1
|
Pickard JM, Porwollik S, Caballero-Flores G, Caruso R, Fukuda S, Soga T, Inohara N, McClelland M, Núñez G. Dietary amino acids regulate Salmonella colonization via microbiota-dependent mechanisms in the mouse gut. Nat Commun 2025; 16:4225. [PMID: 40335509 PMCID: PMC12058977 DOI: 10.1038/s41467-025-59706-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
The gut microbiota confers host protection against pathogen colonization early after infection. Several mechanisms underlying the protection have been described, but the contributions of nutrient competition versus direct inhibition are controversial. Using an ex vivo model of Salmonella growth in the mouse cecum with its indigenous microbes, we find that nutrient limitation and typical inhibitory factors alone cannot prevent pathogen growth. However, the addition of certain amino acids markedly reverses the microbiota's ability to suppress pathogen growth. Enhanced Salmonella colonization after antibiotic treatment is ablated by exclusion of dietary protein, which requires the presence of the microbiota. Thus, dietary protein and amino acids are important regulators of colonization resistance.
Collapse
Affiliation(s)
- Joseph M Pickard
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Steffen Porwollik
- Department of Microbiology and Molecular Genetics, University of California, Irvine, School of Medicine, Irvine, CA, USA
| | - Gustavo Caballero-Flores
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Roberta Caruso
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan
- Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Innovative Microbiome Therapy Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Naohiro Inohara
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, University of California, Irvine, School of Medicine, Irvine, CA, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Fletcher SJ, Ching C, Suprenant MP, Desai D, Zaman MH. Agent-based model of the human colon to investigate mechanisms of pathogen colonization resistance. Biophys J 2025; 124:1446-1461. [PMID: 40156186 DOI: 10.1016/j.bpj.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/31/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Recent global burden of disease studies have shown that bacterial infections are responsible for over 13 million deaths worldwide, or 1 in every 8 deaths, each year. Enteric diarrheal infections, in particular, pose a significant challenge and strain on healthcare systems as many are difficult to address pharmaceutically, and thus rely primarily on the patient's own immune system and gut microbiome to fight the infection. Nonetheless, the specific mechanisms behind gut microbiome colonization resistance of enteric pathogens are not well defined and microbiome diversity is difficult to represent and study experimentally. To address this gap, we have constructed an agent-based computational model of the colonic epithelium cross section to investigate the colonic invasion of enteric pathogens. The model focuses on three main regions: epithelial layer, mucosal bilayer, and adjacent lumen, and utilizes four main cell types as agents: anaerobic bacteria, facultative anaerobic bacteria, human goblet cells, and pathogens. Utilizing this model, we are able to describe the healthy microbiome cell localization and dynamics from our mucosal bilayer. In addition, we are also able to investigate the impact of host dietary fiber consumption and simulate pathogen invasion. The model exemplifies the possibility and potential to explore key gut microbiome colonization resistance mechanisms and environmental impacts on the gut microbiome using computational methods.
Collapse
Affiliation(s)
| | - Carly Ching
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | | | - Darash Desai
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Muhammad Hamid Zaman
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts; Howard Hughes Medical Institute, Boston University, Boston, Massachusetts; Center on Forced Displacement, Boston University, Boston, Massachusetts.
| |
Collapse
|
3
|
Laganenka L, Schubert C, Sichert A, Kalita I, Barthel M, Nguyen BD, Näf J, Lobriglio T, Sauer U, Hardt WD. Interplay between chemotaxis, quorum sensing, and metabolism regulates Escherichia coli-Salmonella Typhimurium interactions in vivo. PLoS Pathog 2025; 21:e1013156. [PMID: 40315408 PMCID: PMC12074654 DOI: 10.1371/journal.ppat.1013156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 05/13/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Motile bacteria use chemotaxis to navigate complex environments like the mammalian gut. These bacteria sense a range of chemoeffector molecules, which can either be of nutritional value or provide a cue for the niche best suited for their survival and growth. One such cue molecule is the intra- and interspecies quorum sensing signaling molecule, autoinducer-2 (AI-2). Apart from controlling collective behavior of Escherichia coli, chemotaxis towards AI-2 contributes to its ability to colonize the murine gut. However, the impact of AI-2-dependent niche occupation by E. coli on interspecies interactions in vivo is not fully understood. Using the C57BL/6J mouse infection model, we show that chemotaxis towards AI-2 contributes to nutrient competition and thereby affects colonization resistance conferred by E. coli against the enteric pathogen Salmonella enterica serovar Typhimurium (S. Tm). Like E. coli, S. Tm also relies on chemotaxis, albeit not towards AI-2, to compete against residing E. coli in a gut inflammation-dependent manner. Finally, utilizing a barcoded S. Tm mutant pool, we investigated the impact of AI-2 signaling in E. coli on carbohydrate utilization and central metabolism of S. Tm. Interestingly, AI-2-dependent niche colonization by E. coli was highly specific, impacting only a limited number of S. Tm mutants at distinct time points during infection. Notably, it significantly altered the fitness of mutants deficient in mannose utilization (ΔmanA, early stage infection) and, to a lesser extent, fumarate respiration (ΔdcuABC, late stage infection). The role of quorum sensing and chemotaxis in metabolic competition among bacteria remains largely unexplored. Here, we provide initial evidence that AI-2-dependent nutrient competition occurs between S. Tm and E. coli at specific time points during infection. These findings represent a crucial step toward understanding how bacteria navigate the gastrointestinal tract and engage in targeted nutrient competition within this complex three-dimensional environment.
Collapse
Affiliation(s)
- Leanid Laganenka
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | | | - Andreas Sichert
- Institute of Molecular Systems Biology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | - Irina Kalita
- Max Planck Institute for Terrestrial Microbiology and Center for Synthetic Microbiology, Marburg, Germany
| | - Manja Barthel
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | - Bidong D. Nguyen
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | - Jana Näf
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | - Thomas Lobriglio
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
4
|
Clavel T, Faber F, Groussin M, Haller D, Overmann J, Pauvert C, Poyet M, Selkrig J, Stecher B, Typas A, Vehreschild MJGT, Westermann AJ, Wylensek D, Maier L. Enabling next-generation anaerobic cultivation through biotechnology to advance functional microbiome research. Nat Biotechnol 2025:10.1038/s41587-025-02660-6. [PMID: 40301656 DOI: 10.1038/s41587-025-02660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Microbiomes are complex communities of microorganisms that are essential for biochemical processes on Earth and for the health of humans, animals and plants. Many environmental and host-associated microbiomes are dominated by anaerobic microbes, some of which cannot tolerate oxygen. Anaerobic microbial communities have been extensively studied over the last 20 years using molecular techniques, especially next-generation sequencing. However, there is a renewed interest in microbial cultivation because isolates provide the basis for understanding the taxonomic and functional units of biodiversity, elucidating novel biochemical pathways and the mechanisms underlying microbe-microbe and microbe-host interactions and opening new avenues for biotechnological and clinical applications. In this Perspective, we present areas of research and applications that will benefit from advancement in anaerobic microbial cultivation. We highlight key technical and infrastructural hurdles associated with the development and deployment of sophisticated cultivation workflows. Improving the performance of cultivation techniques will set new trends in functional microbiome research in the coming years.
Collapse
Affiliation(s)
- Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany.
| | - Franziska Faber
- Institute for Hygiene and Microbiology, Faculty of Medicine, University of Würzburg, Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Mathieu Groussin
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Jörg Overmann
- Leibniz-Institut DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- Microbiology, Technical University of Braunschweig, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Charlie Pauvert
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathilde Poyet
- Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Joel Selkrig
- Host-Microbe Interactomics Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Bärbel Stecher
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
- Chair of Intestinal Microbiome, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Athanasios Typas
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Maria J G T Vehreschild
- Goethe University Frankfurt, University Hospital Frankfurt, Department II of Internal Medicine, Infectious Diseases, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Alexander J Westermann
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Department of Microbiology, Biocentre, University of Würzburg, Würzburg, Germany
| | - David Wylensek
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Lisa Maier
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, University of Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
Lentsch V, Woller A, Rocker A, Aslani S, Moresi C, Ruoho N, Larsson L, Fattinger SA, Wenner N, Barazzone EC, Hardt WD, Loverdo C, Diard M, Slack E. Vaccine-enhanced competition permits rational bacterial strain replacement in the gut. Science 2025; 388:74-81. [PMID: 40179176 DOI: 10.1126/science.adp5011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 02/06/2025] [Indexed: 04/05/2025]
Abstract
Colonization of the intestinal lumen precedes invasive infection for a wide range of enteropathogenic and opportunistic pathogenic bacteria. We show that combining oral vaccination with engineered or selected niche-competitor strains permits pathogen exclusion and strain replacement in the mouse gut lumen. This approach can be applied either prophylactically to prevent invasion of nontyphoidal Salmonella strains, or therapeutically to displace an established Escherichia coli. Both intact adaptive immunity and metabolic niche competition are necessary for efficient vaccine-enhanced competition. Our findings imply that mucosal antibodies have evolved to work in the context of gut microbial ecology by influencing the outcome of competition. This has broad implications for the elimination of pathogenic and antibiotic-resistant bacterial reservoirs and for rational microbiota engineering.
Collapse
Affiliation(s)
- Verena Lentsch
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Medical Research Council (MRC) Translational Immunology Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Aurore Woller
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris, France
- Unité de Chronobiologie théorique, Faculté des Sciences, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Selma Aslani
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Claudia Moresi
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Niina Ruoho
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Louise Larsson
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Stefan A Fattinger
- Institute for Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | | | - Wolf-Dietrich Hardt
- Institute for Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris, France
| | - Médéric Diard
- Biozentrum, University of Basel, Basel, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Emma Slack
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Lee S, Wischmeyer PE, Mintz CD, Serbanescu MA. Recent Insights into the Evolving Role of the Gut Microbiome in Critical Care. Crit Care Clin 2025; 41:379-396. [PMID: 40021286 DOI: 10.1016/j.ccc.2024.11.002] [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: 03/03/2025]
Abstract
This review explores the evolving understanding of gut microbiota's role in critical illness, focusing on how acute illness and exposures in intensive care unit (ICU) environment negatively impact the gut microbiota and the implications of these changes on host responses in critically-ill patients. Focusing on recent findings from clinical and preclinical studies, we discuss the effects of inflammation, enteral nutrient deprivation, and antibiotics on gut microbial dynamics. This review aims to enhance comprehension of microbial dynamics in the ICU and their implications for clinical outcomes and therapeutic strategies.
Collapse
Affiliation(s)
- Seoho Lee
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Paul E Wischmeyer
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA
| | - Cyrus D Mintz
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Mara A Serbanescu
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA.
| |
Collapse
|
7
|
Xu T, Fang D, Xu T, Tao X, Wang Z, Liu Y. Exercise-driven gut microbiota alterations enhance colonization resistance against methicillin-resistant Staphylococcus aureus. Cell Rep 2025; 44:115424. [PMID: 40080501 DOI: 10.1016/j.celrep.2025.115424] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Gut microbiota plays a crucial role in resisting the invasion of pathogens, particularly multidrug-resistant (MDR) bacteria, which pose a significant threat to public health. While exercise offers numerous health benefits, its impact on host colonization resistance remains largely unclear. In this study, we demonstrate that moderate exercise significantly reduces gut colonization by methicillin-resistant Staphylococcus aureus (MRSA), a clinically important MDR pathogen. Moreover, we identify an understudied strain of the intestinal probiotic Dubosiella newyorkensis (L8) as a critical factor in mediating exercise-induced colonization resistance against MRSA. Mechanistically, L8 enhances the deprivation of fucose, a crucial carbon source essential for MRSA growth and pathogenicity. This process relies on the high binding affinity of pyruvate to the ILE257 site of the lactate dehydrogenase in L8. Overall, our work highlights the importance of moderate exercise in maintaining host colonization resistance and demonstrates L8 as a probiotic in protecting against MRSA colonization.
Collapse
Affiliation(s)
- Tingting Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Dan Fang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiuying Tao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
8
|
Shealy NG, Baltagulov M, de Brito C, McGovern A, Castro P, Schrimpe-Rutledge AC, Malekshahi C, Condreanu SG, Sherrod SD, Jana S, Jones K, Ribeiro TM, McLean JA, Beiting DP, Byndloss MX. Short-term alterations in dietary amino acids override host genetic susceptibility and reveal mechanisms of Salmonella Typhimurium small intestine colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645332. [PMID: 40196486 PMCID: PMC11974825 DOI: 10.1101/2025.03.25.645332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
In addition to individual genetics, environmental factors (e.g., dietary changes) may influence host susceptibility to gastrointestinal infection through unknown mechanisms. Herein, we developed a model in which CBA/J mice, a genetically resistant strain that tolerates intestinal colonization by the enteric pathogen Salmonella Typhimurium (S. Tm), rapidly succumb to infection after exposure to a diet rich in L-amino acids (AA). In mice, S. Tm-gastroenteritis is restricted to the large intestine (cecum), limiting their use to understand S. Tm small intestine (ileum) colonization, a feature of human Salmonellosis. Surprisingly, CBA mice fed AA diet developed ileitis with enhanced S. Tm ileal colonization. Using germ-free mice and ileal-fecal slurry transplant, we found diet-mediated S. Tm ileal expansion to be microbiota-dependent. Mechanistically, S. Tm relied on Fructosyl-asparagine utilization to expand in the ileum during infection. We demonstrate how AA diet overrides host genetics by altering the gut microbiota's ability to prevent S. Tm ileal colonization.
Collapse
Affiliation(s)
- Nicolas G. Shealy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Madi Baltagulov
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Camila de Brito
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Anna McGovern
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Pollyana Castro
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | | | - Clara Malekshahi
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, U. S. A
| | - Simona G. Condreanu
- Center for Innovative Technology and Department of Chemistry, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Stacy D. Sherrod
- Center for Innovative Technology and Department of Chemistry, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Somnath Jana
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Katerina Jones
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Tamara Machado Ribeiro
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - John A. McLean
- Center for Innovative Technology and Department of Chemistry, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Daniel P. Beiting
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Mariana X. Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
- Vanderbilt Institute of Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, TN 37235, U.S.A
| |
Collapse
|
9
|
Kroon S, Malcic D, Weidert L, Bircher L, Boldt L, Christen P, Kiefer P, Sintsova A, Nguyen BD, Barthel M, Steiger Y, Clerc M, Herzog MKM, Chen C, Gül E, Guery B, Slack E, Sunagawa S, Vorholt JA, Maier L, Lacroix C, Hausmann A, Hardt WD. Sublethal systemic LPS in mice enables gut-luminal pathogens to bloom through oxygen species-mediated microbiota inhibition. Nat Commun 2025; 16:2760. [PMID: 40113753 PMCID: PMC11926250 DOI: 10.1038/s41467-025-57979-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Endotoxin-driven systemic immune activation is a common hallmark across various clinical conditions. During acute critical illness, elevated plasma lipopolysaccharide triggers non-specific systemic immune activation. In addition, a compositional shift in the gut microbiota, including an increase in gut-luminal opportunistic pathogens, is observed. Whether a causal link exists between acute endotoxemia and abundance of gut-luminal opportunistic pathogens is incompletely understood. Here, we model acute, pathophysiological lipopolysaccharide concentrations in mice and show that systemic exposure promotes a 100-10'000-fold expansion of Klebsiella pneumoniae, Escherichia coli, Enterococcus faecium and Salmonella Typhimurium in the gut within one day, without overt enteropathy. Mechanistically, this is driven by a Toll-like receptor 4-dependent increase in gut-luminal oxygen species levels, which transiently halts microbiota fermentation and fuels growth of gut-luminal facultative anaerobic pathogens through oxidative respiration. Thus, systemic immune activation transiently perturbs microbiota homeostasis and favours opportunistic pathogens, potentially increasing the risk of infection in critically ill patients.
Collapse
Affiliation(s)
- Sanne Kroon
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Dejan Malcic
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Lena Weidert
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Lea Bircher
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Leonardo Boldt
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
- M3 Research Center for Malignome, Metabolome and Microbiome, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Melanie Clerc
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Mathias K-M Herzog
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Carmen Chen
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Benoit Guery
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Lisa Maier
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
- M3 Research Center for Malignome, Metabolome and Microbiome, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
| | - Christophe Lacroix
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Annika Hausmann
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
- reNEW - Novo Nordisk Foundation Center for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
10
|
Walling LK, Gamache MH, González-Pech RA, Harwood VJ, Ibrahim-Hashim A, Jung JH, Lewis DB, Margres MJ, McMinds R, Rasheed K, Reis F, van Riemsdijk I, Santiago-Alarcon D, Sarmiento C, Whelan CJ, Zalamea PC, Parkinson JE, Richards CL. Incorporating microbiome analyses can enhance conservation of threatened species and ecosystem functions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 970:178826. [PMID: 40054249 DOI: 10.1016/j.scitotenv.2025.178826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 03/17/2025]
Abstract
Conservation genomics is a rapidly growing subdiscipline of conservation biology that uses genome-wide information to inform management of biodiversity at all levels. Such efforts typically focus on species or systems of conservation interest, but rarely consider associated microbes. At least three major approaches have been used to study how microorganisms broadly contribute to conservation areas: (1) diversity surveys map out microbial species distribution patterns in a variety of hosts, natural environments or regions; (2) functional surveys associate microbial communities with factors of interest, such as host health, symbiotic interactions, environmental characteristics, ecosystem processes, and biological invasions; and (3) manipulative experiments examine the response of changes to microbial communities or determine the functional roles of specific microbes within hosts or communities by adding, removing, or genetically modifying microbes. In practice, multiple approaches are often applied simultaneously. The results from all three conservation genomics approaches can be used to help design practical interventions and improve management actions, some of which we highlight below. However, experimental manipulations allow for more robust causal inferences and should be the ultimate goal of future work. Here we discuss how further integration of microbial research of a host's microbiome and of free living microbes into conservation biology will be an essential advancement for conservation of charismatic organisms and ecosystem functions in light of ongoing global environmental change.
Collapse
Affiliation(s)
| | - Matthew H Gamache
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA
| | - Raúl A González-Pech
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA; Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Valerie J Harwood
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA
| | - Arig Ibrahim-Hashim
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA; Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Faculty of Education and Arts, Sohar University, Sohar, Oman
| | - Jun Hee Jung
- Plant Evolutionary Ecology Group, University of Tübingen, Tübingen, Germany
| | - David B Lewis
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA
| | - Mark J Margres
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA
| | - Ryan McMinds
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA; Center for Global Health and Interdisciplinary Research (GHIDR), University of South Florida, Tampa, FL, USA; Northwest Indian Fisheries Commission
| | - Kiran Rasheed
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Frank Reis
- Plant Evolutionary Ecology Group, University of Tübingen, Tübingen, Germany
| | - Isolde van Riemsdijk
- Plant Evolutionary Ecology Group, University of Tübingen, Tübingen, Germany; Biodiversity and Evolution, Lund University, Lund, Sweden
| | | | - Carolina Sarmiento
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA; Smithsonian Tropical Research Institute, Panama City, Republic of Panama
| | - Christopher J Whelan
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Paul-Camilo Zalamea
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA; Smithsonian Tropical Research Institute, Panama City, Republic of Panama
| | | | - Christina L Richards
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA; Plant Evolutionary Ecology Group, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
11
|
Cherrak Y, Younes AA, Perez-Molphe-Montoya E, Maurer L, Yilmaz K, Enz U, Zeder C, Kiefer P, Christen P, Gül E, Vorholt JA, von Mering C, Hardt WD. Neutrophil recruitment during intestinal inflammation primes Salmonella elimination by commensal E. coli in a context-dependent manner. Cell Host Microbe 2025; 33:358-372.e4. [PMID: 40023150 DOI: 10.1016/j.chom.2025.02.004] [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: 08/16/2024] [Revised: 12/19/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025]
Abstract
Foodborne bacterial diarrhea involves complex pathogen-microbiota-host interactions. Pathogen-displacing probiotics are increasingly popular, but heterogeneous patient outcomes highlighted the need to understand individualized host-probiotic activity. Using the mouse gut commensal Escherichia coli 8178 and the human probiotic E. coli Nissle 1917, we found that the degree of protection against the enteric pathogen Salmonella enterica serovar Typhimurium (S. Tm) varies across mice with distinct gut microbiotas. Pathogen clearance is linked to enteropathy severity and subsequent recruitment of intraluminal neutrophils, which differs in a microbiota-dependent manner. By combining mouse knockout and antibody-mediated depletion models with bacterial genetics, we show that neutrophils and host-derived reactive oxygen species directly influence E. coli-mediated S. Tm displacement by potentiating siderophore-bound toxin killing. Our work demonstrates how host immune factors shape pathogen-displacing probiotic efficiency while also revealing an unconventional antagonistic interaction where a gut commensal and the host synergize to displace an enteric pathogen.
Collapse
Affiliation(s)
- Yassine Cherrak
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| | - Andrew Abi Younes
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Eugenio Perez-Molphe-Montoya
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Luca Maurer
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Koray Yilmaz
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ursina Enz
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Christophe Zeder
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Science and Technology, 8092 Zurich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
12
|
Vayena E, Fuchs L, Peyhani HM, Lagoda K, Nguyen B, Hardt WD, Hatzimanikatis V. Metabolic network reconstruction as a resource for analyzing Salmonella Typhimurium SL1344 growth in the mouse intestine. PLoS Comput Biol 2025; 21:e1012869. [PMID: 40067815 PMCID: PMC11925469 DOI: 10.1371/journal.pcbi.1012869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 03/20/2025] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Nontyphoidal Salmonella strains (NTS) are among the most common foodborne enteropathogens and constitute a major cause of global morbidity and mortality, imposing a substantial burden on global health. The increasing antibiotic resistance of NTS bacteria has attracted a lot of research on understanding their modus operandi during infection. Growth in the gut lumen is a critical phase of the NTS infection. This might offer opportunities for intervention. However, the metabolic richness of the gut lumen environment and the inherent complexity and robustness of the metabolism of NTS bacteria call for modeling approaches to guide research efforts. In this study, we reconstructed a thermodynamically constrained and context-specific genome-scale metabolic model (GEM) for S. Typhimurium SL1344, a model strain well-studied in infection research. We combined sequence annotation, optimization methods and in vitro and in vivo experimental data. We used GEM to explore the nutritional requirements, the growth limiting metabolic genes, and the metabolic pathway usage of NTS bacteria in a rich environment simulating the murine gut. This work provides insight and hypotheses on the biochemical capabilities and requirements of SL1344 beyond the knowledge acquired through conventional sequence annotation and can inform future research aimed at better understanding NTS metabolism and identifying potential targets for infection prevention.
Collapse
Affiliation(s)
- Evangelia Vayena
- Laboratory of Computational Systems Biotechnology, EPFL, Lausanne, Switzerland
| | - Lea Fuchs
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | | | - Konrad Lagoda
- Laboratory of Computational Systems Biotechnology, EPFL, Lausanne, Switzerland
| | - Bidong Nguyen
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
13
|
Jans M, Vereecke L. A guide to germ-free and gnotobiotic mouse technology to study health and disease. FEBS J 2025; 292:1228-1251. [PMID: 38523409 DOI: 10.1111/febs.17124] [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: 10/30/2023] [Revised: 01/17/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
The intestinal microbiota has major influence on human physiology and modulates health and disease. Complex host-microbe interactions regulate various homeostatic processes, including metabolism and immune function, while disturbances in microbiota composition (dysbiosis) are associated with a plethora of human diseases and are believed to modulate disease initiation, progression and therapy response. The vast complexity of the human microbiota and its metabolic output represents a great challenge in unraveling the molecular basis of host-microbe interactions in specific physiological contexts. To increase our understanding of these interactions, functional microbiota research using animal models in a reductionistic setting are essential. In the dynamic landscape of gut microbiota research, the use of germ-free and gnotobiotic mouse technology, in which causal disease-driving mechanisms can be dissected, represents a pivotal investigative tool for functional microbiota research in health and disease, in which causal disease-driving mechanisms can be dissected. A better understanding of the health-modulating functions of the microbiota opens perspectives for improved therapies in many diseases. In this review, we discuss practical considerations for the design and execution of germ-free and gnotobiotic experiments, including considerations around germ-free rederivation and housing conditions, route and timing of microbial administration, and dosing protocols. This comprehensive overview aims to provide researchers with valuable insights for improved experimental design in the field of functional microbiota research.
Collapse
Affiliation(s)
- Maude Jans
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
| |
Collapse
|
14
|
Beresford-Jones BS, Suyama S, Clare S, Soderholm A, Xia W, Sardar P, Lee J, Harcourt K, Lawley TD, Pedicord VA. Enterocloster clostridioformis protects against Salmonella pathogenesis and modulates epithelial and mucosal immune function. MICROBIOME 2025; 13:61. [PMID: 40022210 PMCID: PMC11869688 DOI: 10.1186/s40168-025-02050-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Promoting resistance to enteric pathogen infection is a core function of the gut microbiota; however, many of the specific host-commensal interactions that mediate this protection remain uncharacterised. To address this knowledge gap, we monocolonised germ-free mice with mouse-derived commensal microbes to screen for microbiota-induced resistance to Salmonella Typhimurium infection. RESULTS We identified Enterocloster clostridioformis as a protective species against S. Typhimurium infection. E. clostridioformis selectively upregulates resistin-like molecule β and cell cycle pathway expression at the level of caecal epithelial cells and increases T-regulatory cells in the underlying mucosal immune system, potentially contributing to reduced infection-induced pathology. CONCLUSIONS We highlight novel mechanisms of host-microbe interactions that can mediate microbiota-induced resistance to acute salmonellosis. In the backdrop of increasing antibiotic resistance, this study identifies novel potential avenues for further research into protective host responses against enteric infections and could lead to new therapeutic approaches. Video Abstract.
Collapse
Affiliation(s)
- Benjamin S Beresford-Jones
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Satoshi Suyama
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Simon Clare
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Amelia Soderholm
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Wangmingyu Xia
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Puspendu Sardar
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Junhee Lee
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Katherine Harcourt
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Trevor D Lawley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Virginia A Pedicord
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
15
|
Schubert C, Nguyen BD, Sichert A, Näpflin N, Sintsova A, Feer L, Näf J, Daniel BBJ, Steiger Y, von Mering C, Sauer U, Hardt WD. Monosaccharides drive Salmonella gut colonization in a context-dependent or -independent manner. Nat Commun 2025; 16:1735. [PMID: 39966379 PMCID: PMC11836396 DOI: 10.1038/s41467-025-56890-y] [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: 08/16/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The carbohydrates that fuel gut colonization by S. Typhimurium are not fully known. To investigate this, we designed a quality-controlled mutant pool to probe the metabolic capabilities of this enteric pathogen. Using neutral genetic barcodes, we tested 35 metabolic mutants across five different mouse models with varying microbiome complexities, allowing us to differentiate between context-dependent and context-independent nutrient sources. Results showed that S. Typhimurium uses D-mannose, D-fructose and likely D-glucose as context-independent carbohydrates across all five mouse models. The utilization of D-galactose, N-acetylglucosamine and hexuronates, on the other hand, was context-dependent. Furthermore, we showed that D-fructose is important in strain-to-strain competition between Salmonella serovars. Complementary experiments confirmed that D-glucose, D-fructose, and D-galactose are excellent niches for S. Typhimurium to exploit during colonization. Quantitative measurements revealed sufficient amounts of carbohydrates, such as D-glucose or D-galactose, in the murine cecum to drive S. Typhimurium colonization. Understanding these key substrates and their context-dependent or -independent use by enteric pathogens will inform the future design of probiotics and therapeutics to prevent diarrheal infections such as non-typhoidal salmonellosis.
Collapse
Affiliation(s)
- Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | - Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Nicolas Näpflin
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Lilith Feer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Benjamin B J Daniel
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
Yang JL, Zhu H, Sadh P, Aumiller K, Guvener ZT, Ludington WB. Commensal acidification of specific gut regions produces a protective priority effect against enteropathogenic bacterial infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637843. [PMID: 39990475 PMCID: PMC11844456 DOI: 10.1101/2025.02.12.637843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The commensal microbiome has been shown to protect against newly introduced enteric pathogens in multiple host species, a phenomenon known as a priority effect. Multiple mechanisms can contribute to this protective priority effect, including antimicrobial compounds, nutrient competition, and pH changes. In Drosophila melanogaster , Lactiplantibacillus plantarum has been shown to protect against enteric pathogens. However, the strains of L. plantarum studied were derived from laboratory flies or non-fly environments and have been found to be unstable colonizers of the fly gut that mainly reside on the food. To study the priority effect using a naturally occurring microbial relationship, we isolated a wild-fly derived strain of L. plantarum that stably colonizes the fly gut in conjunction with a common enteric pathogen, Serratia marcescens . Flies stably associated with the L. plantarum strain were more resilient to oral Serratia marcescens infection as seen by longer lifespan and lower S. marcescens load in the gut. Through in vitro experiments, we found that L. plantarum inhibits S. marcescens growth due to acidification. We used gut imaging with pH-indicator dyes to show that L. plantarum reduces the gut pH to levels that restrict S. marcescens growth in vivo . In flies colonized with L. plantarum prior to S. marcescens infection, L. plantarum and S. marcescens are spatially segregated in the gut and S. marcescens is less abundant where L. plantarum heavily colonizes, indicating that acidification of specific gut regions is a mechanism of a protective priority effect.
Collapse
|
17
|
Yin Q, da Silva AC, Zorrilla F, Almeida AS, Patil KR, Almeida A. Ecological dynamics of Enterobacteriaceae in the human gut microbiome across global populations. Nat Microbiol 2025; 10:541-553. [PMID: 39794474 PMCID: PMC11790488 DOI: 10.1038/s41564-024-01912-6] [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: 07/30/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025]
Abstract
Gut bacteria from the Enterobacteriaceae family are a major cause of opportunistic infections worldwide. Given their prevalence among healthy human gut microbiomes, interspecies interactions may play a role in modulating infection resistance. Here we uncover global ecological patterns linked to Enterobacteriaceae colonization and abundance by leveraging a large-scale dataset of 12,238 public human gut metagenomes spanning 45 countries. Machine learning analyses identified a robust gut microbiome signature associated with Enterobacteriaceae colonization status, consistent across health states and geographic locations. We classified 172 gut microbial species as co-colonizers and 135 as co-excluders, revealing a genus-wide signal of colonization resistance within Faecalibacterium and strain-specific co-colonization patterns of the underexplored Faecalimonas phoceensis. Co-exclusion is linked to functions involved in short-chain fatty acid production, iron metabolism and quorum sensing, while co-colonization is linked to greater functional diversity and metabolic resemblance to Enterobacteriaceae. Our work underscores the critical role of the intestinal environment in the colonization success of gut-associated opportunistic pathogens with implications for developing non-antibiotic therapeutic strategies.
Collapse
Affiliation(s)
- Qi Yin
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- College of Public Health, Chongqing Medical University, Chongqing, China
| | - Ana C da Silva
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Francisco Zorrilla
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Ana S Almeida
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Kiran R Patil
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Alexandre Almeida
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
18
|
Bray AS, Broberg CA, Hudson AW, Wu W, Nagpal RK, Islam M, Valencia-Bacca JD, Shahid F, Hernandez GE, Nutter NA, Walker KA, Bennett EF, Young TM, Barnes AJ, Ornelles DA, Miller VL, Zafar MA. Klebsiella pneumoniae employs a type VI secretion system to overcome microbiota-mediated colonization resistance. Nat Commun 2025; 16:940. [PMID: 39843522 PMCID: PMC11754592 DOI: 10.1038/s41467-025-56309-8] [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: 02/13/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Microbial species must compete for space and nutrients to persist in the gastrointestinal (GI) tract, and our understanding of the complex pathobiont-microbiota interactions is far from complete. Klebsiella pneumoniae, a problematic, often drug-resistant nosocomial pathogen, can colonize the GI tract asymptomatically, serving as an infection reservoir. To provide insight on how K. pneumoniae interacts with the resident gut microbiome, we conduct a transposon mutagenesis screen using a murine model of GI colonization with an intact microbiota. Among the genes identified were those encoding a type VI secretion system (T6SS), which mediates contact-dependent killing of gram-negative bacteria. From several approaches, we demonstrate that the T6SS is critical for K. pneumoniae gut colonization. Metagenomics and in vitro killing assays reveal that K. pneumoniae reduces Betaproteobacteria species in a T6SS-dependent manner, thus identifying specific species targeted by K. pneumoniae. We further show that T6SS gene expression is controlled by several transcriptional regulators and that expression only occurs in vitro under conditions that mimic the gut environment. By enabling K. pneumoniae to thrive in the gut, the T6SS indirectly contributes to the pathogenic potential of this organism. These observations advance our molecular understanding of how K. pneumoniae successfully colonizes the GI tract.
Collapse
Affiliation(s)
- Andrew S Bray
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Christopher A Broberg
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Andrew W Hudson
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Weisheng Wu
- BRCF Bioinformatics Core, University of Michigan, Ann Arbor, MI, USA
| | - Ravinder K Nagpal
- Department of Nutrition & Integrative Physiology, Florida State University College of Health and Human Sciences, Tallahassee, FL, USA
| | - Maidul Islam
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Juan D Valencia-Bacca
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Fawaz Shahid
- Wake Forest University, Winston Salem, Winston Salem, NC, USA
| | - Giovanna E Hernandez
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Noah A Nutter
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Kimberly A Walker
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Emma F Bennett
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Taylor M Young
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Andrew J Barnes
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - David A Ornelles
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Virginia L Miller
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - M Ammar Zafar
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, NC, USA.
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
19
|
Xu Q, Zhao Y, Yuan P, Ma X, Wang S, Li L, Cheng P, Qu S. Functionalized Microsphere Platform Combining Nutrient Restriction and Combination Therapy to Combat Bacterial Infections. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2966-2976. [PMID: 39744763 DOI: 10.1021/acsami.4c16610] [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: 01/18/2025]
Abstract
The escalating prevalence of multidrug-resistant (MDR) bacterial infections has emerged as a critical global health crisis, undermining the efficacy of conventional antibiotic therapies. This pressing challenge necessitates the development of innovative strategies to combat MDR pathogens. Advances in multifunctional drug delivery systems offer promising solutions to reduce or eradicate MDR bacteria. Inspired by the fact that the growth of bacteria requires essential nutrients, core-shell porous poly(lactic-co-glycolic acid) (PLGA) microspheres coated with pH-responsive polydopamine (PDA) were fabricated to improve delivery, resulting in enhanced efficacy through nutrient restriction and combination therapy. The PDA chelates iron ions in the environment, preventing bacteria from absorbing iron and thus suppressing their growth and proliferation. Subsequently, the released antibiotics from the porous PLGA core, rifampicin and polymyxin B, accelerate bacterial eradication by disrupting their inner and outer membrane structures. Such a multifunctional microsphere platform clears 99% Salmonella Typhimurium in 4 h and shows increased efficiency in a lethal intestinal infection model in mice. These findings provide a drug delivery system that integrates bacterial nutrient restriction and antibiotic killing, highlighting the potential of targeting bacterial iron regulation as a strategy for developing new antimicrobial delivery systems to address MDR bacterial infections.
Collapse
Affiliation(s)
- Qingjun Xu
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Yang Zhao
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Pingping Yuan
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Xuan Ma
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Sheng Wang
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Ping Cheng
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
20
|
Chen Z, Yang Y, Chen X, Bei C, Gao Q, Chao Y, Wang C. An RNase III-processed sRNA coordinates sialic acid metabolism of Salmonella enterica during gut colonization. Proc Natl Acad Sci U S A 2025; 122:e2414563122. [PMID: 39792291 PMCID: PMC11745405 DOI: 10.1073/pnas.2414563122] [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: 07/21/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025] Open
Abstract
Sialic acids derived from colonic mucin glycans are crucial nutrients for enteric bacterial pathogens like Salmonella. The uptake and utilization of sialic acid in Salmonella depend on coordinated regulons, each activated by specific metabolites at the transcriptional level. However, the mechanisms enabling crosstalk among these regulatory circuits to synchronize gene expression remain poorly understood. Here, we identify ManS, a small noncoding RNA derived from the 3' UTR of STM1128 mRNA transcribed from a Salmonella enterica-specific genetic locus, as an important posttranscriptional regulator coordinating sialic acid metabolism regulons. ManS is primarily processed by RNase III and, along with its parental transcripts, is specifically activated by N-acetylmannosamine (ManNAc), the initial degradation product of sialic acid. We found that the imperfect stem-loop structure at the 5' end of ManS allows RNase III to cleave in a noncanonical manner, generating two functional types of ManS with the assistance of RNase E and other RNases: short isoforms with a single seed region that regulate the uptake of N-acetylglucosamine, an essential intermediate in sialic acid metabolism; and long isoforms with an additional seed region that regulate multiple genes involved in central and secondary metabolism. This sophisticated regulation by ManS significantly impacts ManNAc metabolism and S. enterica's competitive behavior during infection. Our findings highlight the role of sRNA in coordinating transcriptional circuits and advance our understanding of RNase III-mediated processing of 3' UTR-derived sRNAs, underscoring the important role of ManNAc in Salmonella adaptation within host environments.
Collapse
Affiliation(s)
- Ziying Chen
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai200031, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Yaomei Yang
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Xiaomin Chen
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Cheng Bei
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Qian Gao
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Yanjie Chao
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai200031, China
| | - Chuan Wang
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| |
Collapse
|
21
|
Virgo M, Mostowy S, Ho BT. Emerging models to study competitive interactions within bacterial communities. Trends Microbiol 2025:S0966-842X(24)00325-1. [PMID: 39799088 DOI: 10.1016/j.tim.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/15/2025]
Abstract
Within both abiotic and host environments, bacteria typically exist as diverse, multispecies communities and have crucial roles in human health, agriculture, and industry. In these communities, bacteria compete for resources, and these competitive interactions can shape the overall population structure and community function. Studying bacterial community dynamics requires experimental model systems that capture the different interaction networks between bacteria and their surroundings. We examine the recent literature advancing such systems, including (i) in silico models establishing the theoretical basis for how cell-to-cell interactions can influence population level dynamics, (ii) in vitro models characterizing specific interbacterial interactions, (iii) organ-on-a-chip models revealing the physiologically relevant parameters, such as spatial structure and mechanical forces, that bacteria encounter within a host, and (iv) in vivo plant and animal models connecting the host responses to interbacterial interactions. Each of these systems has greatly contributed to our understanding of bacterial community dynamics and can be used synergistically to understand how bacterial competition influences population architecture.
Collapse
Affiliation(s)
- Mollie Virgo
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, UK; Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK.
| | - Brian T Ho
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, UK; Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK.
| |
Collapse
|
22
|
Almási ÉDH, Eisenhard L, Osbelt L, Lesker TR, Vetter AC, Knischewski N, Bielecka AA, Gronow A, Muthukumarasamy U, Wende M, Tawk C, Neumann-Schaal M, Brönstrup M, Strowig T. Klebsiella oxytoca facilitates microbiome recovery via antibiotic degradation and restores colonization resistance in a diet-dependent manner. Nat Commun 2025; 16:551. [PMID: 39789003 PMCID: PMC11717976 DOI: 10.1038/s41467-024-55800-y] [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: 03/28/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Competition among bacteria for carbohydrates is pivotal for colonization resistance (CR). However, the impact of Western-style diets on CR remains unclear. Here we show how the competition between Klebsiella oxytoca and Klebsiella pneumoniae is modulated by consuming one of three Western-style diets characterized by high-starch, high-sucrose, or high-fat/high-sucrose content. In vivo competition experiments in ampicillin-treated mice reveal that K. oxytoca promotes K. pneumoniae decolonization on all dietary backgrounds. However, mice on the high-fat/high-sucrose diet show reduced pathogen clearance. Microbiome analysis reveals that the combination of Western-style diets and ampicillin treatment synergize in microbiome impairment, particularly noticeable in the presence of high dietary fat content. The diet-independent degradation of ampicillin in the gut lumen by K. oxytoca beta-lactamases facilitates rapid commensal outgrowth, which is required for subsequent pathogen clearance. Our findings provide insights into how diet modulates functional microbiome recovery and K. oxytoca-mediated pathogen elimination from the gut.
Collapse
Affiliation(s)
- Éva D H Almási
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Lea Eisenhard
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Lisa Osbelt
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Anna C Vetter
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Nele Knischewski
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Agata Anna Bielecka
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Achim Gronow
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Uthayakumar Muthukumarasamy
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Caroline Tawk
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Meina Neumann-Schaal
- Bacterial Metabolomics, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
- Center for Individualized Infection Medicine, Hannover, Germany.
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany.
| |
Collapse
|
23
|
Dong Q, Harper S, McSpadden E, Son SS, Allen MM, Lin H, Smith RC, Metcalfe C, Burgo V, Woodson C, Sundararajan A, Rose A, McMillin M, Moran D, Little J, Mullowney MW, Sidebottom AM, Fortier LC, Shen A, Pamer EG. Protection against Clostridioides difficile disease by a naturally avirulent strain. Cell Host Microbe 2025; 33:59-70.e4. [PMID: 39610252 PMCID: PMC11731898 DOI: 10.1016/j.chom.2024.11.003] [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: 05/15/2024] [Revised: 09/24/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024]
Abstract
Clostridioides difficile is a leading cause of healthcare infections. Gut dysbiosis promotes C. difficile infection (CDI) and CDIs promote gut dysbiosis, leading to frequent CDI recurrence. Although therapies preventing recurrent CDI have been developed, including live biotherapeutic products, existing therapies are costly and do not prevent primary infections. Here, we show that an avirulent C. difficile isolate, ST1-75, protects mice from developing colitis induced by a virulent R20291 strain when coinfected at a 1:1 ratio. In metabolic analyses, avirulent ST1-75 depletes amino acids more rapidly than virulent R20291 and supplementation with amino acids ablates this competitive advantage, indicating that ST1-75 limits the growth of virulent R20291 through amino acid depletion. Overall, our study identifies inter-strain nutrient depletion as a potentially exploitable mechanism to reduce the incidence of CDI and reveals that the ST1-75 strain may be a biotherapeutic agent that can prevent CDI in high-risk patients.
Collapse
Affiliation(s)
- Qiwen Dong
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA.
| | - Stephen Harper
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Emma McSpadden
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Sophie S Son
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
| | - Marie-Maude Allen
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Huaiying Lin
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Rita C Smith
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Carolyn Metcalfe
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Victoria Burgo
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Che Woodson
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | | | - Amber Rose
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Mary McMillin
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - David Moran
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Jessica Little
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | | | | | - Louis-Charles Fortier
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Aimee Shen
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Eric G Pamer
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
24
|
Cruz-Cruz A, Schreeg ME, Gunn JS. A temporary cholesterol-rich diet and bacterial extracellular matrix factors favor Salmonella spp. biofilm formation in the cecum. mBio 2025; 16:e0324224. [PMID: 39636114 PMCID: PMC11708031 DOI: 10.1128/mbio.03242-24] [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: 10/17/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Asymptomatic chronic carriers occur in approximately 5% of humans infected with Salmonella enterica serovar Typhi (S. Typhi) and represent a critical reservoir for bacterial dissemination. While chronic carriage primarily occurs in the gallbladder (GB) through biofilms on gallstones, additional anatomic sites have been suggested that could also harbor Salmonella. S. Typhimurium, orally infected 129 × 1/SvJ mice were pre-treated with a cholesterol-rich diet as a gallstone model for chronic carriage. We observed S. Typhimurium in feces and the cecum during early and persistent infection. Furthermore, bacterial biofilm-like aggregates were associated with the cecum epithelium at 7 and 21 days post-infection (DPI) in mice on a lithogenic diet (Ld) and correlated with an increase in cecal cholesterol at 21 DPI. Salmonella's extracellular matrix (ECM) was demonstrated as important in colonizing the cecum, as survival and aggregate formation significantly decreased when mice were infected with a quadruple ECM mutant strain. Gallbladder Salmonella counts were low at 36 DPI while cecal Salmonella were high, suggesting that gallbladder colonization was likely not responsible for the high cecal burden. All cecum phenotypes were significantly diminished in mice fed a normal diet (Nd). Finally, we examined the capability of S. Typhi to colonize the cecum and showed S. Typhi in feces and in aggregates in the cecum up to 7 DPI, with slightly higher counts in mice fed a Ld compared to Nd. Our findings suggest that the cecum, particularly under cholesterol-rich conditions, serves as an adaptative niche for Salmonella spp. aggregates/biofilms and is a putative site for long-term infection.IMPORTANCETyphoid fever is a systemic infectious disease triggered by the gastrointestinal dissemination of Salmonella Typhi and Paratyphi in humans. Three to five percent of infected individuals become chronic carriers, a state in which gallstone biofilm formation facilitates spread of the bacteria in feces. Notably, surgical removal of the gallbladder (GB) in some chronic carriers (22%) does not guarantee the elimination of the bacteria, and the rationale for this remains poorly understood. This study is significant as it explores other tissues associated with the chronic carrier state. It highlights not only a cholesterol-rich diet as an important etiological factor for Salmonella colonization but also identifies the cecum as a crucial tissue promoting fecal shedding. Additionally, we determined that biofilm matrix components of Salmonella are key factors contributing to these effects. A greater understanding of these mechanisms will allow the formulation of new therapeutic strategies specifically targeted at preventing typhoid fever dissemination from chronic carriers.
Collapse
Affiliation(s)
- Alonso Cruz-Cruz
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Megan E. Schreeg
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - John S. Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
25
|
Doranga S, Krogfelt KA, Cohen PS, Conway T. Nutrition of Escherichia coli within the intestinal microbiome. EcoSal Plus 2024; 12:eesp00062023. [PMID: 38417452 PMCID: PMC11636361 DOI: 10.1128/ecosalplus.esp-0006-2023] [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: 05/31/2023] [Accepted: 11/03/2023] [Indexed: 03/01/2024]
Abstract
In this chapter, we update our 2004 review of "The Life of Commensal Escherichia coli in the Mammalian Intestine" (https://doi.org/10.1128/ecosalplus.8.3.1.2), with a change of title that reflects the current focus on "Nutrition of E. coli within the Intestinal Microbiome." The earlier part of the previous two decades saw incremental improvements in understanding the carbon and energy sources that E. coli and Salmonella use to support intestinal colonization. Along with these investigations of electron donors came a better understanding of the electron acceptors that support the respiration of these facultative anaerobes in the gastrointestinal tract. Hundreds of recent papers add to what was known about the nutrition of commensal and pathogenic enteric bacteria. The fact that each biotype or pathotype grows on a different subset of the available nutrients suggested a mechanism for succession of commensal colonizers and invasion by enteric pathogens. Competition for nutrients in the intestine has also come to be recognized as one basis for colonization resistance, in which colonized strain(s) prevent colonization by a challenger. In the past decade, detailed investigations of fiber- and mucin-degrading anaerobes added greatly to our understanding of how complex polysaccharides support the hundreds of intestinal microbiome species. It is now clear that facultative anaerobes, which usually cannot degrade complex polysaccharides, live in symbiosis with the anaerobic degraders. This concept led to the "restaurant hypothesis," which emphasizes that facultative bacteria, such as E. coli, colonize the intestine as members of mixed biofilms and obtain the sugars they need for growth locally through cross-feeding from polysaccharide-degrading anaerobes. Each restaurant represents an intestinal niche. Competition for those niches determines whether or not invaders are able to overcome colonization resistance and become established. Topics centered on the nutritional basis of intestinal colonization and gastrointestinal health are explored here in detail.
Collapse
Affiliation(s)
- Sudhir Doranga
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Karen A. Krogfelt
- Department of Science and Environment, Pandemix Center Roskilde University, Roskilde, Denmark
| | - Paul S. Cohen
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, Rhode Island, USA
| | - Tyrrell Conway
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
26
|
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
|
27
|
Kokkinias K, Sabag-Daigle A, Kim Y, Leleiwi I, Shaffer M, Kevorkian R, Daly RA, Wysocki VH, Borton MA, Ahmer BMM, Wrighton KC. Time-resolved multi-omics reveals diverse metabolic strategies of Salmonella during diet-induced inflammation. mSphere 2024; 9:e0053424. [PMID: 39254340 PMCID: PMC11520297 DOI: 10.1128/msphere.00534-24] [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: 06/24/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024] Open
Abstract
With a rise in antibiotic resistance and chronic infection, the metabolic response of Salmonella enterica serovar Typhimurium to various dietary conditions over time remains an understudied avenue for novel, targeted therapeutics. Elucidating how enteric pathogens respond to dietary variation not only helps us decipher the metabolic strategies leveraged for expansion but also assists in proposing targets for therapeutic interventions. In this study, we use a multi-omics approach to identify the metabolic response of Salmonella enterica serovar Typhimurium in mice on both a fibrous diet and high-fat diet over time. When comparing Salmonella gene expression between diets, we found a preferential use of respiratory electron acceptors consistent with increased inflammation in high-fat diet mice. Looking at the high-fat diet over the course of infection, we noticed heterogeneity in samples based on Salmonella ribosomal activity, which is separated into three infection phases: early, peak, and late. We identified key respiratory, carbon, and pathogenesis gene expressions descriptive of each phase. Surprisingly, we identified genes associated with host cell entry expressed throughout infection, suggesting subpopulations of Salmonella or stress-induced dysregulation. Collectively, these results highlight not only the sensitivity of Salmonella to its environment but also identify phase-specific genes that may be used as therapeutic targets to reduce infection.IMPORTANCEIdentifying novel therapeutic strategies for Salmonella infection that occur in relevant diets and over time is needed with the rise of antibiotic resistance and global shifts toward Western diets that are high in fat and low in fiber. Mice on a high-fat diet are more inflamed compared to those on a fibrous diet, creating an environment that results in more favorable energy generation for Salmonella. We observed differential gene expression across infection phases in mice over time on a high-fat diet. Together, these findings reveal the metabolic tuning of Salmonella to dietary and temporal perturbations. Research like this, which explores the dimensions of pathogen metabolic plasticity, can pave the way for rationally designed strategies to control disease.
Collapse
Affiliation(s)
- Katherine Kokkinias
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Yongseok Kim
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Ikaia Leleiwi
- Department of Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Michael Shaffer
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Richard Kevorkian
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Rebecca A. Daly
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Mikayla A. Borton
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Brian M. M. Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Kelly C. Wrighton
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
- Department of Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
28
|
Nguyen BD, Sintsova A, Schubert C, Sichert A, Scheidegger C, Näf J, Huttman J, Lentsch V, Keys T, Rutschmann C, Christen P, Kiefer P, Keller P, Barthel M, Cuenca M, Christen B, Sauer U, Slack E, Vorholt JA, Sunagawa S, Hardt WD. Salmonella Typhimurium screen identifies shifts in mixed-acid fermentation during gut colonization. Cell Host Microbe 2024; 32:1758-1773.e4. [PMID: 39293436 DOI: 10.1016/j.chom.2024.08.015] [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: 11/23/2023] [Revised: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024]
Abstract
How enteric pathogens adapt their metabolism to a dynamic gut environment is not yet fully understood. To investigate how Salmonella enterica Typhimurium (S.Tm) colonizes the gut, we conducted an in vivo transposon mutagenesis screen in a gnotobiotic mouse model. Our data implicate mixed-acid fermentation in efficient gut-luminal growth and energy conservation throughout infection. During initial growth, the pathogen utilizes acetate fermentation and fumarate respiration. After the onset of gut inflammation, hexoses appear to become limiting, as indicated by carbohydrate analytics and the increased need for gluconeogenesis. In response, S.Tm adapts by ramping up ethanol fermentation for redox balancing and supplying the TCA cycle with α-ketoglutarate for additional energy. Our findings illustrate how S.Tm flexibly adapts mixed fermentation and its use of the TCA cycle to thrive in the changing gut environment. Similar metabolic wiring in other pathogenic Enterobacteriaceae may suggest a broadly conserved mechanism for gut colonization.
Collapse
Affiliation(s)
- Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Clio Scheidegger
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julien Huttman
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Verena Lentsch
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Tim Keys
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | | | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Philipp Keller
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Miguelangel Cuenca
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Beat Christen
- Institute of Microbiology, University of Stuttgart, Stuttgart, Germany
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Emma Slack
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
29
|
Singh S, Gola C, Singh B, Agrawal V, Chaba R. D-galactonate metabolism in enteric bacteria: a molecular and physiological perspective. Curr Opin Microbiol 2024; 81:102524. [PMID: 39137493 DOI: 10.1016/j.mib.2024.102524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024]
Abstract
D-galactonate, a widely prevalent sugar acid, was first reported as a nutrient source for enteric bacteria in the 1970s. Since then, decades of research enabled a description of the modified Entner-Doudoroff pathway involved in its degradation and reported the structural and biochemical features of its metabolic enzymes, primarily in Escherichia coli K-12. However, only in the last few years, the D-galactonate transporter has been characterized, and the regulation of the dgo operon, encoding the structural genes for the transporter and enzymes of D-galactonate metabolism, has been detailed. Notably, in recent years, multiple evolutionary studies have identified the dgo operon as a dominant target for adaptation of E. coli in the mammalian gut. Despite considerable research on dgo operon, numerous fundamental questions remain to be addressed. The emerging relevance of the dgo operon in host-bacterial interactions further necessitates the study of D-galactonate metabolism in other enterobacterial strains.
Collapse
Affiliation(s)
- Swati Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab 140306, India
| | - Chetna Gola
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab 140306, India
| | - Bhupinder Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab 140306, India
| | - Vishal Agrawal
- Amity School of Biological Sciences, Amity University Punjab, Mohali, SAS Nagar, Punjab 140306, India
| | - Rachna Chaba
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab 140306, India.
| |
Collapse
|
30
|
Cherrak Y, Salazar MA, Näpflin N, Malfertheiner L, Herzog MKM, Schubert C, von Mering C, Hardt WD. Non-canonical start codons confer context-dependent advantages in carbohydrate utilization for commensal E. coli in the murine gut. Nat Microbiol 2024; 9:2696-2709. [PMID: 39160293 PMCID: PMC11445065 DOI: 10.1038/s41564-024-01775-x] [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: 10/02/2023] [Accepted: 07/04/2024] [Indexed: 08/21/2024]
Abstract
Resource competition is a driver of gut microbiota composition. Bacteria can outcompete metabolically similar rivals through the limitation of shared growth-fuelling nutrients. The mechanisms underlying this remain unclear for bacteria with identical sets of metabolic genes. Here we analysed the lactose utilization operon in the murine commensal Escherichia coli 8178. Using in vitro and in vivo approaches, we showed that translation of the lactose utilization repressor gene lacI from its native non-canonical GTG start codon increases the basal expression of the lactose utilization cluster, enhancing adaptation to lactose consumption. Consequently, a strain carrying the wild type lacI GTG start codon outperformed the lacI ATG start codon mutant in the mouse intestine. This advantage was attenuated upon limiting host lactose intake through diet shift or altering the mutant frequency, emphasizing the context-dependent effect of a single nucleotide change on the bacterial fitness of a common member of the gut microbiota. Coupled with a genomic analysis highlighting the selection of non-ATG start codons in sugar utilization regulator genes across the Enterobacteriaceae family, our data exposed an unsuspected function of non-canonical start codons in metabolic competition.
Collapse
Affiliation(s)
- Yassine Cherrak
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | - Miguel Angel Salazar
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Nicolas Näpflin
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Lukas Malfertheiner
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Mathias K-M Herzog
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Bray AS, Zafar MA. Deciphering the gastrointestinal carriage of Klebsiella pneumoniae. Infect Immun 2024; 92:e0048223. [PMID: 38597634 PMCID: PMC11384780 DOI: 10.1128/iai.00482-23] [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] [Indexed: 04/11/2024] Open
Abstract
Bacterial infections pose a significant global health threat, accounting for an estimated 7.7 million deaths. Hospital outbreaks driven by multi-drug-resistant pathogens, notably Klebsiella pneumoniae (K. pneumoniae), are of grave concern. This opportunistic pathogen causes pneumonia, urinary tract infections, and bacteremia, particularly in immunocompromised individuals. The rise of hypervirulent K. pneumoniae adds complexity, as it increasingly infects healthy individuals. Recent epidemiological data suggest that asymptomatic gastrointestinal carriage serves as a reservoir for infections in the same individual and allows for host-to-host transmission via the fecal-oral route. This review focuses on K. pneumoniae's gastrointestinal colonization, delving into epidemiological evidence, current animal models, molecular colonization mechanisms, and the protective role of the resident gut microbiota. Moreover, the review sheds light on in vivo high-throughput approaches that have been crucial for identifying K. pneumoniae factors in gut colonization. This comprehensive exploration aims to enhance our understanding of K. pneumoniae gut pathogenesis, guiding future intervention and prevention strategies.
Collapse
Affiliation(s)
- Andrew S. Bray
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - M. Ammar Zafar
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
32
|
Pamer EG. Gut microbes fend off harmful bacteria by depriving them of nutrients. Nature 2024; 633:774-775. [PMID: 39294279 DOI: 10.1038/d41586-024-02803-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
|
33
|
Furuichi M, Kawaguchi T, Pust MM, Yasuma-Mitobe K, Plichta DR, Hasegawa N, Ohya T, Bhattarai SK, Sasajima S, Aoto Y, Tuganbaev T, Yaginuma M, Ueda M, Okahashi N, Amafuji K, Kiridoshi Y, Sugita K, Stražar M, Avila-Pacheco J, Pierce K, Clish CB, Skelly AN, Hattori M, Nakamoto N, Caballero S, Norman JM, Olle B, Tanoue T, Suda W, Arita M, Bucci V, Atarashi K, Xavier RJ, Honda K. Commensal consortia decolonize Enterobacteriaceae via ecological control. Nature 2024; 633:878-886. [PMID: 39294375 PMCID: PMC11424487 DOI: 10.1038/s41586-024-07960-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 08/19/2024] [Indexed: 09/20/2024]
Abstract
Persistent colonization and outgrowth of potentially pathogenic organisms in the intestine can result from long-term antibiotic use or inflammatory conditions, and may perpetuate dysregulated immunity and tissue damage1,2. Gram-negative Enterobacteriaceae gut pathobionts are particularly recalcitrant to conventional antibiotic treatment3,4, although an emerging body of evidence suggests that manipulation of the commensal microbiota may be a practical alternative therapeutic strategy5-7. Here we isolated and down-selected commensal bacterial consortia from stool samples from healthy humans that could strongly and specifically suppress intestinal Enterobacteriaceae. One of the elaborated consortia, comprising 18 commensal strains, effectively controlled ecological niches by regulating gluconate availability, thereby re-establishing colonization resistance and alleviating Klebsiella- and Escherichia-driven intestinal inflammation in mice. Harnessing these activities in the form of live bacterial therapies may represent a promising solution to combat the growing threat of proinflammatory, antimicrobial-resistant Enterobacteriaceae infection.
Collapse
Affiliation(s)
- Munehiro Furuichi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takaaki Kawaguchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Marie-Madlen Pust
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Keiko Yasuma-Mitobe
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Damian R Plichta
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Naomi Hasegawa
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Ohya
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shakti K Bhattarai
- Department of Microbiology and Physiological Systems, Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Satoshi Sasajima
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshimasa Aoto
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Timur Tuganbaev
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan
| | - Mizuki Yaginuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Ueda
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Nobuyuki Okahashi
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Kimiko Amafuji
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kiridoshi
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Kayoko Sugita
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Martin Stražar
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julian Avila-Pacheco
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kerry Pierce
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clary B Clish
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashwin N Skelly
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masahira Hattori
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cooperative Major in Advanced Health Science, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | - Takeshi Tanoue
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cooperative Major in Advanced Health Science, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Makoto Arita
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Koji Atarashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan
| | - Ramnik J Xavier
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan.
| |
Collapse
|
34
|
Chen Q, Yu Y, Xu Y, Quan H, Liu D, Li C, Liu M, Gong X. Salmonella Typhimurium alters galactitol metabolism under ciprofloxacin treatment to balance resistance and virulence. J Bacteriol 2024; 206:e0017824. [PMID: 39082861 PMCID: PMC11340313 DOI: 10.1128/jb.00178-24] [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: 05/03/2024] [Accepted: 07/02/2024] [Indexed: 08/23/2024] Open
Abstract
Ciprofloxacin-resistant Salmonella Typhimurium (S. Typhimurium) causes a significant health burden worldwide. A wealth of studies has been published on the contributions of different mechanisms to ciprofloxacin resistance in Salmonella spp. But we still lack a deep understanding of the physiological responses and genetic changes that underlie ciprofloxacin exposure. This study aims to know how phenotypic and genotypic characteristics are impacted by ciprofloxacin exposure, from ciprofloxacin-susceptible to ciprofloxacin-resistant strains in vitro. Here, we investigated the multistep evolution of resistance in replicate populations of S. Typhimurium during 24 days of continuously increasing ciprofloxacin exposure and assessed how ciprofloxacin impacts physiology and genetics. Numerous studies have demonstrated that RamA is a global transcriptional regulator that prominently perturbs the transcriptional landscape of S. Typhimurium, resulting in a ciprofloxacin-resistant phenotype appearing first; the quinolone resistance-determining region mutation site can only be detected later. Comparing the microbial physiological changes and RNA sequencing (RNA-Seq) results of ancestral and selectable mutant strains, the selectable mutant strains had some fitness costs, such as decreased virulence, an increase of biofilm-forming ability, a change of "collateral" sensitivity to other drugs, and inability to utilize galactitol. Importantly, in the ciprofloxacin induced, RamA directly binds and activates the gatR gene responsible for the utilization of galactitol, but RamA deletion strains could not activate gatR. The elevated levels of RamA, which inhibit the galactitol metabolic pathway through the activation of gatR, can lead to a reduction in the growth rate, adhesion, and colonization resistance of S. Typhimurium. This finding is supported by studies conducted in M9 medium as well as in vivo infection models. IMPORTANCE Treatment of antibiotic resistance can significantly benefit from a deeper understanding of the interactions between drugs and genetics. The physiological responses and genetic mechanisms in antibiotic-exposed bacteria are not well understood. Traditional resistance studies, often retrospective, fail to capture the entire resistance development process and typically exhibit unpredictable dynamics. To explore how clinical isolates of S. Typhimurium respond to ciprofloxacin, we analyzed their adaptive responses. We found that S. Typhimurium RamA-mediated regulation disrupts microbial metabolism under ciprofloxacin exposure, affecting genes in the galactitol metabolic pathways. This disruption facilitates adaptive responses to drug therapy and enhances the efficiency of intracellular survival. A more comprehensive and integrated understanding of these physiological and genetic changes is crucial for improving treatment outcomes.
Collapse
Affiliation(s)
- Qiwei Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Animal Medicine and Biosafety, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yongfeng Yu
- State Key Laboratory for Animal Disease Control and Prevention, College of Animal Medicine and Biosafety, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yongchang Xu
- Department of Immunology and Pathogen Biology, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Heng Quan
- State Key Laboratory for Animal Disease Control and Prevention, College of Animal Medicine and Biosafety, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Donghui Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Animal Medicine and Biosafety, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Caiyu Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Animal Medicine and Biosafety, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Mengyao Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Animal Medicine and Biosafety, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xiaowei Gong
- State Key Laboratory for Animal Disease Control and Prevention, College of Animal Medicine and Biosafety, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
35
|
Sima CM, Buzilă ER, Trofin F, Păduraru D, Luncă C, Duhaniuc A, Dorneanu OS, Nastase EV. Emerging Strategies against Non-Typhoidal Salmonella: From Pathogenesis to Treatment. Curr Issues Mol Biol 2024; 46:7447-7472. [PMID: 39057083 PMCID: PMC11275306 DOI: 10.3390/cimb46070442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Even with the intensive efforts by public health programs to control and prevent it, non-typhoidal Salmonella (NTS) infection remains an important public health challenge. It is responsible for approximately 150 million illnesses and 60,000 deaths worldwide annually. NTS infection poses significant risks with high rates of morbidity and mortality, leading to potential short- and long-term complications. There is growing concern among health authorities about the increasing incidence of antimicrobial resistance, with multidrug resistance totaling 22.6% in Europe, highlighting an urgent need for new therapeutic approaches. Our review aims to provide a comprehensive overview of NTS infection. We outline the molecular mechanisms involved in the pathogenesis of NTS infection, as well as the events leading to invasive NTS infection and the subsequent complications associated with it. Given the widespread implications of antimicrobial resistance, our review also presents the global landscape of resistance, including multidrug resistance, and delve into the underlying mechanisms driving this resistance. The rising rates of antibiotic resistance frequently lead to treatment failures, emphasizing the importance of investigating alternative therapeutic options. Therefore, in this review we also explore potential alternative therapies that could offer promising approaches to treating NTS infections.
Collapse
Affiliation(s)
- Cristina Mihaela Sima
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
| | - Elena Roxana Buzilă
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Iasi Regional Center for Public Health, National Institute of Public Health, 700465 Iasi, Romania
| | - Felicia Trofin
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
| | - Diana Păduraru
- “Dr. C.I. Parhon” Clinical Hospital, 700503 Iasi, Romania;
| | - Cătălina Luncă
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania
| | - Alexandru Duhaniuc
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Iasi Regional Center for Public Health, National Institute of Public Health, 700465 Iasi, Romania
| | - Olivia Simona Dorneanu
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
| | - Eduard Vasile Nastase
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
- Department of Internal Medicine II—Infectious Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
36
|
Bejarano E, Domenech-Bendaña A, Avila-Portillo N, Rowan S, Edirisinghe S, Taylor A. Glycative stress as a cause of macular degeneration. Prog Retin Eye Res 2024; 101:101260. [PMID: 38521386 PMCID: PMC11699537 DOI: 10.1016/j.preteyeres.2024.101260] [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: 01/05/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
People are living longer and rates of age-related diseases such as age-related macular degeneration (AMD) are accelerating, placing enormous burdens on patients and health care systems. The quality of carbohydrate foods consumed by an individual impacts health. The glycemic index (GI) is a kinetic measure of the rate at which glucose arrives in the blood stream after consuming various carbohydrates. Consuming diets that favor slowly digested carbohydrates releases sugar into the bloodstream gradually after consuming a meal (low glycemic index). This is associated with reduced risk for major age-related diseases including AMD, cardiovascular disease, and diabetes. In comparison, consuming the same amounts of different carbohydrates in higher GI diets, releases glucose into the blood rapidly, causing glycative stress as well as accumulation of advanced glycation end products (AGEs). Such AGEs are cytotoxic by virtue of their forming abnormal proteins and protein aggregates, as well as inhibiting proteolytic and other protective pathways that might otherwise selectively recognize and remove toxic species. Using in vitro and animal models of glycative stress, we observed that consuming higher GI diets perturbs metabolism and the microbiome, resulting in a shift to more lipid-rich metabolomic profiles. Interactions between aging, diet, eye phenotypes and physiology were observed. A large body of laboratory animal and human clinical epidemiologic data indicates that consuming lower GI diets, or lower glycemia diets, is protective against features of early AMD (AMDf) in mice and AMD prevalence or AMD progression in humans. Drugs may be optimized to diminish the ravages of higher glycemic diets. Human trials are indicated to determine if AMD progression can be retarded using lower GI diets. Here we summarized the current knowledge regarding the pathological role of glycative stress in retinal dysfunction and how dietary strategies might diminish retinal disease.
Collapse
Affiliation(s)
- Eloy Bejarano
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Alicia Domenech-Bendaña
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | | | - Sheldon Rowan
- JM USDA Human Nutrition Research Center on Aging at Tufts University, United States
| | - Sachini Edirisinghe
- Tufts University Friedman School of Nutrition Science and Policy, United States
| | - Allen Taylor
- Tufts University Friedman School of Nutrition Science and Policy, United States.
| |
Collapse
|
37
|
Osbelt L, Almási ÉDH, Wende M, Kienesberger S, Voltz A, Lesker TR, Muthukumarasamy U, Knischewski N, Nordmann E, Bielecka AA, Giralt-Zúñiga M, Kaganovitch E, Kühne C, Baier C, Pietsch M, Müsken M, Greweling-Pils MC, Breinbauer R, Flieger A, Schlüter D, Müller R, Erhardt M, Zechner EL, Strowig T. Klebsiella oxytoca inhibits Salmonella infection through multiple microbiota-context-dependent mechanisms. Nat Microbiol 2024; 9:1792-1811. [PMID: 38862602 PMCID: PMC11222139 DOI: 10.1038/s41564-024-01710-0] [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: 06/06/2023] [Accepted: 04/22/2024] [Indexed: 06/13/2024]
Abstract
The Klebsiella oxytoca species complex is part of the human microbiome, especially during infancy and childhood. K. oxytoca species complex strains can produce enterotoxins, namely, tilimycin and tilivalline, while also contributing to colonization resistance (CR). The relationship between these seemingly contradictory roles is not well understood. Here, by coupling ex vivo assays with CRISPR-mutagenesis and various mouse models, we show that K. oxytoca provides CR against Salmonella Typhimurium. In vitro, the antimicrobial activity against various Salmonella strains depended on tilimycin production and was induced by various simple carbohydrates. In vivo, CR against Salmonella depended on toxin production in germ-free mice, while it was largely toxin-independent in mice with residual microbiota. This was linked to the relative levels of toxin-inducing carbohydrates in vivo. Finally, dulcitol utilization was essential for toxin-independent CR in gnotobiotic mice. Together, this demonstrates that nutrient availability is key to both toxin-dependent and substrate-driven competition between K. oxytoca and Salmonella.
Collapse
Affiliation(s)
- Lisa Osbelt
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
- ESF International Graduate School on Analysis, Imaging and Modelling of Neuronal and Inflammatory Processes, Otto-von-Guericke University, Magdeburg, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Éva D H Almási
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
- ESF International Graduate School on Analysis, Imaging and Modelling of Neuronal and Inflammatory Processes, Otto-von-Guericke University, Magdeburg, Germany
| | - Sabine Kienesberger
- Institute of Molecular Biosciences, University of Graz, BioTechMed-Graz, Graz, Austria
| | - Alexander Voltz
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Till R Lesker
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | | - Nele Knischewski
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Elke Nordmann
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Agata A Bielecka
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - María Giralt-Zúñiga
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eugen Kaganovitch
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Caroline Kühne
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claas Baier
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Michael Pietsch
- Division of Enteropathogenic Bacteria and Legionella (FG11)/National Reference Centre for Salmonella and other Bacterial Enteric Pathogens, Robert Koch Institute, Wernigerode, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Rolf Breinbauer
- BioTechMed-Graz, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Antje Flieger
- Division of Enteropathogenic Bacteria and Legionella (FG11)/National Reference Centre for Salmonella and other Bacterial Enteric Pathogens, Robert Koch Institute, Wernigerode, Germany
| | - Dirk Schlüter
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF),Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Rolf Müller
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- German Center for Infection Research (DZIF),Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Marc Erhardt
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | - Ellen L Zechner
- Institute of Molecular Biosciences, University of Graz, BioTechMed-Graz, Graz, Austria
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
- German Center for Infection Research (DZIF),Partner Site Hannover-Braunschweig, Braunschweig, Germany.
- Center for Individualized Infection Medicine, Hannover, Germany.
| |
Collapse
|
38
|
Fan Y, Wang Y, Xiao H, Sun H. Advancements in understanding the role of intestinal dysbacteriosis mediated mucosal immunity in IgA nephropathy. BMC Nephrol 2024; 25:203. [PMID: 38907188 PMCID: PMC11191200 DOI: 10.1186/s12882-024-03646-3] [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: 04/14/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024] Open
Abstract
IgA nephropathy, presently recognized as the foremost primary glomerular disorder, emerges as a principal contributor to renal failure globally, with its pathogenesis yet to be fully elucidated. Extensive research has highlighted the critical role of gut microbiome in the onset and progression of IgA nephropathy, underscoring its importance in accurately delineating the disease's etiology. For example, gut microbiome dysbacteriosis can lead to the production of nephritogenic IgA1 antibodies, which form immune complexes that deposit in the kidneys, causing inflammation and damage. The gut microbiome, a source of numerous bioactive compounds, interacts with the host and plays a regulatory role in gut-immune axis modulation, earning it the moniker of the "second brain." Recent investigations have particularly emphasized a significant correlation between IgA nephropathy and gut microbiome dysbacteriosis. This article offers a detailed overview of the pathogenic mechanisms of IgA nephropathy, specifically focusing on elucidating how alterations in the gut microbiome are associated with anomalies in the intestinal mucosal system in IgA nephropathy. Additionally, it describes the possible influence of gut microbiome on recurrent IgA nephropathy following kidney transplantation. Furthermore, it compiles potential therapeutic interventions, offering both theoretical and practical foundations for the management of IgA nephropathy. Lastly, the challenges currently faced in the therapeutic approaches to IgA nephropathy are discussed.
Collapse
Affiliation(s)
- Yitao Fan
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Yan Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Han Xiao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Hui Sun
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China.
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
39
|
Yoo W, Shealy NG, Zieba JK, Torres TP, Baltagulov M, Thomas JD, Shelton CD, McGovern AG, Foegeding NJ, Olsan EE, Byndloss MX. Salmonella Typhimurium expansion in the inflamed murine gut is dependent on aspartate derived from ROS-mediated microbiota lysis. Cell Host Microbe 2024; 32:887-899.e6. [PMID: 38806059 PMCID: PMC11189616 DOI: 10.1016/j.chom.2024.05.001] [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: 08/08/2023] [Revised: 03/20/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024]
Abstract
Inflammation boosts the availability of electron acceptors in the intestinal lumen, creating a favorable niche for pathogenic Enterobacteriaceae. However, the mechanisms linking intestinal inflammation-mediated changes in luminal metabolites and pathogen expansion remain unclear. Here, we show that mucosal inflammation induced by Salmonella enterica serovar Typhimurium (S. Tm) infection increases intestinal levels of the amino acid aspartate. S. Tm used aspartate-ammonia lyase (aspA)-dependent fumarate respiration for growth in the murine gut only during inflammation. AspA-dependent growth advantage was abolished in the gut of germ-free mice and restored in gnotobiotic mice colonized with members of the classes Bacteroidia and Clostridia. Reactive oxygen species (ROS) produced during the host response caused lysis of commensal microbes, resulting in the release of microbiota-derived aspartate that was used by S. Tm, in concert with nitrate-dependent anaerobic respiration, to outcompete commensal Enterobacteriaceae. Our findings demonstrate the role of microbiota-derived amino acids in driving respiration-dependent S. Tm expansion during colitis.
Collapse
Affiliation(s)
- Woongjae Yoo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Nicolas G Shealy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jacob K Zieba
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Teresa P Torres
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madi Baltagulov
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Julia D Thomas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Catherine D Shelton
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Anna G McGovern
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nora J Foegeding
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Erin E Olsan
- Department of Biological Sciences, California State University, Sacramento, CA 95819, USA
| | - Mariana X Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Digestive Disease Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, TN 37235, USA; Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
40
|
Muramatsu MK, Winter SE. Nutrient acquisition strategies by gut microbes. Cell Host Microbe 2024; 32:863-874. [PMID: 38870902 PMCID: PMC11178278 DOI: 10.1016/j.chom.2024.05.011] [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: 03/25/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
The composition and function of the gut microbiota are intimately tied to nutrient acquisition strategies and metabolism, with significant implications for host health. Both dietary and host-intrinsic factors influence community structure and the basic modes of bacterial energy metabolism. The intestinal tract is rich in carbon and nitrogen sources; however, limited access to oxygen restricts energy-generating reactions to fermentation. By contrast, increased availability of electron acceptors during episodes of intestinal inflammation results in phylum-level changes in gut microbiota composition, suggesting that bacterial energy metabolism is a key driver of gut microbiota function. In this review article, we will illustrate diverse examples of microbial nutrient acquisition strategies in the context of habitat filters and anatomical location and the central role of energy metabolism in shaping metabolic strategies to support bacterial growth in the mammalian gut.
Collapse
Affiliation(s)
- Matthew K Muramatsu
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis, Davis, CA 95616, USA
| | - Sebastian E Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis, Davis, CA 95616, USA.
| |
Collapse
|
41
|
Rojas VK, Winter MG, Jimenez AG, Tanner NW, Crockett SL, Spiga L, Hendrixson DR, Winter SE. Infection-associated gene regulation of L-tartrate metabolism in Salmonella enterica serovar Typhimurium. mBio 2024; 15:e0035024. [PMID: 38682906 PMCID: PMC11237755 DOI: 10.1128/mbio.00350-24] [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: 02/02/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
Enteric pathogens such as Salmonella enterica serovar Typhimurium experience spatial and temporal changes to the metabolic landscape throughout infection. Host reactive oxygen and nitrogen species non-enzymatically convert monosaccharides to alpha hydroxy acids, including L-tartrate. Salmonella utilizes L-tartrate early during infection to support fumarate respiration, while L-tartrate utilization ceases at later time points due to the increased availability of exogenous electron acceptors such as tetrathionate, nitrate, and oxygen. It remains unknown how Salmonella regulates its gene expression to metabolically adapt to changing nutritional environments. Here, we investigated how the transcriptional regulation for L-tartrate metabolism in Salmonella is influenced by infection-relevant cues. L-tartrate induces the transcription of ttdBAU, genes involved in L-tartrate utilization. L-tartrate metabolism is negatively regulated by two previously uncharacterized transcriptional regulators TtdV (STM3357) and TtdW (STM3358), and both TtdV and TtdW are required for the sensing of L-tartrate. The electron acceptors nitrate, tetrathionate, and oxygen repress ttdBAU transcription via the two-component system ArcAB. Furthermore, the regulation of L-tartrate metabolism is required for optimal fitness in a mouse model of Salmonella-induced colitis. TtdV, TtdW, and ArcAB allow for the integration of two cues, i.e., substrate availability and availability of exogenous electron acceptors, to control L-tartrate metabolism. Our findings provide novel insights into how Salmonella prioritizes the utilization of different electron acceptors for respiration as it experiences transitional nutrient availability throughout infection. IMPORTANCE Bacterial pathogens must adapt their gene expression profiles to cope with diverse environments encountered during infection. This coordinated process is carried out by the integration of cues that the pathogen senses to fine-tune gene expression in a spatiotemporal manner. Many studies have elucidated the regulatory mechanisms of how Salmonella sense metabolites in the gut to activate or repress its virulence program; however, our understanding of how Salmonella coordinates its gene expression to maximize the utilization of carbon and energy sources found in transitional nutrient niches is not well understood. In this study, we discovered how Salmonella integrates two infection-relevant cues, substrate availability and exogenous electron acceptors, to control L-tartrate metabolism. From our experiments, we propose a model for how L-tartrate metabolism is regulated in response to different metabolic cues in addition to characterizing two previously unknown transcriptional regulators. This study expands our understanding of how microbes combine metabolic cues to enhance fitness during infection.
Collapse
Affiliation(s)
- Vivian K. Rojas
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
| | - Maria G. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
| | - Angel G. Jimenez
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Natasha W. Tanner
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
| | - Stacey L. Crockett
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luisella Spiga
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David R. Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sebastian E. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
- Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, California, USA
| |
Collapse
|
42
|
Woelfel S, Silva MS, Stecher B. Intestinal colonization resistance in the context of environmental, host, and microbial determinants. Cell Host Microbe 2024; 32:820-836. [PMID: 38870899 DOI: 10.1016/j.chom.2024.05.002] [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: 02/15/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 06/15/2024]
Abstract
Microbial communities that colonize the human gastrointestinal (GI) tract defend against pathogens through a mechanism known as colonization resistance (CR). Advances in technologies such as next-generation sequencing, gnotobiotic mouse models, and bacterial cultivation have enhanced our understanding of the underlying mechanisms and the intricate microbial interactions involved in CR. Rather than being attributed to specific microbial clades, CR is now understood to arise from a dynamic interplay between microbes and the host and is shaped by metabolic, immune, and environmental factors. This evolving perspective underscores the significance of contextual factors, encompassing microbiome composition and host conditions, in determining CR. This review highlights recent research that has shifted its focus toward elucidating how these factors interact to either promote or impede enteric infections. It further discusses future research directions to unravel the complex relationship between host, microbiota, and environmental determinants in safeguarding against GI infections to promote human health.
Collapse
Affiliation(s)
- Simon Woelfel
- Max von Pettenkofer-Institute for Hygiene and Clinical Microbiology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Marta Salvado Silva
- Max von Pettenkofer-Institute for Hygiene and Clinical Microbiology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer-Institute for Hygiene and Clinical Microbiology, Ludwig Maximilian University of Munich, 80336 Munich, Germany; German Center for Infection Research (DZIF), partner site LMU Munich, Munich, Germany.
| |
Collapse
|
43
|
Cherrak Y, Salazar MA, Yilmaz K, Kreuzer M, Hardt WD. Commensal E. coli limits Salmonella gut invasion during inflammation by producing toxin-bound siderophores in a tonB-dependent manner. PLoS Biol 2024; 22:e3002616. [PMID: 38865418 PMCID: PMC11168627 DOI: 10.1371/journal.pbio.3002616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/05/2024] [Indexed: 06/14/2024] Open
Abstract
The gastrointestinal tract is densely colonized by a polymicrobial community known as the microbiota which serves as primary line of defence against pathogen invasion. The microbiota can limit gut-luminal pathogen growth at different stages of infection. This can be traced to specific commensal strains exhibiting direct or indirect protective functions. Although these mechanisms hold the potential to develop new approaches to combat enteric pathogens, they remain far from being completely described. In this study, we investigated how a mouse commensal Escherichia coli can outcompete Salmonella enterica serovar Typhimurium (S. Tm). Using a salmonellosis mouse model, we found that the commensal E. coli 8178 strain relies on a trojan horse trap strategy to limit S. Tm expansion in the inflamed gut. Combining mutants and reporter tools, we demonstrated that inflammation triggers the expression of the E. coli 8178 antimicrobial microcin H47 toxin which, when fused to salmochelin siderophores, can specifically alter S. Tm growth. This protective function was compromised upon disruption of the E. coli 8178 tonB-dependent catecholate siderophore uptake system, highlighting a previously unappreciated crosstalk between iron intake and microcin H47 activity. By identifying the genetic determinants mediating S. Tm competition, our work not only provides a better mechanistic understanding of the protective function displayed by members of the gut microbiota but also further expands the general contribution of microcins in bacterial antagonistic relationships. Ultimately, such insights can open new avenues for developing microbiota-based approaches to better control intestinal infections.
Collapse
Affiliation(s)
- Yassine Cherrak
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Miguel Angel Salazar
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Koray Yilmaz
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Markus Kreuzer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Dong Q, Harper S, McSpadden E, Son SS, Allen MM, Lin H, Smith RC, Metcalfe C, Burgo V, Woodson C, Sundararajan A, Rose A, McMillin M, Moran D, Little J, Mullowney M, Sidebottom AM, Shen A, Fortier LC, Pamer EG. Protection against Clostridioides difficile disease by a naturally avirulent C. difficile strain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592814. [PMID: 38766138 PMCID: PMC11100753 DOI: 10.1101/2024.05.06.592814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Clostridioides difficile (C. difficile) strains belonging to the epidemic BI/NAP1/027 (RT027) group have been associated with increased transmissibility and disease severity. In addition to the major toxin A and toxin B virulence factors, RT027 strains also encode the CDT binary toxin. Our lab previously identified a toxigenic RT027 isolate, ST1-75, that is avirulent in mice despite densely colonizing the colon. Here, we show that coinfecting mice with the avirulent ST1-75 and virulent R20291 strains protects mice from colitis due to rapid clearance of the virulent strain and persistence of the avirulent strain. Although avirulence of ST1-75 is due to a mutation in the cdtR gene, which encodes a response regulator that modulates the production of all three C. difficile toxins, the ability of ST1-75 to protect against acute colitis is not directly attributable to the cdtR mutation. Metabolomic analyses indicate that the ST1-75 strain depletes amino acids more rapidly than the R20291 strain and supplementation with amino acids ablates ST1-75's competitive advantage, suggesting that the ST1-75 strain limits the growth of virulent R20291 bacteria by amino acid depletion. Since the germination kinetics and sensitivity to the co-germinant glycine are similar for the ST1-75 and R20291 strains, our results identify the rapidity of in vivo nutrient depletion as a mechanism providing strain-specific, virulence-independent competitive advantages to different BI/NAP1/027 strains. They also suggest that the ST1-75 strain may, as a biotherapeutic agent, enhance resistance to CDI in high-risk patients.
Collapse
Affiliation(s)
- Qiwen Dong
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Stephen Harper
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Emma McSpadden
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Sophie S. Son
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, Illinois, USA
| | - Marie-Maude Allen
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Huaiying Lin
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Rita C. Smith
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Carolyn Metcalfe
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Victoria Burgo
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Che Woodson
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | | | - Amber Rose
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Mary McMillin
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - David Moran
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Jessica Little
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | - Michael Mullowney
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | | | - Aimee Shen
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, USA
| | - Louis-Charles Fortier
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric G. Pamer
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
45
|
Almási ÉDH, Knischewski N, Osbelt L, Muthukumarasamy U, El Mouali Y, Vialetto E, Beisel CL, Strowig T. An adapted method for Cas9-mediated editing reveals the species-specific role of β-glucoside utilization driving competition between Klebsiella species. J Bacteriol 2024; 206:e0031723. [PMID: 38353529 PMCID: PMC10955844 DOI: 10.1128/jb.00317-23] [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: 09/27/2023] [Accepted: 01/26/2024] [Indexed: 03/22/2024] Open
Abstract
Cas9-based gene editing tools have revolutionized genetics, enabling the fast and precise manipulation of diverse bacterial species. However, widely applicable genetic tools for non-model gut bacteria are unavailable. Here, we present a two-plasmid Cas9-based system designed for gene deletion and knock-in complementation in three members of the Klebsiella oxytoca species complex (KoSC), which we applied to study the genetic factors underlying the role of these bacteria in competition against Klebsiella pneumoniae. Firstly, the system allowed efficient and precise full-length gene deletion via enhanced lambda Red expression. Furthermore, we tested the efficiency of two independent, functionally validated complementation strategies. Ultimately, the insertion of universal "bookmark" targets during gene deletion subsequently allows the most optimal genetic complementation in K. oxytoca, Klebsiella michiganensis, and Klebsiella grimontii. This approach offers a significant advantage by enabling the use of a single high-efficiency "bookmark" for complementing other loci or strains, eliminating the need for site-specific design. We revealed that the carbohydrate permease CasA is critical in ex vivo assays for K. pneumoniae inhibition by K. oxytoca but is neither sufficient nor required for K. michiganensis and K. grimontii. Thus, the adaptation of state-of-the-art genetic tools to KoSC allows the identification of species-specific functions in microbial competition. IMPORTANCE Cas9-based gene editing tools have revolutionized bacterial genetics, yet, their application to non-model gut bacteria is frequently hampered by various limitations. We utilized a two-plasmid Cas9-based system designed for gene deletion in Klebsiella pneumoniae and demonstrate after optimization its utility for gene editing in three members of the Klebsiella oxytoca species complex (KoSC) namely K. oxytoca, Klebsiella michiganensis, and Klebsiella grimontii. We then adapted a recently developed protocol for functional complementation based on universal "bookmark" targets applicable to all tested species. In summary, species-specific adaptation of state-of-the-art genetic tools allows efficient gene deletion and complementation in type strains as well as natural isolates of KoSC members to study microbial interactions.
Collapse
Affiliation(s)
- Éva d. H. Almási
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Nele Knischewski
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Lisa Osbelt
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | | | - Youssef El Mouali
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Elena Vialetto
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Chase L. Beisel
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Medical Faculty, University of Würzburg, Würzburg, Germany
| | - Till Strowig
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
- Center for Individualized Infection Medicine, Hannover, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| |
Collapse
|
46
|
Wu L, Wang XW, Tao Z, Wang T, Zuo W, Zeng Y, Liu YY, Dai L. Data-driven prediction of colonization outcomes for complex microbial communities. Nat Commun 2024; 15:2406. [PMID: 38493186 PMCID: PMC10944475 DOI: 10.1038/s41467-024-46766-y] [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: 07/07/2023] [Accepted: 03/08/2024] [Indexed: 03/18/2024] Open
Abstract
Microbial interactions can lead to different colonization outcomes of exogenous species, be they pathogenic or beneficial in nature. Predicting the colonization of exogenous species in complex communities remains a fundamental challenge in microbial ecology, mainly due to our limited knowledge of the diverse mechanisms governing microbial dynamics. Here, we propose a data-driven approach independent of any dynamics model to predict colonization outcomes of exogenous species from the baseline compositions of microbial communities. We systematically validate this approach using synthetic data, finding that machine learning models can predict not only the binary colonization outcome but also the post-invasion steady-state abundance of the invading species. Then we conduct colonization experiments for commensal gut bacteria species Enterococcus faecium and Akkermansia muciniphila in hundreds of human stool-derived in vitro microbial communities, confirming that the data-driven approaches can predict the colonization outcomes in experiments. Furthermore, we find that while most resident species are predicted to have a weak negative impact on the colonization of exogenous species, strongly interacting species could significantly alter the colonization outcomes, e.g., Enterococcus faecalis inhibits the invasion of E. faecium invasion. The presented results suggest that the data-driven approaches are powerful tools to inform the ecology and management of microbial communities.
Collapse
Affiliation(s)
- Lu Wu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xu-Wen Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zining Tao
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shandong Agricultural University, Tai'an, China
| | - Tong Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wenlong Zuo
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yu Zeng
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| | - Lei Dai
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
47
|
Ellermann M. Designing microbial communities to starve out invading pathogens. Cell Host Microbe 2024; 32:145-146. [PMID: 38359793 DOI: 10.1016/j.chom.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 02/17/2024]
Abstract
Identifying key features required for specific community-level functions can be challenging, especially considering the complexity of the gut microbiome. In a recent study in Science, Spragge et al. present a high-throughput experimental framework to rationally design microbial communities that can resist invasion by specific bacterial pathogens.
Collapse
Affiliation(s)
- Melissa Ellermann
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
48
|
Beutler M, Eberl C, Garzetti D, Herp S, Münch P, Ring D, Dolowschiak T, Brugiroux S, Schiller P, Hussain S, Basic M, Bleich A, Stecher B. Contribution of bacterial and host factors to pathogen "blooming" in a gnotobiotic mouse model for Salmonella enterica serovar Typhimurium-induced enterocolitis. Infect Immun 2024; 92:e0031823. [PMID: 38189339 PMCID: PMC10863408 DOI: 10.1128/iai.00318-23] [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: 08/17/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Inflammation has a pronounced impact on the intestinal ecosystem by driving an expansion of facultative anaerobic bacteria at the cost of obligate anaerobic microbiota. This pathogen "blooming" is also a hallmark of enteric Salmonella enterica serovar Typhimurium (S. Tm) infection. Here, we analyzed the contribution of bacterial and host factors to S. Tm "blooming" in a gnotobiotic mouse model for S. Tm-induced enterocolitis. Mice colonized with the Oligo-Mouse-Microbiota (OMM12), a minimal bacterial community, develop fulminant colitis by day 4 after oral infection with wild-type S. Tm but not with an avirulent mutant. Inflammation leads to a pronounced reduction in overall intestinal bacterial loads, distinct microbial community shifts, and pathogen blooming (relative abundance >50%). S. Tm mutants attenuated in inducing gut inflammation generally elicit less pronounced microbiota shifts and reduction in total bacterial loads. In contrast, S. Tm mutants in nitrate respiration, salmochelin production, and ethanolamine utilization induced strong inflammation and S. Tm "blooming." Therefore, individual Salmonella-specific inflammation-fitness factors seem to be of minor importance for competition against this minimal microbiota in the inflamed gut. Finally, we show that antibody-mediated neutrophil depletion normalized gut microbiota loads but not intestinal inflammation or microbiota shifts. This suggests that neutrophils equally reduce pathogen and commensal bacterial loads in the inflamed gut.
Collapse
Affiliation(s)
- Markus Beutler
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Claudia Eberl
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Debora Garzetti
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Simone Herp
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Philipp Münch
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
- Computational Biology of Infection Research, Helmholtz Center for Infection Research, Braunschweig, Germany
- Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Diana Ring
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Tamas Dolowschiak
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zurich, Zürich, Switzerland
| | - Sandrine Brugiroux
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Patrick Schiller
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Saib Hussain
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, 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 site LMU Munich, Munich, Germany
| |
Collapse
|
49
|
Rojas VK, Winter MG, Jimenez AG, Tanner NW, Crockett SL, Spiga L, Hendrixson DR, Winter SE. Gene regulation of infection-associated L-tartrate metabolism in Salmonella enterica serovar Typhimurium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578992. [PMID: 38370731 PMCID: PMC10871181 DOI: 10.1101/2024.02.05.578992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Enteric pathogens such as Salmonella enterica serovar Typhimurium experience spatial and temporal changes to the metabolic landscape throughout infection. Host reactive oxygen and nitrogen species non-enzymatically convert monosaccharides to alpha hydroxy acids, including L-tartrate. Salmonella utilizes L-tartrate early during infection to support fumarate respiration, while L-tartrate utilization ceases at later time points due to the increased availability of exogenous electron acceptors such as tetrathionate, nitrate, and oxygen. It remains unknown how Salmonella regulates its gene expression to metabolically adapt to changing nutritional environments. Here, we investigated how the transcriptional regulation for L-tartrate metabolism in Salmonella is influenced by infection-relevant cues. L-tartrate induces the transcription of ttdBAU, genes involved in L-tartrate utilization. L-tartrate metabolism is negatively regulated by two previously uncharacterized transcriptional regulators TtdV (STM3357) and TtdW (STM3358), and both TtdV and TtdW are required for sensing of L-tartrate. The electron acceptors nitrate, tetrathionate, and oxygen repress ttdBAU transcription via the two-component system ArcAB. Furthermore, regulation of L-tartrate metabolism is required for optimal fitness in a mouse model of Salmonella-induced colitis. TtdV, TtdW, and ArcAB allow for the integration of two cues, substrate availability and availability of exogenous electron acceptors, to control L-tartrate metabolism. Our findings provide novel insights into how Salmonella prioritizes utilization of different electron acceptors for respiration as it experiences transitional nutrient availability throughout infection.
Collapse
Affiliation(s)
- Vivian K. Rojas
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
| | - Maria G. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
| | - Angel G. Jimenez
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Current address: Infectious Diseases, Genentech, South San Francisco, California, USA
| | - Natasha W. Tanner
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
| | - Stacey L. Crockett
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Luisella Spiga
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Current address: Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David R. Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sebastian E. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
- Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
50
|
Felsl A, Brokatzky D, Kröger C, Heermann R, Fuchs TM. Hierarchic regulation of a metabolic pathway: H-NS, CRP, and SsrB control myo-inositol utilization by Salmonella enterica. Microbiol Spectr 2024; 12:e0272423. [PMID: 38095474 PMCID: PMC10783015 DOI: 10.1128/spectrum.02724-23] [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: 07/03/2023] [Accepted: 11/07/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE The capacity to utilize myo-inositol (MI) as sole carbon and energy source is widespread among bacteria, among them the intestinal pathogen S. Typhimurium. This study elucidates the complex and hierarchical regulation that underlies the utilization of MI by S. Typhimurium under substrate limitation. A total of seven regulatory factors have been identified so far, allowing the pathogen an environment-dependent, efficient, and fine-tuned regulation of a metabolic property that provides growth advantages in different environments.
Collapse
Affiliation(s)
- Angela Felsl
- Lehrstuhl für Mikrobielle Ökologie, ZIEL-Institute for Food and Health, School of Life Science, Technische Universität München, Freising, Germany
| | - Dominik Brokatzky
- Lehrstuhl für Mikrobielle Ökologie, ZIEL-Institute for Food and Health, School of Life Science, Technische Universität München, Freising, Germany
| | - Carsten Kröger
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Ralf Heermann
- Johannes Gutenberg University Mainz, Institute of Molecular Physiology (imP), Biocenter II, Microbiology and Biotechnology, Mainz, Germany
| | - Thilo M. Fuchs
- Lehrstuhl für Mikrobielle Ökologie, ZIEL-Institute for Food and Health, School of Life Science, Technische Universität München, Freising, Germany
- Friedrich-Loeffler-Institut, Institute of Molecular Pathogenesis, Jena, Germany
| |
Collapse
|