1
|
Rawat N, Sivanesan S, Kanade GS, Bafana A. Interaction of environmental fluoride exposure and gut microbes: Potential implication in the development of fluorosis in human subjects. Food Chem Toxicol 2025; 200:115388. [PMID: 40086585 DOI: 10.1016/j.fct.2025.115388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Fluoride exposure primarily occurs through contaminated water and leads to fluorosis, which is a global health concern. After ingestion, fluoride is absorbed via gastrointestinal tract, where it interacts with the gut microbiota. While animal studies have explored fluoride's effects on gut microbiota, no human studies have yet been conducted. Most research emphasizes metagenomic diversity, neglecting isolation and characterization of pure cultures for further applications. Additionally, the association between gut microbiota with fluorosis outcomes in fluoride-exposed populations is unexplored. This study characterizes and compares the cultivable gut microbiota in the fluoride-exposed population with (symptomatic, group II) or without (asymptomatic, group I) signs of skeletal fluorosis along with unexposed control (group III). Group I displayed higher abundance of Firmicutes (58.58 %), group II had predominance of Proteobacteria (61.25 %) while group III showed similar abundance of Proteobacteria (50.38 %) and Firmicutes (49.51 %). On analyzing short-chain fatty acid (SCFA) profiles, group I isolates produced higher isobutyric acid (1.31 ± 0.9 mM) than group II (0.71 ± 0.35 mM), while group II produced more isovaleric acid (0.8 ± 0.41 mM) than group I (0.61 ± 0.08 mM) (p < 0.05). These findings suggest that gut microbiota and SCFAs alteration may influence bone metabolism, affecting the fluorosis progression.
Collapse
Affiliation(s)
- Neha Rawat
- CSIR-NEERI (National Environmental Engineering Research Institute), Nehru Marg, Nagpur 440020, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Saravanadevi Sivanesan
- CSIR-NEERI (National Environmental Engineering Research Institute), Nehru Marg, Nagpur 440020, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Gajanan Sitaramji Kanade
- CSIR-NEERI (National Environmental Engineering Research Institute), Nehru Marg, Nagpur 440020, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Amit Bafana
- CSIR-NEERI (National Environmental Engineering Research Institute), Nehru Marg, Nagpur 440020, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
2
|
Amen RA, Hassan YM, Essmat RA, Ahmed RH, Azab MM, Shehata NR, Elgazzar MM, El-Sayed WM. Harnessing the Microbiome: CRISPR-Based Gene Editing and Antimicrobial Peptides in Combating Antibiotic Resistance and Cancer. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10573-8. [PMID: 40377870 DOI: 10.1007/s12602-025-10573-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
The growing crisis of antibiotic resistance and the increasing incidence of cancer have prompted the exploration of innovative approaches, such as gene editing and antimicrobial peptides (AMPs). The human microbiome is integral to various aspects of health, disease, and therapeutic development, influencing metabolic pathways, immune function, and pathogen resistance. Recent advances in gene editing technologies, particularly CRISPR (clustered regularly interspaced short palindromic repeats), have opened new avenues for leveraging the microbiome to address complex medical challenges, including combating multidrug-resistant pathogens and cancer. The microbiome plays a crucial role in combating antibiotic resistance by modulating microbial communities, influencing pathogen survival and susceptibility to treatments. This review explores the microbiome's dynamic role in metabolic regulation, its contribution to cancer management, and how AMPs help maintain homeostasis and exhibit emerging anticancer properties, supported by both preclinical findings and clinical evidence. Additionally, CRISPR-based microbiome engineering offers potential to enhance host-microbiome interactions, optimizing therapeutic outcomes. The integration of microbiome metagenomics and proteomics has led to the discovery of novel AMPs with targeted anticancer effects. Innovative strategies, such as engineered probiotics and CRISPR-based microbiome engineering, present exciting prospects for next-generation therapies. Despite these advances, the translation of microbiome-based therapies into clinical settings remains challenging due to ethical, regulatory, and ecological hurdles. This review underscores the transformative potential of microbiome-based interventions, emphasizing the role of personalized medicine in maximizing therapeutic efficacy. Furthermore, we also address critical research gaps, limitations, and future directions, including optimizing AMP stability, delivery, and bioavailability, as well as overcoming the regulatory and ethical challenges in clinical translation.
Collapse
Affiliation(s)
- Radwa A Amen
- Department of Biotechnology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Yaser M Hassan
- Biotechnology Program, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Rawan A Essmat
- Faculty of Pharmacy, Modern University for Information and Technology, Cairo, 11728, Egypt
| | - Rana H Ahmed
- Biotechnology Program, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Marwan M Azab
- Molecular Biotechnology Program, Faculty of Science, Helwan University, Ain Helwan, Cairo, Egypt
| | - Nadia R Shehata
- College of Biotechnology, Misr University for Science and Technology, Giza, 12596, Egypt
| | | | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia 11566, Cairo, Egypt.
| |
Collapse
|
3
|
Schoultz I, Claesson MJ, Dominguez-Bello MG, Fåk Hållenius F, Konturek P, Korpela K, Laursen MF, Penders J, Roager H, Vatanen T, Öhman L, Jenmalm MC. Gut microbiota development across the lifespan: Disease links and health-promoting interventions. J Intern Med 2025. [PMID: 40270478 DOI: 10.1111/joim.20089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
The gut microbiota plays a pivotal role in human life and undergoes dynamic changes throughout the human lifespan, from infancy to old age. During our life, the gut microbiota influences health and disease across life stages. This review summarizes the discussions and presentations from the symposium "Gut microbiota development from infancy to old age" held in collaboration with the Journal of Internal Medicine. In early infancy, microbial colonization is shaped by factors such as mode of delivery, antibiotic exposure, and milk-feeding practices, laying the foundation for subsequent increased microbial diversity and maturation. Throughout childhood and adolescence, microbial maturation continues, influencing immune development and metabolic health. In adulthood, the gut microbiota reaches a relatively stable state, influenced by genetics, diet, and lifestyle. Notably, disruptions in gut microbiota composition have been implicated in various inflammatory diseases-including inflammatory bowel disease, Type 1 diabetes, and allergies. Furthermore, emerging evidence suggests a connection between gut dysbiosis and neurodegenerative disorders such as Alzheimer's disease. Understanding the role of the gut microbiota in disease pathogenesis across life stages provides insights into potential therapeutic interventions. Probiotics, prebiotics, and dietary modifications, as well as fecal microbiota transplantation, are being explored as promising strategies to promote a healthy gut microbiota and mitigate disease risks. This review focuses on the gut microbiota's role in infancy, adulthood, and aging, addressing its development, stability, and alterations linked to health and disease across these critical life stages. It outlines future research directions aimed at optimizing the gut microbiota composition to improve health.
Collapse
Affiliation(s)
- Ida Schoultz
- School of Medical Sciences, Faculty of Medicine and Health Örebro University, Orebro, Sweden
| | | | - Maria Gloria Dominguez-Bello
- Department of Biochemistry & Microbiology and of Anthropology, Rutgers University-New Brunswick, New Brunswick, New Jersey, USA
| | - Frida Fåk Hållenius
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Peter Konturek
- Department of Medicine, Thuringia Clinic Saalfeld, Teaching Hospital of the University Jena, Jena, Germany
| | - Katri Korpela
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - H Roager
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Tommi Vatanen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Cappio Barazzone E, Diard M, Hug I, Larsson L, Slack E. Diagnosing and engineering gut microbiomes. EMBO Mol Med 2024; 16:2660-2677. [PMID: 39468301 PMCID: PMC11554810 DOI: 10.1038/s44321-024-00149-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
The microbes, nutrients and toxins that we are exposed to can have a profound effect on the composition and function of the gut microbiome. Thousands of peer-reviewed publications link microbiome composition and function to health from the moment of birth, right through to centenarians, generating a tantalizing glimpse of what might be possible if we could intervene rationally. Nevertheless, there remain relatively few real-world examples where successful microbiome engineering leads to beneficial health effects. Here we aim to provide a framework for the progress needed to turn gut microbiome engineering from a trial-and-error approach to a rational medical intervention. The workflow starts with truly understanding and accurately diagnosing the problems that we are trying to fix, before moving on to developing technologies that can achieve the desired changes.
Collapse
Affiliation(s)
- Elisa Cappio Barazzone
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Médéric Diard
- Basel Research Centre for Child Health, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Isabelle Hug
- Basel Research Centre for Child Health, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Louise Larsson
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Emma Slack
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland.
- Basel Research Centre for Child Health, Basel, Switzerland.
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
Yao G, Zhao Z, Yang C, Zuo B, Sun Z, Wang J, Zhang H. Evaluating the probiotic effects of spraying lactiplantibacillus plantarum P-8 in neonatal piglets. BMC Microbiol 2024; 24:253. [PMID: 38982403 PMCID: PMC11232343 DOI: 10.1186/s12866-024-03332-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/13/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Gut microbes play an important role in the growth and health of neonatal piglets. Probiotics can promote the healthy growth of neonatal piglets by regulating their gut microbes. The study investigated the effects of spraying Lactiplantibacillus plantarum P-8 (L. plantarum P-8) fermentation broth on the growth performance and gut microbes of neonatal piglets. RESULTS The animals were randomly divided into probiotics groups (109 neonatal piglets) and control groups (113 neonatal piglets). The probiotics group was sprayed with L. plantarum P-8 fermented liquid from 3 day before the expected date of the sow to the 7-day-old of piglets, while the control group was sprayed with equal dose of PBS. Average daily gain (ADG), immune and antioxidant status and metagenome sequencing were used to assess the changes in growth performance and gut microbiota of neonatal piglets. The results showed that L. plantarum P-8 treatment significantly improved the average daily gain (P < 0.05) of neonatal piglets. L. plantarum P-8 increased the activities of CAT and SOD but reduced the levels of IL-2 and IL-6, effectively regulating the antioxidant capacity and immunity in neonatal piglets. L. plantarum P-8 adjusted the overall structure of gut microflora improving gut homeostasis to a certain extent, and significantly increased the relative abundance of gut beneficial bacteria such as L. mucosae and L. plantarum. CONCLUSION Spraying L. plantarum P-8 can be a feasible and effective probiotic intervention not only improving the growth of neonatal piglets, regulating the antioxidant capacity and immunity of neonatal piglets, but also improving the gut homeostasis to a certain extent.
Collapse
Affiliation(s)
- Guoqiang Yao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Huhhot, Inner Mongolia, China
| | - Zhixin Zhao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Huhhot, Inner Mongolia, China
| | - Chengcong Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Huhhot, Inner Mongolia, China
| | - Bin Zuo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Huhhot, Inner Mongolia, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Huhhot, Inner Mongolia, China.
| |
Collapse
|
6
|
Luo Y, Liao H, Huang X, Zhang C, Gao L, Wang Z, Xia X. Unraveling the Metabolic Behavior and Interspecific Interaction Pattern of Lactic Acid Bacteria within Chinese Rice Wine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14899-14911. [PMID: 38913831 DOI: 10.1021/acs.jafc.4c02461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The synthetic community of lactic acid bacteria (LAB) is commonly utilized in the food industry for manipulating product properties. However, the intermediate interactions and ecological stability resulting from metabolic differences among various LAB types remain poorly understood. We aimed to analyze the metabolic behavior of single and combined lactic acid bacteria in China rice wine based on microbial succession. Three-stage succession patterns with obligate heterofermentative LAB dominating prefermentation and homofermentative LAB prevailing in main fermentation were observed. Facultative heterofermentative LAB exhibited significant growth. Pairwise coculture interactions revealed 63.5% positive, 34.4% negative, and 2.1% neutral interactions, forming nontransitive and transitive competition modes. Nontransitive competitive combinations demonstrated stability over ∼200 generations through amino acid (mainly aspartic acid, glutamine, and serine) cross-feeding and lactic acid detoxification, which also showed potential for controlling biogenic amines and developing LAB starter cultures. Our findings offer insights into the mechanistic underpinnings of LAB interaction networks.
Collapse
Affiliation(s)
- Yi Luo
- Institute of Environmental Processes and Pollution Control, School of Environmental and Civil Engineering, Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi 214122, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, P. R. China
| | - Hui Liao
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, P. R. China
| | - Xinlei Huang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, P. R. China
| | - Chenhao Zhang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, P. R. China
| | - Ling Gao
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, P. R. China
- Jiangsu Tanggou Liangxianghe Liquor Co., Ltd., Lianyungang, Jiangsu 222535, P. R. China
| | - Zhenyu Wang
- Institute of Environmental Processes and Pollution Control, School of Environmental and Civil Engineering, Jiangsu Engineering Laboratory for Biomass Energy and Carbon Reduction Technology, Jiangsu Key Laboratory of Anaerobic Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Xiaole Xia
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, P. R. China
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300000, P. R. China
| |
Collapse
|
7
|
Jennings SAV, Clavel T. Synthetic Communities of Gut Microbes for Basic Research and Translational Approaches in Animal Health and Nutrition. Annu Rev Anim Biosci 2024; 12:283-300. [PMID: 37963399 DOI: 10.1146/annurev-animal-021022-025552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Microbes and animals have a symbiotic relationship that greatly influences nutrient uptake and animal health. This relationship can be studied using selections of microbes termed synthetic communities, or SynComs. SynComs are used in many different animal hosts, including agricultural animals, to investigate microbial interactions with nutrients and how these affect animal health. The most common host focuses for SynComs are currently mouse and human, from basic mechanistic research through to translational disease models and live biotherapeutic products (LBPs) as treatments. We discuss SynComs used in basic research models and findings that relate to human and animal health and nutrition. Translational use cases of SynComs are discussed, followed by LBPs, especially within the context of agriculture. SynComs still face challenges, such as standardization for reproducibility and contamination risks. However, the future of SynComs is hopeful, especially in the areas of genome-guided SynCom design and custom SynCom-based treatments.
Collapse
Affiliation(s)
- Susan A V Jennings
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany;
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany;
| |
Collapse
|
8
|
Stevick RJ, Audrain B, Bedu S, Dray N, Ghigo JM, Pérez-Pascual D. Anti-diarrheal drug loperamide induces dysbiosis in zebrafish microbiota via bacterial inhibition. MICROBIOME 2023; 11:252. [PMID: 37951983 PMCID: PMC10638762 DOI: 10.1186/s40168-023-01690-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/04/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Perturbations of animal-associated microbiomes from chemical stress can affect host physiology and health. While dysbiosis induced by antibiotic treatments and disease is well known, chemical, nonantibiotic drugs have recently been shown to induce changes in microbiome composition, warranting further exploration. Loperamide is an opioid-receptor agonist widely prescribed for treating acute diarrhea in humans. Loperamide is also used as a tool to study the impact of bowel dysfunction in animal models by inducing constipation, but its effect on host-associated microbiota is poorly characterized. RESULTS We used conventional and gnotobiotic larval zebrafish models to show that in addition to host-specific effects, loperamide also has anti-bacterial activities that directly induce changes in microbiota diversity. This dysbiosis is due to changes in bacterial colonization, since gnotobiotic zebrafish mono-colonized with bacterial strains sensitive to loperamide are colonized up to 100-fold lower when treated with loperamide. Consistently, the bacterial diversity of gnotobiotic zebrafish colonized by a mix of 5 representative bacterial strains is affected by loperamide treatment. CONCLUSION Our results demonstrate that loperamide, in addition to host effects, also induces dysbiosis in a vertebrate model, highlighting that established treatments can have underlooked secondary effects on microbiota structure and function. This study further provides insights for future studies exploring how common medications directly induce changes in host-associated microbiota. Video Abstract.
Collapse
Affiliation(s)
- Rebecca J Stevick
- Genetics of Biofilms Laboratory, UMR 6047, Institut Pasteur Université Paris Cité, CNRS, Paris, France
| | - Bianca Audrain
- Genetics of Biofilms Laboratory, UMR 6047, Institut Pasteur Université Paris Cité, CNRS, Paris, France
| | - Sébastien Bedu
- Zebrafish Neurogenetics Laboratory, UMR 3738, Institut Pasteur Université Paris Cité, CNRS, Paris, France
| | - Nicolas Dray
- Zebrafish Neurogenetics Laboratory, UMR 3738, Institut Pasteur Université Paris Cité, CNRS, Paris, France
| | - Jean-Marc Ghigo
- Genetics of Biofilms Laboratory, UMR 6047, Institut Pasteur Université Paris Cité, CNRS, Paris, France.
| | - David Pérez-Pascual
- Genetics of Biofilms Laboratory, UMR 6047, Institut Pasteur Université Paris Cité, CNRS, Paris, France.
| |
Collapse
|
9
|
Adade EE, Stevick RJ, Pérez-Pascual D, Ghigo JM, Valm AM. Gnotobiotic zebrafish microbiota display inter-individual variability affecting host physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526612. [PMID: 36778358 PMCID: PMC9915576 DOI: 10.1101/2023.02.01.526612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Gnotobiotic animal models reconventionalized under controlled laboratory conditions with multi-species bacterial communities are commonly used to study host-microbiota interactions under presumably more reproducible conditions than conventional animals. The usefulness of these models is however limited by inter-animal variability in bacterial colonization and our general lack of understanding of the inter-individual fluctuation and spatio-temporal dynamics of microbiota assemblies at the micron to millimeter scale. Here, we show underreported variability in gnotobiotic models by analyzing differences in gut colonization efficiency, bacterial composition, and host intestinal mucus production between conventional and gnotobiotic zebrafish larvae re-conventionalized with a mix of 9 bacteria isolated from conventional microbiota. Despite similar bacterial community composition, we observed high variability in the spatial distribution of bacteria along the intestinal tract in the reconventionalized model. We also observed that, whereas bacteria abundance and intestinal mucus per fish were not correlated, reconventionalized fish had lower intestinal mucus compared to conventional animals, indicating that the stimulation of mucus production depends on the microbiota composition. Our findings, therefore, suggest that variable colonization phenotypes affect host physiology and impact the reproducibility of experimental outcomes in studies that use gnotobiotic animals. This work provides insights into the heterogeneity of gnotobiotic models and the need to accurately assess re-conventionalization for reproducibility in host-microbiota studies.
Collapse
Affiliation(s)
- Emmanuel E. Adade
- Department of Biological Sciences, State University of New York at Albany, Albany, NY 12222, USA
- The RNA Institute, State University of New York at Albany, Albany, NY 12222, USA
| | - Rebecca J. Stevick
- Institut Pasteur, Université de Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris F-75015, France
| | - David Pérez-Pascual
- Institut Pasteur, Université de Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris F-75015, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Université de Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris F-75015, France
| | - Alex M. Valm
- Department of Biological Sciences, State University of New York at Albany, Albany, NY 12222, USA
- The RNA Institute, State University of New York at Albany, Albany, NY 12222, USA
| |
Collapse
|
10
|
Fonseca RJC, Mourão PAS. Pharmacological Activities of Sulfated Fucose-Rich Polysaccharides after Oral Administration: Perspectives for the Development of New Carbohydrate-Based Drugs. Mar Drugs 2021; 19:425. [PMID: 34436263 PMCID: PMC8400256 DOI: 10.3390/md19080425] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Marine organisms are a source of active biomolecules with immense therapeutic and nutraceutical potential. Sulfated fucose-rich polysaccharides are present in large quantities in these organisms with important pharmacological effects in several biological systems. These polysaccharides include sulfated fucan (as fucoidan) and fucosylated chondroitin sulfate. The development of these polysaccharides as new drugs involves several important steps, among them, demonstration of the effectiveness of these compounds after oral administration. The oral route is the more practical, comfortable and preferred by patients for long-term treatments. In the past 20 years, reports of various pharmacological effects of these polysaccharides orally administered in several animal experimental models and some trials in humans have sparked the possibility for the development of drugs based on sulfated polysaccharides and/or the use of these marine organisms as functional food. This review focuses on the main pharmacological effects of sulfated fucose-rich polysaccharides, with an emphasis on the antidislipidemic, immunomodulatory, antitumor, hypoglycemic and hemostatic effects.
Collapse
Affiliation(s)
- Roberto J. C. Fonseca
- Laboratório de Tecido Conjuntivo, Hospital Universitário Clementino Fraga Filho, Rio de Janeiro 21941-913, Brazil;
- Centro de Ciências da Saúde, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Paulo A. S. Mourão
- Laboratório de Tecido Conjuntivo, Hospital Universitário Clementino Fraga Filho, Rio de Janeiro 21941-913, Brazil;
- Centro de Ciências da Saúde, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| |
Collapse
|
11
|
Bayer F, Ascher S, Kiouptsi K, Kittner JM, Stauber RH, Reinhardt C. Colonization with Altered Schaedler Flora Impacts Leukocyte Adhesion in Mesenteric Ischemia-Reperfusion Injury. Microorganisms 2021; 9:1601. [PMID: 34442681 PMCID: PMC8401286 DOI: 10.3390/microorganisms9081601] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
The microbiota impacts mesenteric ischemia-reperfusion injury, aggravating the interaction of leukocytes with endothelial cells in mesenteric venules. The role of defined gut microbiomes in this life-threatening pathology is unknown. To investigate how a defined model microbiome affects the adhesion of leukocytes in mesenteric ischemia-reperfusion, we took advantage of gnotobiotic isolator technology and transferred altered Schaedler flora (ASF) from C3H/HeNTac to germ-free C57BL/6J mice. We were able to detect all eight bacterial taxa of ASF in fecal samples of colonized C57BL/6J mice by PCR. Applying qRT-PCR for quantification of species-specific 16S rDNA sequences of ASF bacteria, we found a major shift in the abundance of ASF 500, which was greater in C57BL/6J mice relative to the C3H/HeNTac founder breeding pair. Using high-speed epifluorescence intravital microscopy to visualize the venules of the small bowel mesentery, we found that gnotobiotic ASF-colonized mice showed reduced leukocyte adherence, both pre- and post-ischemia. Relative to germ-free mice, the counts of adhering leukocytes were increased pre-ischemia but did not significantly increase in ASF-colonized mice in the post-ischemic state. Collectively, our results suggest a protective role of the minimal microbial consortium ASF in mesenteric ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Franziska Bayer
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (S.A.); (K.K.)
| | - Stefanie Ascher
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (S.A.); (K.K.)
- Department of Chemistry, Biochemistry, Johannes Gutenberg-University Mainz, 55128 Mainz, Germany
| | - Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (S.A.); (K.K.)
| | - Jens M. Kittner
- Department of Medicine, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany;
- Diakonie Klinikum Neunkirchen, Brunnenstraße 20, 66538 Neunkirchen, Germany
| | - Roland H. Stauber
- Department of Nanobiomedicine/ENT, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany;
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (S.A.); (K.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, 55131 Mainz, Germany
| |
Collapse
|
12
|
Kumar N, Hitch TCA, Haller D, Lagkouvardos I, Clavel T. MiMiC: a bioinformatic approach for generation of synthetic communities from metagenomes. Microb Biotechnol 2021; 14:1757-1770. [PMID: 34081399 PMCID: PMC8313253 DOI: 10.1111/1751-7915.13845] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 01/23/2023] Open
Abstract
Environmental and host-associated microbial communities are complex ecosystems, of which many members are still unknown. Hence, it is challenging to study community dynamics and important to create model systems of reduced complexity that mimic major community functions. Therefore, we developed MiMiC, a computational approach for data-driven design of simplified communities from shotgun metagenomes. We first built a comprehensive database of species-level bacterial and archaeal genomes (n = 22 627) consisting of binary (presence/absence) vectors of protein families (Pfam = 17 929). MiMiC predicts the composition of minimal consortia using an iterative scoring system based on maximal match-to-mismatch ratios between this database and the Pfam binary vector of any input metagenome. Pfam vectorization retained enough resolution to distinguish metagenomic profiles between six environmental and host-derived microbial communities (n = 937). The calculated number of species per minimal community ranged between 5 and 11, with MiMiC selected communities better recapitulating the functional repertoire of the original samples than randomly selected species. The inferred minimal communities retained habitat-specific features and were substantially different from communities consisting of most abundant members. The use of a mixture of known microbes revealed the ability to select 23 of 25 target species from the entire genome database. MiMiC is open source and available at https://github.com/ClavelLab/MiMiC.
Collapse
Affiliation(s)
- Neeraj Kumar
- Functional Microbiome Research GroupInstitute of Medical MicrobiologyUniversity Hospital of RWTHAachenGermany
- ZIEL‐ Institute for Food and HealthTechnical University of MunichFreisingGermany
| | - Thomas C. A. Hitch
- Functional Microbiome Research GroupInstitute of Medical MicrobiologyUniversity Hospital of RWTHAachenGermany
| | - Dirk Haller
- ZIEL‐ Institute for Food and HealthTechnical University of MunichFreisingGermany
- Chair of Nutrition and ImmunologyTechnical University of MunichFreisingGermany
| | - Ilias Lagkouvardos
- ZIEL‐ Institute for Food and HealthTechnical University of MunichFreisingGermany
- Institute of Marine Biology, Biotechnology and AquacultureHellenic Center of Marine ResearchHeraklionGreece
| | - Thomas Clavel
- Functional Microbiome Research GroupInstitute of Medical MicrobiologyUniversity Hospital of RWTHAachenGermany
| |
Collapse
|
13
|
Basic M, Bolsega S, Smoczek A, Gläsner J, Hiergeist A, Eberl C, Stecher B, Gessner A, Bleich A. Monitoring and contamination incidence of gnotobiotic experiments performed in microisolator cages. Int J Med Microbiol 2021; 311:151482. [PMID: 33636479 DOI: 10.1016/j.ijmm.2021.151482] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/14/2021] [Accepted: 02/16/2021] [Indexed: 10/22/2022] Open
Abstract
With the increased interest in the microbiome research, gnotobiotic animals and techniques emerged again as valuable tools to investigate functional effects of host-microbe and microbe-microbe interactions. The increased demand for gnotobiotic experiments has resulted in the greater need for housing systems for short-term maintenance of gnotobiotic animals. During the last six years, the gnotobiotic facility of the Hannover Medical School has worked intensively with different housing systems for gnotobiotic animals. Here, we report our experience in handling, contamination incidence, and monitoring strategies that we apply for controlling gnotobiotic experiments. From our experience, the risk of introducing contaminants to animals housed in microisolator cages is higher than in isolators. However, with strict operating protocols, the contamination rate in these systems can be minimized. In addition to spore-forming bacteria and fungi from the environment, spore-forming bacteria from defined bacterial communities used in experiments represent the major risk for contamination of gnotobiotic experiments performed in microisolator cages. The presence/absence of contaminants in germ-free animals can be easily monitored by preparation of wet mounts and Gram staining of fecal samples. Contaminants in animals colonized with specific microorganisms need to be tracked with methods such as next-generation sequencing. However, when using PCR-based methods it is important to consider that relatively small amounts of bacterial DNA detected likely originates from food, bedding, or reagents and is not to be interpreted as true contamination.
Collapse
Affiliation(s)
- Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Anna Smoczek
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Joachim Gläsner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Claudia Eberl
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Germany; German Center of Infection Research (DZIF), Partner Site Munich, Germany
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
14
|
Adhikari B, Kwon YM. Cell density alters bacterial community structure in culture-enriched 16S rRNA gene microbiota profiling. BMC Res Notes 2020; 13:269. [PMID: 32493423 PMCID: PMC7268277 DOI: 10.1186/s13104-020-05113-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/26/2020] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Microbial community profiling using 16S rRNA gene has provided invaluable insights into diverse microbial communities. Recently a few studies have attempted to use 16S rRNA gene microbiota profiling in combination with the conventional culture methods to explore bacterial communities. In this "culture-enriched microbiota profiling" approach, microbes in a sample are cultured on solid media, and the resulting colonies are combined and subjected to 16S rRNA gene microbiota profiling. Here we investigated the effect of cell densities as determined by varying levels of sample dilution on the culture-enriched microbiota profiles using De Man, Rogosa and Sharpe (MRS) agar medium as a model system. RESULTS Cecal samples collected from 10 healthy chickens were serially diluted to 102 fold (M-LOW), 104 fold (M-MEDIUM), and 106 fold (M-HIGH), and the dilutions were plated on MRS agar. 16S rRNA gene profiling showed that the relative abundance of certain genera showed gradual increase (Pediococcus and Enterococcus) or decrease (Lactobacillus and Turicibacter) with higher dilutions, though it was significant only for Pediococcus (p < 0.05). The result indicates that the dilution levels of the samples can alter the resulting microbiota profiles via unknown density-dependent mechanisms and thus should be considered for designing experiments using culture-enriched microbiota profiling.
Collapse
Affiliation(s)
- Bishnu Adhikari
- Department of Poultry Science, College of Agricultural, Food and Life Sciences, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Young Min Kwon
- Department of Poultry Science, College of Agricultural, Food and Life Sciences, University of Arkansas, Fayetteville, AR, 72701, USA. .,Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
15
|
Poeker SA, Lacroix C, de Wouters T, Spalinger MR, Scharl M, Geirnaert A. Stepwise Development of an in vitro Continuous Fermentation Model for the Murine Caecal Microbiota. Front Microbiol 2019; 10:1166. [PMID: 31191488 PMCID: PMC6548829 DOI: 10.3389/fmicb.2019.01166] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/07/2019] [Indexed: 12/24/2022] Open
Abstract
Murine models are valuable tools to study the role of gut microbiota in health or disease. However, murine and human microbiota differ in species composition, so further investigation of the murine gut microbiota is important to gain a better mechanistic understanding. Continuous in vitro fermentation models are powerful tools to investigate microbe-microbe interactions while circumventing animal testing and host confounding factors, but are lacking for murine gut microbiota. We therefore developed a novel continuous fermentation model based on the PolyFermS platform adapted to the murine caecum and inoculated with immobilized caecal microbiota. We followed a stepwise model development approach by adjusting parameters [pH, retention time (RT), growth medium] to reach fermentation metabolite profiles and marker bacterial levels similar to the inoculum. The final model had a stable and inoculum-alike fermentation profile during continuous operation. A lower pH during startup and continuous operation stimulated bacterial fermentation (115 mM short-chain fatty acids at pH 7 to 159 mM at pH 6.5). Adjustments to nutritive medium, a decreased pH and increased RT helped control the in vitro Enterobacteriaceae levels, which often bloom in fermentation models, to 6.6 log gene copies/mL in final model. In parallel, the Lactobacillus, Lachnospiraceae, and Ruminococcaceae levels were better maintained in vitro with concentrations of 8.5 log gene copies/mL, 8.8 log gene copies/mL and 7.5 log gene copies/mL, respectively, in the final model. An independent repetition with final model parameters showed reproducible results in maintaining the inoculum fermentation metabolite profile and its marker bacterial levels. Microbiota community analysis of the final model showed a decreased bacterial diversity and compositional differences compared to caecal inoculum microbiota. Most of the caecal bacterial families were represented in vitro, but taxa of the Muribaculaceae family were not maintained. Functional metagenomics prediction showed conserved metabolic and functional KEGG pathways between in vitro and caecal inoculum microbiota. To conclude, we showed that a rational and stepwise approach allowed us to model in vitro the murine caecal microbiota and functions. Our model is a first step to develop murine microbiota model systems and offers the potential to study microbiota functionality and structure ex vivo.
Collapse
Affiliation(s)
- Sophie A Poeker
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Tomas de Wouters
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Marianne R Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Reconstructing functional networks in the human intestinal tract using synthetic microbiomes. Curr Opin Biotechnol 2019; 58:146-154. [PMID: 30959425 DOI: 10.1016/j.copbio.2019.03.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/06/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022]
Abstract
The human intestinal tract harbors one of the most densely populated and open microbial ecosystems. The application of multi-omics approaches has provided insight into a wide array of complex interactions between the various groups of mainly anaerobic colonic microbes as well as the host-microbe dialogue. Integration of multi-omics techniques in cultivation based experiments that vary in complexity from monocultures to synthetic microbial communities identified key metabolic players in the trophic interactions as well as their ecological dynamics. A synergy between these approaches will be of utmost importance to reconstruct the functional interaction networks at the ecosystem level within the human intestinal microbiome. The improved understanding of microbiome functioning at ecosystem level will further aid in developing better predictive models and design of effective microbiome modulation strategies for health benefits.
Collapse
|
17
|
Lagkouvardos I, Lesker TR, Hitch TCA, Gálvez EJC, Smit N, Neuhaus K, Wang J, Baines JF, Abt B, Stecher B, Overmann J, Strowig T, Clavel T. Sequence and cultivation study of Muribaculaceae reveals novel species, host preference, and functional potential of this yet undescribed family. MICROBIOME 2019; 7:28. [PMID: 30782206 PMCID: PMC6381624 DOI: 10.1186/s40168-019-0637-2] [Citation(s) in RCA: 488] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/29/2019] [Indexed: 05/19/2023]
Abstract
BACKGROUND Bacteria within family S24-7 (phylum Bacteroidetes) are dominant in the mouse gut microbiota and detected in the intestine of other animals. Because they had not been cultured until recently and the family classification is still ambiguous, interaction with their host was difficult to study and confusion still exists regarding sequence data annotation. METHODS We investigated family S24-7 by combining data from large-scale 16S rRNA gene analysis and from functional and taxonomic studies of metagenomic and cultured species. RESULTS A total of 685 species was inferred by full-length 16S rRNA gene sequence clustering. While many species could not be assigned ecological habitats (93,045 samples analyzed), the mouse was the most commonly identified host (average of 20% relative abundance and nine co-occurring species). Shotgun metagenomics allowed reconstruction of 59 molecular species, of which 34 were representative of the 16S rRNA gene-derived species clusters. In addition, cultivation efforts allowed isolating five strains representing three species, including two novel taxa. Genome analysis revealed that S24-7 spp. are functionally distinct from neighboring families and versatile with respect to complex carbohydrate degradation. CONCLUSIONS We provide novel data on the diversity, ecology, and description of bacterial family S24-7, for which the name Muribaculaceae is proposed.
Collapse
Affiliation(s)
- Ilias Lagkouvardos
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Till R. Lesker
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas C. A. Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Eric J. C. Gálvez
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Nathiana Smit
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klaus Neuhaus
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Jun Wang
- Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - John F. Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany
- Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Birte Abt
- Leibniz-Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Center for Infection Research (DZIF), partner sites Hannover-Braunschweig and Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner sites Hannover-Braunschweig and Munich, Germany
| | - Jörg Overmann
- Leibniz-Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Center for Infection Research (DZIF), partner sites Hannover-Braunschweig and Munich, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Clavel
- ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
18
|
Shibayama J, Kuda T, Shikano A, Fukunaga M, Takahashi H, Kimura B, Ishizaki S. Effects of rice bran and fermented rice bran suspensions on caecal microbiota in dextran sodium sulphate-induced inflammatory bowel disease model mice. FOOD BIOSCI 2018. [DOI: 10.1016/j.fbio.2018.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
19
|
Yasushi Y, Haraguchi Y, Shikano A, Kuda T, Takahashi H, Kimura B. Induction of gut Lactobacillus reuteri
in normal ICR mice by oral administration of L. plantarum
AN1. J Food Biochem 2018. [DOI: 10.1111/jfbc.12589] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Yokota Yasushi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Yutaka Haraguchi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Ayane Shikano
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Takashi Kuda
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Hajime Takahashi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Bon Kimura
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| |
Collapse
|
20
|
Yokota Y, Shikano A, Kuda T, Takei M, Takahashi H, Kimura B. Lactobacillus plantarum AN1 cells increase caecal L. reuteri in an ICR mouse model of dextran sodium sulphate-induced inflammatory bowel disease. Int Immunopharmacol 2018; 56:119-127. [PMID: 29414641 DOI: 10.1016/j.intimp.2018.01.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/06/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
Abstract
To clarify the different effects of live and heat-killed probiotics on inflammatory bowel disease (IBD), the anti-inflammatory and protective effects of Lactobacillus plantarum AN1 cells isolated from the fermented fish aji-narezushi on murine macrophage RAW264.7 cells and in ICR mice with dextran sodium sulphate (DSS)-induced IBD were determined. L. plantarum AN1 cells showed anti-inflammatory activities in vitro, indicated by secretion of nitric oxide (NO) from RAW264.7 cells in the presence and absence of Escherichia coli 0111 lipopolysaccharide (LPS). L. plantarum AN1 cells also protected RAW264.7 cells against hydrogen peroxide toxicity. There was no difference between these effects in heat-killed and ultraviolet irradiation-killed cells. In the murine IBD model, both live and heat-killed L. plantarum AN1 cells via drinking water tended to ameliorate atrophy of colon length, mucosal tissue damage, and spleen enlargement. Amplicon sequencing of 16S rDNA (V4) revealed that both live and heat-killed AN1 cells increased abundance of indigenous lactic acid bacteria, particularly Lactobacillus reuteri. The results suggest that increased indigenous lactic acid bacterial abundance and feeding with L. plantarum AN1 cells synergistically improve effects against IBD.
Collapse
Affiliation(s)
- Yasushi Yokota
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-city, Tokyo 108-8477, Japan
| | - Ayane Shikano
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-city, Tokyo 108-8477, Japan
| | - Takashi Kuda
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-city, Tokyo 108-8477, Japan.
| | - Moemi Takei
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-city, Tokyo 108-8477, Japan
| | - Hajime Takahashi
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-city, Tokyo 108-8477, Japan
| | - Bon Kimura
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-city, Tokyo 108-8477, Japan
| |
Collapse
|
21
|
Clavel T, Neto JCG, Lagkouvardos I, Ramer-Tait AE. Deciphering interactions between the gut microbiota and the immune system via microbial cultivation and minimal microbiomes. Immunol Rev 2017; 279:8-22. [PMID: 28856739 PMCID: PMC5657458 DOI: 10.1111/imr.12578] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The community of microorganisms in the mammalian gastrointestinal tract, referred to as the gut microbiota, influences host physiology and immunity. The last decade of microbiome research has provided significant advancements for the field and highlighted the importance of gut microbes to states of both health and disease. Novel molecular techniques have unraveled the tremendous diversity of intestinal symbionts that potentially influence the host, many proof-of-concept studies have demonstrated causative roles of gut microbial communities in various pathologies, and microbiome-based approaches are beginning to be implemented in the clinic for diagnostic purposes or for personalized treatments. However, several challenges for the field remain: purely descriptive reports outnumbering mechanistic studies and slow translation of experimental results obtained in animal models into the clinics. Moreover, there is a dearth of knowledge regarding how gut microbes, including novel species that have yet to be identified, impact host immune responses. The sheer complexity of the gut microbial ecosystem makes it difficult, in part, to fully understand the microbiota-host networks that regulate immunity. In the present manuscript, we review key findings on the interactions between gut microbiota members and the immune system. Because culturing microbes allows performing functional studies, we have emphasized the impact of specific taxa or communities thereof. We also highlight underlying molecular mechanisms and discuss opportunities to implement minimal microbiome-based strategies.
Collapse
Affiliation(s)
- Thomas Clavel
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - João Carlos Gomes Neto
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Ilias Lagkouvardos
- ZIEL Institute for Food and Health, Core Facility Microbiome/NGS, Technical University of Munich, Germany
| | - Amanda E. Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|