. Park
- Department of Food Science and Technology, Oregon State University, Corvallis, OR, United States
| | - Kyle D. Teague
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Lucas E. Graham
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Brittany D. Mahaffey
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Mikayla F. A. Baxter
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | | | - Young M. Kwon
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Steven C. Ricke
- Department of Food Science, Center of Food Safety, University of Arkansas, Fayetteville, AR, United States
| | - Lisa R. Bielke
- Department of Animal Science, The Ohio State University, Columbus, OH, United States
| | - Billy M. Hargis
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Guillermo Tellez
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
146
|
Effect of probiotic Bifidobacterium bifidum G9-1 on the relationship between gut microbiota profile and stress sensitivity in maternally separated rats. Sci Rep 2018; 8:12384. [PMID: 30120330 PMCID: PMC6098190 DOI: 10.1038/s41598-018-30943-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022] Open
Abstract
Although gut microbiota and early life events are likely involved in the development of irritable bowel syndrome (IBS), it remains unclear how these factors interact in the pathophysiology of IBS. In the present study, using rats subjected to maternal separation (MS) as a model of IBS, we investigated interrelationships among gut microbiota, stress susceptibility and intestinal permeability, and examined the effect of the probiotic Bifidobacterium bifidum G9-1 (BBG9-1) on those interrelationships. When compared with the controls at postnatal day 20, MS rats showed hypercorticosteronemia, enhanced intestinal permeability and changes in gut microbiota structure. All of these changes in MS rats were prevented by treatment with BBG9-1. Although the gut microbiota profile and basal serum corticosterone level did not differ between MS and control rats at postnatal day 56, MS rats showed hypersensitivity to restraint stress in terms of serum corticosterone level and fecal frequency. However, such hypersensitivity was not observed in MS rats treated with BBG9-1. These findings suggest that MS initiates the link between gut microbiota alteration and hypersensitivity to stress and that the triggering of this process can be prevented by the treatment with the probiotic BBG9-1.
Collapse
|
147
|
Corfield AP. The Interaction of the Gut Microbiota with the Mucus Barrier in Health and Disease in Human. Microorganisms 2018; 6:microorganisms6030078. [PMID: 30072673 PMCID: PMC6163557 DOI: 10.3390/microorganisms6030078] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023] Open
Abstract
Glycoproteins are major players in the mucus protective barrier in the gastrointestinal and other mucosal surfaces. In particular the mucus glycoproteins, or mucins, are responsible for the protective gel barrier. They are characterized by their high carbohydrate content, present in their variable number, tandem repeat domains. Throughout evolution the mucins have been maintained as integral components of the mucosal barrier, emphasizing their essential biological status. The glycosylation of the mucins is achieved through a series of biosynthetic pathways processes, which generate the wide range of glycans found in these molecules. Thus mucins are decorated with molecules having information in the form of a glycocode. The enteric microbiota interacts with the mucosal mucus barrier in a variety of ways in order to fulfill its many normal processes. How bacteria read the glycocode and link to normal and pathological processes is outlined in the review.
Collapse
Affiliation(s)
- Anthony P Corfield
- Mucin Research Group, School of Clinical Sciences, Bristol Royal Infirmary, Level 7, Marlborough Street, Bristol BS2 8HW, UK.
| |
Collapse
|
148
|
Quintana-Hayashi MP, Padra M, Padra JT, Benktander J, Lindén SK. Mucus-Pathogen Interactions in the Gastrointestinal Tract of Farmed Animals. Microorganisms 2018; 6:E55. [PMID: 29912166 PMCID: PMC6027344 DOI: 10.3390/microorganisms6020055] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/09/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Gastrointestinal infections cause significant challenges and economic losses in animal husbandry. As pathogens becoming resistant to antibiotics are a growing concern worldwide, alternative strategies to treat infections in farmed animals are necessary in order to decrease the risk to human health and increase animal health and productivity. Mucosal surfaces are the most common route used by pathogens to enter the body. The mucosal surface that lines the gastrointestinal tract is covered by a continuously secreted mucus layer that protects the epithelial surface. The mucus layer is the first barrier the pathogen must overcome for successful colonization, and is mainly composed of densely glycosylated proteins called mucins. The vast array of carbohydrate structures present on the mucins provide an important setting for host-pathogen interactions. This review summarizes the current knowledge on gastrointestinal mucins and their role during infections in farmed animals. We examine the interactions between mucins and animal pathogens, with a focus on how pathogenic bacteria can modify the mucin environment in the gut, and how this in turn affects pathogen adhesion and growth. Finally, we discuss analytical challenges and complexities of the mucus-based defense, as well as its potential to control infections in farmed animals.
Collapse
Affiliation(s)
- Macarena P Quintana-Hayashi
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - Médea Padra
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - János Tamás Padra
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - John Benktander
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| | - Sara K Lindén
- Department of Medical Biochemistry and Cell biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, 405 30 Gothenburg, Sweden.
| |
Collapse
|
149
|
Mucus: An Underestimated Gut Target for Environmental Pollutants and Food Additives. Microorganisms 2018; 6:microorganisms6020053. [PMID: 29914144 PMCID: PMC6027178 DOI: 10.3390/microorganisms6020053] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/08/2018] [Accepted: 06/14/2018] [Indexed: 12/22/2022] Open
Abstract
Synthetic chemicals (environmental pollutants, food additives) are widely used for many industrial purposes and consumer-related applications, which implies, through manufactured products, diet, and environment, a repeated exposure of the general population with growing concern regarding health disorders. The gastrointestinal tract is the first physical and biological barrier against these compounds, and thus their first target. Mounting evidence indicates that the gut microbiota represents a major player in the toxicity of environmental pollutants and food additives; however, little is known on the toxicological relevance of the mucus/pollutant interplay, even though mucus is increasingly recognized as essential in gut homeostasis. Here, we aimed at describing how environmental pollutants (heavy metals, pesticides, and other persistent organic pollutants) and food additives (emulsifiers, nanomaterials) might interact with mucus and mucus-related microbial species; that is, “mucophilic” bacteria such as mucus degraders. This review highlights that intestinal mucus, either directly or through its crosstalk with the gut microbiota, is a key, yet underestimated gut player that must be considered for better risk assessment and management of environmental pollution.
Collapse
|
150
|
Kazou M, Alexandraki V, Blom J, Pot B, Tsakalidou E, Papadimitriou K. Comparative Genomics of Lactobacillus acidipiscis ACA-DC 1533 Isolated From Traditional Greek Kopanisti Cheese Against Species Within the Lactobacillus salivarius Clade. Front Microbiol 2018; 9:1244. [PMID: 29942291 PMCID: PMC6004923 DOI: 10.3389/fmicb.2018.01244] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/23/2018] [Indexed: 02/05/2023] Open
Abstract
Lactobacillus acidipiscis belongs to the Lactobacillus salivarius clade and it is found in a variety of fermented foods. Strain ACA-DC 1533 was isolated from traditional Greek Kopanisti cheese and among the available L. acidipiscis genomes it is the only one with a fully sequenced chromosome. L. acidipiscis strains exhibited a high degree of conservation at the genome level. Investigation of the distribution of prophages and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPRs) among the three strains suggests the potential existence of lineages within the species. Based on the presence/absence patterns of these genomic traits, strain ACA-DC 1533 seems to be more related to strain JCM 10692T than strain KCTC 13900. Interestingly, strains ACA-DC 1533 and JCM 10692T which lack CRISPRs, carry two similar prophages. In contrast, strain KCTC 13900 seems to have acquired immunity to these prophages according to the sequences of spacers in its CRISPRs. Nonetheless, strain KCTC 13900 has a prophage that is absent from strains ACA-DC 1533 and JCM 10692T. Furthermore, comparative genomic analysis was performed among L. acidipiscis ACA-DC 1533, L. salivarius UCC118 and Lactobacillus ruminis ATCC 27782. The chromosomes of the three species lack long-range synteny. Important differences were also determined in the number of glycobiome related proteins, proteolytic enzymes, transporters, insertion sequences and regulatory proteins. Moreover, no obvious genomic traits supporting a probiotic potential of L. acidipiscis ACA-DC 1533 were detected when compared to the probiotic L. salivarius UCC118. However, the existence of more than one glycine-betaine transporter within the genome of ACA-DC 1533 may explain the ability of L. acidipiscis to grow in fermented foods containing high salt concentrations. Finally, in silico analysis of the L. acidipiscis ACA-DC 1533 genome revealed pathways that could underpin the production of major volatile compounds during the catabolism of amino acids that may contribute to the typical piquant flavors of Kopanisti cheese.
Collapse
Affiliation(s)
- Maria Kazou
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | - Voula Alexandraki
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | - Jochen Blom
- Bioinformatics and Systems Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Bruno Pot
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Department of Bioengineering Sciences (DBIT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Effie Tsakalidou
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | - Konstantinos Papadimitriou
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| |
Collapse
|