101
|
Nguyen BD, Cuenca V M, Hartl J, Gül E, Bauer R, Meile S, Rüthi J, Margot C, Heeb L, Besser F, Escriva PP, Fetz C, Furter M, Laganenka L, Keller P, Fuchs L, Christen M, Porwollik S, McClelland M, Vorholt JA, Sauer U, Sunagawa S, Christen B, Hardt WD. Import of Aspartate and Malate by DcuABC Drives H 2/Fumarate Respiration to Promote Initial Salmonella Gut-Lumen Colonization in Mice. Cell Host Microbe 2020; 27:922-936.e6. [PMID: 32416061 PMCID: PMC7292772 DOI: 10.1016/j.chom.2020.04.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/16/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Initial enteropathogen growth in the microbiota-colonized gut is poorly understood. Salmonella Typhimurium is metabolically adaptable and can harvest energy by anaerobic respiration using microbiota-derived hydrogen (H2) as an electron donor and fumarate as an electron acceptor. As fumarate is scarce in the gut, the source of this electron acceptor is unclear. Here, transposon sequencing analysis along the colonization trajectory of S. Typhimurium implicates the C4-dicarboxylate antiporter DcuABC in early murine gut colonization. In competitive colonization assays, DcuABC and enzymes that convert the C4-dicarboxylates aspartate and malate into fumarate (AspA, FumABC), are required for fumarate/H2-dependent initial growth. Thus, S. Typhimurium obtains fumarate by DcuABC-mediated import and conversion of L-malate and L-aspartate. Fumarate reduction yields succinate, which is exported by DcuABC in exchange for L-aspartate and L-malate. This cycle allows S. Typhimurium to harvest energy by H2/fumarate respiration in the microbiota-colonized gut. This strategy may also be relevant for commensal E. coli diminishing the S. Typhimurium infection.
Collapse
Affiliation(s)
- Bidong D Nguyen
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | | | - Johannes Hartl
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Ersin Gül
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Rebekka Bauer
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Susanne Meile
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Joel Rüthi
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Céline Margot
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Laura Heeb
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Franziska Besser
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Pau Pérez Escriva
- Institute of Molecular Systems Biology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Céline Fetz
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Markus Furter
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Leanid Laganenka
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Philipp Keller
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Lea Fuchs
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Matthias Christen
- Institute of Molecular Systems Biology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Steffen Porwollik
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92697-4025, USA
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92697-4025, USA
| | - Julia A Vorholt
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland.
| | - Beat Christen
- Institute of Molecular Systems Biology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland.
| | | |
Collapse
|
102
|
Developing a new class of engineered live bacterial therapeutics to treat human diseases. Nat Commun 2020; 11:1738. [PMID: 32269218 PMCID: PMC7142098 DOI: 10.1038/s41467-020-15508-1] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/13/2020] [Indexed: 12/29/2022] Open
Abstract
A complex interplay of metabolic and immunological mechanisms underlies many diseases that represent a substantial unmet medical need. There is an increasing appreciation of the role microbes play in human health and disease, and evidence is accumulating that a new class of live biotherapeutics comprised of engineered microbes could address specific mechanisms of disease. Using the tools of synthetic biology, nonpathogenic bacteria can be designed to sense and respond to environmental signals in order to consume harmful compounds and deliver therapeutic effectors. In this perspective, we describe considerations for the design and development of engineered live biotherapeutics to achieve regulatory and patient acceptance. The role microbes play in human health and the ability of synthetic biology to engineer microbial properties opens up new ways of treating disease. In this perspective, the authors describe the design and development of these living therapeutics.
Collapse
|
103
|
Study of the Histopathologic Effects of Probiotic Lactobacillus acidophilus in Exposure to E. coli O157: H7 in Zebrafish Intestine. IRANIAN RED CRESCENT MEDICAL JOURNAL 2020. [DOI: 10.5812/ircmj.99400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
104
|
Abstract
Mucus in the gastrointestinal (GI) tract is the primary point-of-interaction between humans and their gut microbiota. This intimates that mucus not only ensures protection against endogenous and exogenous opportunists but also provisions for the human microbiota to reside and flourish. With the emergence of living therapeutics, engineered microbes can deliver and produce increasingly complex medicine, and controlling the mucoadhesive properties of different microbial chassis can dictate dose-response in a patient. Here we present a redesigned, in vitro, plate-based assay to measure the mucus adhesion of various probiotics. Cell-mucus interactions were isolated by immobilizing mucus to the plate surface. Binding parameters were derived for each probiotic strain by measuring cell adhesion over a wide range of cell concentrations, providing dose-dependent adhesion metrics. Surface proteins and cell components known to influence mucoadhesion were then heterologously expressed or altered in Lactococcus lactis MG1363 and Escherichia coli Nissle 1917 to control mucus-binding capacity, avidity, and cooperativity.
Collapse
Affiliation(s)
- Zachary J. S. Mays
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Todd C. Chappell
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Nikhil U. Nair
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
105
|
Gut Microbiota, Antibiotic Therapy and Antimicrobial Resistance: A Narrative Review. Microorganisms 2020; 8:microorganisms8020269. [PMID: 32079318 PMCID: PMC7074698 DOI: 10.3390/microorganisms8020269] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/08/2020] [Accepted: 02/08/2020] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial resistance is a major concern. Epidemiological studies have demonstrated direct relationships between antibiotic consumption and emergence/dissemination of resistant strains. Within the last decade, authors confounded spectrum activity and ecological effects and did not take into account several other factors playing important roles, such as impact on anaerobic flora, biliary elimination and sub-inhibitory concentration. The ecological impact of antibiotics on the gut microbiota by direct or indirect mechanisms reflects the breaking of the resistance barrier to colonization. To limit the impact of antibiotic therapy on gut microbiota, consideration of the spectrum of activity and route of elimination must be integrated into the decision. Various strategies to prevent (antimicrobial stewardship, action on residual antibiotics at colonic level) or cure dysbiosis (prebiotic, probiotic and fecal microbiota transplantation) have been introduced or are currently being developed.
Collapse
|
106
|
Identification of Clostridioides difficile-Inhibiting Gut Commensals Using Culturomics, Phenotyping, and Combinatorial Community Assembly. mSystems 2020; 5:5/1/e00620-19. [PMID: 32019832 PMCID: PMC7002114 DOI: 10.1128/msystems.00620-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A major function of the gut microbiota is to provide colonization resistance, wherein pathogens are inhibited or suppressed below infectious levels. However, the fraction of gut microbiota required for colonization resistance remains unclear. We used culturomics to isolate a gut microbiota culture collection comprising 1,590 isolates belonging to 102 species. This culture collection represents 34.57% of the taxonomic diversity and 70% functional capacity, as estimated by metagenomic sequencing of the fecal samples used for culture. Using whole-genome sequencing, we characterized species representatives from this collection and predicted their phenotypic traits, further characterizing isolates by defining nutrient utilization profiles and short-chain fatty acid production. When screened with a coculture assay, 66 species in our culture collection inhibited Clostridioides difficile Several phenotypes, particularly, growth rate, production of SCFAs, and the utilization of mannitol, sorbitol, or succinate, correlated with C. difficile inhibition. We used a combinatorial community assembly approach to formulate defined bacterial mixes inhibitory to C. difficile We tested 256 combinations and found that both species composition and blend size were important in inhibition. Our results show that the interaction of bacteria with one another in a mix and with other members of gut commensals must be investigated to design defined bacterial mixes for inhibiting C. difficile in vivo IMPORTANCE Antibiotic treatment causes instability of gut microbiota and the loss of colonization resistance, thus allowing pathogens such as Clostridioides difficile to colonize and causing recurrent infection and mortality. Although fecal microbiome transplantation has been shown to be an effective treatment for C. difficile infection (CDI), a more desirable approach would be the use of a defined mix of inhibitory gut bacteria. The C. difficile-inhibiting species and bacterial combinations identified herein improve the understanding of the ecological interactions controlling colonization resistance against C. difficile and could aid in the design of defined bacteriotherapy as a nonantibiotic alternative against CDI.
Collapse
|
107
|
Oliveira RA, Ng KM, Correia MB, Cabral V, Shi H, Sonnenburg JL, Huang KC, Xavier KB. Klebsiella michiganensis transmission enhances resistance to Enterobacteriaceae gut invasion by nutrition competition. Nat Microbiol 2020; 5:630-641. [PMID: 31959968 DOI: 10.1038/s41564-019-0658-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/10/2019] [Indexed: 01/18/2023]
Abstract
Intestinal microbiotas contain beneficial microorganisms that protect against pathogen colonization; treatment with antibiotics disrupts the microbiota and compromises colonization resistance. Here, we determine the impact of exchanging microorganisms between hosts on resilience to the colonization of invaders after antibiotic-induced dysbiosis. We assess the functional consequences of dysbiosis using a mouse model of colonization resistance against Escherichia coli. Antibiotics caused stochastic loss of members of the microbiota, but the microbiotas of co-housed mice remained more similar to each other compared with the microbiotas among singly housed animals. Strikingly, co-housed mice maintained colonization resistance after treatment with antibiotics, whereas most singly housed mice were susceptible to E. coli. The ability to retain or share the commensal Klebsiella michiganensis, a member of the Enterobacteriaceae family, was sufficient for colonization resistance after treatment with antibiotics. K. michiganensis generally outcompeted E. coli in vitro, but in vivo administration of galactitol-a nutrient that supports the growth of only E. coli-to bi-colonized gnotobiotic mice abolished the colonization-resistance capacity of K. michiganensis against E. coli, supporting the idea that nutrient competition is the primary interaction mechanism. K. michiganensis also hampered colonization of the pathogen Salmonella, prolonging host survival. Our results address functional consequences of the stochastic effects of microbiota perturbations, whereby microbial transmission through host interactions can facilitate reacquisition of beneficial commensals, minimizing the negative impact of antibiotics.
Collapse
Affiliation(s)
| | - Katharine M Ng
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Vitor Cabral
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Handuo Shi
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Justin L Sonnenburg
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.,Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | |
Collapse
|
108
|
Ebrahimnezhad H, Barzegar L, Esmaeili D, Student of pharmacy, Department of Pharmaceutics, Baqiyatallah University of Medical Sciences,Tehran,Iran, Department of Microbiology and Applied Microbiology Research Center, Systems Biology and Poisonings Institute, BaqiyatallahUniversity of Medical Sciences, Tehran, Iran, Applied VirologyResearch Center, BaqiyatallahUniversity of Medical Sciences, Tehran, Iran, and Department of Microbiology and Applied Microbiology Research Center, Systems Biology and Poisonings Institute, BaqiyatallahUniversity ofMedical Sciences, Tehran, Iran. Antibacterial Effects of Compound Bifilact on E.coli and Campylobacter jejuni. MEDICAL LABORATORY JOURNAL 2020. [DOI: 10.29252/mlj.14.1.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
109
|
Jimenez AG, Ellermann M, Abbott W, Sperandio V. Diet-derived galacturonic acid regulates virulence and intestinal colonization in enterohaemorrhagic Escherichia coli and Citrobacter rodentium. Nat Microbiol 2019; 5:368-378. [PMID: 31873206 PMCID: PMC6992478 DOI: 10.1038/s41564-019-0641-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022]
Abstract
Enteric pathogens sense the complex chemistry within the gastrointestinal (GI) tract to efficiently compete with the resident microbiota and establish a colonization niche. Here we show that enterohemorrhagic E. coli (EHEC), and its surrogate murine infection model Citrobacter rodentium, sense galacturonic-acid to initiate a multi-layered program towards successful mammalian infection. Galacturonic-acid utilization as a carbon source aids the initial pathogen expansion. The main source of galacturonic-acid is dietary pectin, which is broken into galacturonic-acid by the prominent member of the microbiota, Bacteroides thetaiotamicron (Bt). This regulation occurs through the ExuR transcription factor. However, galacturonic-acid is also sensed as a signal through ExuR to modulate the expression of the genes encoding a molecular syringe known as a type three secretion system (T3SS) leading to infectious colitis and inflammation. Galacturonic-acid moonlights as a nutrient and a signal directing the exquisite microbiota-pathogen relationships within the GI tract. Importantly, this work highlights that differential dietary sugar availability impacts the relationship between the microbiota and enteric pathogens, as well as disease outcomes.
Collapse
Affiliation(s)
- Angel G Jimenez
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melissa Ellermann
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wade Abbott
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, Alberta, Canada.,Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Vanessa Sperandio
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
110
|
Wu D, Zheng J, Mao G, Hu W, Ye X, Linhardt RJ, Chen S. Rethinking the impact of RG-I mainly from fruits and vegetables on dietary health. Crit Rev Food Sci Nutr 2019; 60:2938-2960. [PMID: 31607142 DOI: 10.1080/10408398.2019.1672037] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Rhamnogalacturonan I (RG-I) pectin is composed of backbone of repeating disaccharide units →2)-α-L-Rhap-(1→4)-α-D-GalpA-(1→ and neutral sugar side-chains mainly consisting of arabinose and galactose having variable types of linkages. However, since traditional pectin extraction methods damages the RG-I structure, the characteristics and health effects of RG-I remains unclear. Recently, many studies have focused on RG-I, which is often more active than the homogalacturonan (HG) portion of pectic polysaccharides. In food products, RG-I is common to fruits and vegetables and possesses many health benefits. This timely and comprehensive review describes the many different facets of RG-I, including its dietary sources, history, metabolism and potential functionalities, all of which have been compiled to establish a platform for taking full advantage of the functional value of RG-I pectin.
Collapse
Affiliation(s)
- Dongmei Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Jiaqi Zheng
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Guizhu Mao
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Weiwei Hu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Robert J Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| |
Collapse
|
111
|
Metabolic output defines Escherichia coli as a health-promoting microbe against intestinal Pseudomonas aeruginosa. Sci Rep 2019; 9:14463. [PMID: 31595010 PMCID: PMC6783455 DOI: 10.1038/s41598-019-51058-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
Gut microbiota acts as a barrier against intestinal pathogens, but species-specific protection of the host from infection remains relatively unexplored. Although lactobacilli and bifidobacteria produce beneficial lactic and short-chain fatty acids in the mammalian gut, the significance of intestinal Escherichia coli producing these acids is debatable. Taking a Koch’s postulates approach in reverse, we define Escherichia coli as health-promoting for naturally colonizing the gut of healthy mice and protecting them against intestinal colonization and concomitant mortality by Pseudomonas aeruginosa. Reintroduction of faecal bacteria and E. coli in antibiotic-treated mice establishes a high titre of E. coli in the host intestine and increases defence against P. aeruginosa colonization and mortality. Strikingly, high sugar concentration favours E. coli fermentation to lactic and acetic acid and inhibits P. aeruginosa growth and virulence in aerobic cultures and in a model of aerobic metabolism in flies, while dietary vegetable fats - not carbohydrates or proteins - favour E. coli fermentation and protect the host in the anaerobic mouse gut. Thus E. coli metabolic output is an important indicator of resistance to infection. Our work may also suggest that the lack of antimicrobial bacterial metabolites in mammalian lungs and wounds allows P. aeruginosa to be a formidable microbe at these sites.
Collapse
|
112
|
Teixeira PJP, Colaianni NR, Fitzpatrick CR, Dangl JL. Beyond pathogens: microbiota interactions with the plant immune system. Curr Opin Microbiol 2019; 49:7-17. [PMID: 31563068 DOI: 10.1016/j.mib.2019.08.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/15/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022]
Abstract
Plant immune receptors perceive microbial molecules and initiate an array of biochemical responses that are effective against most invaders. The role of the plant immune system in detecting and controlling pathogenic microorganism has been well described. In contrast, much less is known about plant immunity in the context of the wealth of commensals that inhabit plants. Recent research indicates that, just like pathogens, commensals in the plant microbiome can suppress or evade host immune responses. Moreover, the plant immune system has an active role in microbiome assembly and controls microbial homeostasis in response to environmental variation. We propose that the plant immune system shapes the microbiome, and that the microbiome expands plant immunity and acts as an additional layer of defense against pathogenic organisms.
Collapse
Affiliation(s)
- Paulo José Pl Teixeira
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nicholas R Colaianni
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Connor R Fitzpatrick
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jeffery L Dangl
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
113
|
Ruano-Gallego D, Yara DA, Di Ianni L, Frankel G, Schüller S, Fernández LÁ. A nanobody targeting the translocated intimin receptor inhibits the attachment of enterohemorrhagic E. coli to human colonic mucosa. PLoS Pathog 2019; 15:e1008031. [PMID: 31465434 PMCID: PMC6738647 DOI: 10.1371/journal.ppat.1008031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/11/2019] [Accepted: 08/15/2019] [Indexed: 12/31/2022] Open
Abstract
Enterohemorrhagic E. coli (EHEC) is a human intestinal pathogen that causes hemorrhagic colitis and hemolytic uremic syndrome. No vaccines or specific therapies are currently available to prevent or treat these infections. EHEC tightly attaches to the intestinal epithelium by injecting the intimin receptor Tir into the host cell via a type III secretion system (T3SS). In this project, we identified a camelid single domain antibody (nanobody), named TD4, that recognizes a conserved Tir epitope overlapping the binding site of its natural ligand intimin with high affinity and stability. We show that TD4 inhibits attachment of EHEC to cultured human HeLa cells by preventing Tir clustering by intimin, activation of downstream actin polymerization and pedestal formation. Furthermore, we demonstrate that TD4 significantly reduces EHEC adherence to human colonic mucosa in in vitro organ cultures. Altogether, these results suggest that nanobody-based therapies hold potential in the development of much needed treatment and prevention strategies against EHEC infection. Currently, there is no effective treatment or vaccine against enterohemorrhagic E. coli (EHEC), a bacterial pathogen that infects human colon after the ingestion of contaminated food. It thrives in the colon thanks to its ability to attach intimately to the intestinal epithelium. Here, we have identified and characterised a small antibody fragment (nanobody) that recognises Tir, a receptor injected by the bacterium into the host cell to mediate intimate attachment. This nanobody shows higher affinity against Tir than its natural bacterial ligand (intimin) and, most importantly, blocks the intimate attachment of the pathogen to the human colonic tissue. Our results show the potential of this nanobody to prevent and treat EHEC infection.
Collapse
Affiliation(s)
- David Ruano-Gallego
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus UAM-Cantoblanco, Madrid, Spain
- MRC Centre for Molecular Bacteriology and Infection, Life Sciences Department, Imperial College London, London, United Kingdom
| | - Daniel A. Yara
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Lorenza Di Ianni
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus UAM-Cantoblanco, Madrid, Spain
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Life Sciences Department, Imperial College London, London, United Kingdom
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus UAM-Cantoblanco, Madrid, Spain
- * E-mail:
| |
Collapse
|
114
|
Ducarmon QR, Zwittink RD, Hornung BVH, van Schaik W, Young VB, Kuijper EJ. Gut Microbiota and Colonization Resistance against Bacterial Enteric Infection. Microbiol Mol Biol Rev 2019; 83:e00007-19. [PMID: 31167904 PMCID: PMC6710460 DOI: 10.1128/mmbr.00007-19] [Citation(s) in RCA: 310] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome is critical in providing resistance against colonization by exogenous microorganisms. The mechanisms via which the gut microbiota provide colonization resistance (CR) have not been fully elucidated, but they include secretion of antimicrobial products, nutrient competition, support of gut barrier integrity, and bacteriophage deployment. However, bacterial enteric infections are an important cause of disease globally, indicating that microbiota-mediated CR can be disturbed and become ineffective. Changes in microbiota composition, and potential subsequent disruption of CR, can be caused by various drugs, such as antibiotics, proton pump inhibitors, antidiabetics, and antipsychotics, thereby providing opportunities for exogenous pathogens to colonize the gut and ultimately cause infection. In addition, the most prevalent bacterial enteropathogens, including Clostridioides difficile, Salmonella enterica serovar Typhimurium, enterohemorrhagic Escherichia coli, Shigella flexneri, Campylobacter jejuni, Vibrio cholerae, Yersinia enterocolitica, and Listeria monocytogenes, can employ a wide array of mechanisms to overcome colonization resistance. This review aims to summarize current knowledge on how the gut microbiota can mediate colonization resistance against bacterial enteric infection and on how bacterial enteropathogens can overcome this resistance.
Collapse
Affiliation(s)
- Q R Ducarmon
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - R D Zwittink
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - B V H Hornung
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - W van Schaik
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - V B Young
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - E J Kuijper
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
- Clinical Microbiology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
- Netherlands Donor Feces Bank, Leiden, Netherlands
| |
Collapse
|
115
|
Kim WJ, Higashi D, Goytia M, Rendón MA, Pilligua-Lucas M, Bronnimann M, McLean JA, Duncan J, Trees D, Jerse AE, So M. Commensal Neisseria Kill Neisseria gonorrhoeae through a DNA-Dependent Mechanism. Cell Host Microbe 2019; 26:228-239.e8. [PMID: 31378677 DOI: 10.1016/j.chom.2019.07.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/25/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022]
Abstract
The mucosa is colonized with commensal Neisseria. Some of these niches are sites of infection for the STD pathogen Neisseria gonorrhoeae (Ngo). Given the antagonistic behavior of commensal bacteria toward their pathogenic relatives, we hypothesized that commensal Neisseria may negatively affect Ngo colonization. Here, we report that commensal species of Neisseria kill Ngo through a mechanism based on genetic competence and DNA methylation state. Specifically, commensal-triggered killing occurs when the pathogen takes up commensal DNA containing a methylation pattern that it does not recognize. Indeed, any DNA will kill Ngo if it can enter the cell, is differentially methylated, and has homology to the pathogen genome. Consistent with these findings, commensal Neisseria elongata accelerates Ngo clearance from the mouse in a DNA-uptake-dependent manner. Collectively, we propose that commensal Neisseria antagonizes Ngo infection through a DNA-mediated mechanism and that DNA is a potential microbicide against this highly drug-resistant pathogen.
Collapse
Affiliation(s)
- Won Jong Kim
- Department of Immunobiology and the BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Dustin Higashi
- Department of Immunobiology and the BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Maira Goytia
- Department of Biology, Spelman College, Atlanta, GA 30314, USA
| | - Maria A Rendón
- Department of Immunobiology and the BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Michelle Pilligua-Lucas
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - Matthew Bronnimann
- Department of Immunobiology and the BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Jeanine A McLean
- Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Joseph Duncan
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David Trees
- Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Ann E Jerse
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - Magdalene So
- Department of Immunobiology and the BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
116
|
Tang C, Lu Z. Health promoting activities of probiotics. J Food Biochem 2019; 43:e12944. [PMID: 31368544 DOI: 10.1111/jfbc.12944] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
Abstract
In recent years, probiotics have received increasing attention and become one type of popular functional food because of their many biological functions. Among these desirable biological functions, the immune regulation, antioxidative activities, and antimicrobial effects are essential properties to maintain host health. Probiotics can regulate the immune system and improve the antioxidative system by producing microbial components and metabolites. Meanwhile, probiotics also possess antimicrobial abilities owing to their competition for nutrient requirements and mucus adherence, reducing pathogenic toxins, producing antimicrobial metabolites (short-chain fatty acids, bacteriocins, reuterin, linoleic acid, and secondary bile acids) and enhancing intestinal, or systemic immunity. Therefore, probiotics could be used to alleviate heavy metal toxicity and metabolic disorders by improving immunity, the antioxidative system, and intestinal micro-environment. This comprehensive review mainly highlights the potential health promoting activities of probiotics based on their antioxidative, antimicrobial, and immune regulatory effects. PRACTICAL APPLICATIONS: The antioxidative defense and the immune system are essential to maintain human health. However, many factors may result in microbial dysbiosis in the gut, which subsequently leads to pathogenic expansion, oxidative stress, and inflammatory responses. Therefore, it is important to explore beneficial foods to prevent or suppress these abnormal responses. Successful application of probiotics in the functional foods has attracted increasing attention due to their immune regulatory, antioxidative, and antimicrobial properties. The aim of this review is to introduce immune regulatory antioxidative and antimicrobial effects of probiotics, which provides some basic theories for scientific research and development of potential functional foods.
Collapse
Affiliation(s)
- Chao Tang
- Laboratory of Enzyme Engineering, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhaoxin Lu
- Laboratory of Enzyme Engineering, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
117
|
Hagey JV, Bhatnagar S, Heguy JM, Karle BM, Price PL, Meyer D, Maga EA. Fecal Microbial Communities in a Large Representative Cohort of California Dairy Cows. Front Microbiol 2019; 10:1093. [PMID: 31156599 PMCID: PMC6532609 DOI: 10.3389/fmicb.2019.01093] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/30/2019] [Indexed: 11/13/2022] Open
Abstract
Improved sequencing and analytical techniques allow for better resolution of microbial communities; however, the agriculture field lacks an updated analysis surveying the fecal microbial populations of dairy cattle in California. This study is a large-scale survey to determine the composition of the bacterial community present in the feces of lactating dairy cattle on commercial dairy operations. For the study, 10 dairy farms across northern and central California representing a variety of feeding and management systems were enrolled. The farms represented three typical housing types including five freestall, two drylot and three pasture-based management systems. Fresh feces were collected from 15 randomly selected cows on each farm and analyzed using 16S rRNA gene amplicon sequencing. This study found that housing type, individual farm, and dietary components significantly affected the alpha diversity of the fecal microbiota. While only one Operational Taxonomic Unit (OTU) was common among all the sampled individuals, 15 bacterial families and 27 genera were shared among 95% of samples. The ratio of the families Coriobacteriaceae to Bifidobacteriaceae was significantly different between housing types and farms with pasture fed animals having a higher relative abundance of Coriobacteriaceae. A majority of samples were positive for at least one OTU assigned to Enterobacteriaceae and 31% of samples contained OTUs assigned to Campylobacter. However, the relative abundance of both taxa was <0.1%. The microbial composition displays individual farm specific signatures, but housing type plays a role. These data provide insights into the composition of the core fecal microbiota of commercial dairy cows in California and will further generate hypotheses for strategies to manipulate the microbiome of cattle.
Collapse
Affiliation(s)
- Jill V. Hagey
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Srijak Bhatnagar
- Geomicrobiology Group, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Jennifer M. Heguy
- Division of Agriculture and Natural Resources, University of California Cooperative Extension, Hayward, CA, United States
| | - Betsy M. Karle
- Division of Agriculture and Natural Resources, University of California Cooperative Extension, Hayward, CA, United States
| | - Patricia L. Price
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Deanne Meyer
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Elizabeth A. Maga
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| |
Collapse
|
118
|
Drolia R, Bhunia AK. Crossing the Intestinal Barrier via Listeria Adhesion Protein and Internalin A. Trends Microbiol 2019; 27:408-425. [PMID: 30661918 DOI: 10.1016/j.tim.2018.12.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/11/2018] [Accepted: 12/14/2018] [Indexed: 12/24/2022]
Abstract
The intestinal epithelial cell lining provides the first line of defense, yet foodborne pathogens such as Listeria monocytogenes can overcome this barrier; however, the underlying mechanism is not well understood. Though the host M cells in Peyer's patch and the bacterial invasion protein internalin A (InlA) are involved, L. monocytogenes can cross the gut barrier in their absence. The interaction of Listeria adhesion protein (LAP) with the host cell receptor (heat shock protein 60) disrupts the epithelial barrier, promoting bacterial translocation. InlA aids L. monocytogenes transcytosis via interaction with the E-cadherin receptor, which is facilitated by epithelial cell extrusion and goblet cell exocytosis; however, LAP-induced cell junction opening may be an alternative bacterial strategy for InlA access to E-cadherin and its translocation. Here, we summarize the strategies that L. monocytogenes employs to circumvent the intestinal epithelial barrier and compare and contrast these strategies with other enteric bacterial pathogens. Additionally, we provide implications of recent findings for food safety regulations.
Collapse
Affiliation(s)
- Rishi Drolia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
119
|
Liu Y, Zheng Z, Yu L, Wu S, Sun L, Wu S, Xu Q, Cai S, Qin N, Bao W. Examination of the temporal and spatial dynamics of the gut microbiome in newborn piglets reveals distinct microbial communities in six intestinal segments. Sci Rep 2019; 9:3453. [PMID: 30837612 PMCID: PMC6400902 DOI: 10.1038/s41598-019-40235-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 02/12/2019] [Indexed: 02/08/2023] Open
Abstract
Intestinal microbiota plays a crucial role in immune development and disease progression in mammals from birth onwards. The gastrointestinal tract of newborn mammals is rapidly colonized by microbes with tremendous biomass and diversity. Understanding how this complex of segmental communities evolves in different gastrointestinal sites over time has great biological significance and medical implications. However, most previous reports examining intestinal microbiota have focused on fecal samples, a strategy that overlooks the spatial microbial dynamics in different intestinal segments. Using intestinal digesta from six intestinal segments (duodenum, jejunum, ileum, cecum, colon and rectum) of newborn piglets, we herein conducted a large-scale 16S rRNA gene sequencing-based study to characterize the segmental dynamics of porcine gut microbiota at eight postnatal intervals (days 1, 7, 14, 21, 28, 35, 120 and 180). A total of 4,465 OTUs were obtained and showed that the six intestinal segments could be divided into three parts; in the duodenum-jejunum section, the most abundant genera included Lactobacillus and Bacteroides; in the ileum, Fusobacterium and Escherichia; and in the cecum-rectum section, Prevotella. Although the microbial communities of the piglets were similar among the six intestinal segments on postnatal day 1, they evolved and quickly differentiated at later intervals. An examination of time-dependent alterations in the dominant microbes revealed that the microbiome in the large intestine was very different from and much more stable than that in the small intestine. The gut microbiota in newborn piglets exhibited apparent temporal and spatial variations in different intestinal segments. The database of gut microbes in piglets could be a referable resource for future studies on mammalian gut microbiome development in early host growth phases.
Collapse
Affiliation(s)
- Ying Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.,School of Life Science, Huaiyin Normal University, Huaian, 223001, China
| | - Zhijun Zheng
- Realbio Genomics Institute, Shanghai, 200123, China.,Shenzhen Jinrui Biotechnology, Co. Ltd., Shenzhen, 518000, China
| | - Lihuai Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Sen Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Li Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Shenglong Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Qian Xu
- Realbio Genomics Institute, Shanghai, 200123, China.,Shenzhen Jinrui Biotechnology, Co. Ltd., Shenzhen, 518000, China
| | - Shunfeng Cai
- Realbio Genomics Institute, Shanghai, 200123, China.,Shenzhen Jinrui Biotechnology, Co. Ltd., Shenzhen, 518000, China
| | - Nan Qin
- Realbio Genomics Institute, Shanghai, 200123, China. .,Shenzhen Jinrui Biotechnology, Co. Ltd., Shenzhen, 518000, China.
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China. .,Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
120
|
Iacob S, Iacob DG, Luminos LM. Intestinal Microbiota as a Host Defense Mechanism to Infectious Threats. Front Microbiol 2019; 9:3328. [PMID: 30761120 PMCID: PMC6362409 DOI: 10.3389/fmicb.2018.03328] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
The intestinal microbiota is a complex microbial community, with diverse and stable populations hosted by the gastrointestinal tract since birth. This ecosystem holds multiple anti-infectious, anti-inflammatory, and immune modulating roles decisive for intestinal homeostasis. Among these, colonization resistance refers to the dynamic antagonistic interactions between commensals and pathogenic flora. Hence, gut bacteria compete for the same intestinal niches and substrates, while also releasing antimicrobial substances such as bacteriocines and changing the environmental conditions. Short chain fatty acids (SCFAs) generated in anaerobic conditions prompt epigenetic regulatory mechanisms that favor a tolerogenic immune response. In addition, the commensal flora is involved in the synthesis of bactericidal products, namely secondary biliary acids or antimicrobial peptides (AMPs) such as cathellicidin-LL37, an immunomodulatory, antimicrobial, and wound healing peptide. Gut microbiota is protected through symbiotic relations with the hosting organism and by quorum sensing, a specific cell-to-cell communication system. Any alterations of these relationships favor the uncontrollable multiplication of the resident pathobionts or external entero-pathogens, prompting systemic translocations, inflammatory reactions, or exacerbations of bacterial virulence mechanisms (T6SS, T3SS) and ultimately lead to gastrointestinal or systemic infections. The article describes the metabolic and immunological mechanisms through which the intestinal microbiota is both an ally of the organism against enteric pathogens and an enemy that favors the development of infections.
Collapse
Affiliation(s)
- Simona Iacob
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| | - Diana Gabriela Iacob
- National Institute of Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| | - Luminita Monica Luminos
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| |
Collapse
|
121
|
Renouf MJ, Cho YH, McPhee JB. Emergent Behavior of IBD-Associated Escherichia coli During Disease. Inflamm Bowel Dis 2019; 25:33-44. [PMID: 30321333 DOI: 10.1093/ibd/izy312] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel diseases are becoming increasingly common throughout the world, both in developed countries and increasingly in rapidly developing countries. Multiple lines of evidence point to a role for the microbial composition of the gastrointestinal tract in the etiology of IBD, but to date, attempts to define a specific microbial cause for IBD have proved unsuccessful. Microbial 16S rRNA profiling shows that IBD patients have elevated levels of Enterobacteriaceae, in particular Escherichia coli, and reduced levels of Faecalibacterium prausnitzii. The observed E. coli have been assigned to a specific pathovar, adherent-invasive E. coli (AIEC). Adherent-invasive E. coli are a genomically heterogenous group, and whereas many groups have attempted to identify specific genetic markers that differentiate AIEC from non-AIEC strains, very few concrete genetic associations have been uncovered. Here, we highlight the advantages of applying a phenotyping approach to the study of these organisms, rather than solely depending on a sequencing or genomic-based screening strategy because virulence-associated phenotypes exhibit behaviors of emergent systems. In this respect, attempts at genetic reductionism are prone to failure because there are numerous metabolic, regulatory or genetic paths that can underlie these virulence-associated behaviors. Here, we review these IBD-associated phenotypes in E. coli and make recommendations for experimental approaches to advance our understanding of IBD-associated bacteria more generally. With advances in high-throughput screening and nongenetically based metabolomic characterization of IBD-associated bacteria, we anticipate a fuller understanding of how altered microbial communities contribute to the development of IBD.
Collapse
Affiliation(s)
| | - Youn Hee Cho
- Department of Chemistry and Biology, Ryerson University, Toronto ON, Canada
| | - Joseph B McPhee
- Department of Chemistry and Biology, Ryerson University, Toronto ON, Canada
| |
Collapse
|
122
|
Sorbara MT, Pamer EG. Interbacterial mechanisms of colonization resistance and the strategies pathogens use to overcome them. Mucosal Immunol 2019; 12:1-9. [PMID: 29988120 PMCID: PMC6312114 DOI: 10.1038/s41385-018-0053-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/15/2018] [Accepted: 05/27/2018] [Indexed: 02/08/2023]
Abstract
The communities of bacteria that reside in the intestinal tract are in constant competition within this dynamic and densely colonized environment. At homeostasis, the equilibrium that exists between these species and strains is shaped by their metabolism and also by pathways of active antagonism, which drive competition with related and unrelated strains. Importantly, these normal activities contribute to colonization resistance by the healthy microbiota, which includes the ability to prevent the expansion of potential pathogens. Disruption of the microbiota, resulting from, for example, inflammation or antibiotic use, can reduce colonization resistance. Pathogens that engraft following disruption of the microbiota are often adapted to expand into newly created niches and compete in an altered gut environment. In this review, we examine both the interbacterial mechanisms of colonization resistance and the strategies of pathogenic strains to exploit gaps in colonization resistance.
Collapse
Affiliation(s)
- Matthew T. Sorbara
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Eric G. Pamer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
- Center for Microbes, Inflammation and Cancer, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
123
|
Ignacio A, Terra FF, Watanabe IKM, Basso PJ, Câmara NOS. Role of the Microbiome in Intestinal Barrier Function and Immune Defense. MICROBIOME AND METABOLOME IN DIAGNOSIS, THERAPY, AND OTHER STRATEGIC APPLICATIONS 2019:127-138. [DOI: 10.1016/b978-0-12-815249-2.00013-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
124
|
Ndeh D, Gilbert HJ. Biochemistry of complex glycan depolymerisation by the human gut microbiota. FEMS Microbiol Rev 2018; 42:146-164. [PMID: 29325042 DOI: 10.1093/femsre/fuy002] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/06/2018] [Indexed: 12/21/2022] Open
Abstract
The human gut microbiota (HGM) makes an important contribution to health and disease. It is a complex microbial community of trillions of microbes with a majority of its members represented within two phyla, the Bacteroidetes and Firmicutes, although it also contains species of Actinobacteria and Proteobacteria. Reflecting its importance, the HGM is sometimes referred to as an 'organ' as it performs functions analogous to systemic tissues within the human host. The major nutrients available to the HGM are host and dietary complex carbohydrates. To utilise these nutrient sources, the HGM has developed elaborate, variable and sophisticated systems for the sensing, capture and utilisation of these glycans. Understanding nutrient acquisition by the HGM can thus provide mechanistic insights into the dynamics of this ecosystem, and how it impacts human health. Dietary nutrient sources include a wide variety of simple and complex plant and animal-derived glycans most of which are not degraded by enzymes in the digestive tract of the host. Here we review how various adaptive mechanisms that operate across the major phyla of the HGM contribute to glycan utilisation, focusing on the most complex carbohydrates presented to this ecosystem.
Collapse
Affiliation(s)
- Didier Ndeh
- Institute for Cell and Molecular Biosciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Harry J Gilbert
- Institute for Cell and Molecular Biosciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
125
|
Modulation of Enterohaemorrhagic Escherichia coli Survival and Virulence in the Human Gastrointestinal Tract. Microorganisms 2018; 6:microorganisms6040115. [PMID: 30463258 PMCID: PMC6313751 DOI: 10.3390/microorganisms6040115] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 01/05/2023] Open
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) is a major foodborne pathogen responsible for human diseases ranging from diarrhoea to life-threatening complications. Survival of the pathogen and modulation of virulence gene expression along the human gastrointestinal tract (GIT) are key features in bacterial pathogenesis, but remain poorly described, due to a paucity of relevant model systems. This review will provide an overview of the in vitro and in vivo studies investigating the effect of abiotic (e.g., gastric acid, bile, low oxygen concentration or fluid shear) and biotic (e.g., gut microbiota, short chain fatty acids or host hormones) parameters of the human gut on EHEC survival and/or virulence (especially in relation with motility, adhesion and toxin production). Despite their relevance, these studies display important limitations considering the complexity of the human digestive environment. These include the evaluation of only one single digestive parameter at a time, lack of dynamic flux and compartmentalization, and the absence of a complex human gut microbiota. In a last part of the review, we will discuss how dynamic multi-compartmental in vitro models of the human gut represent a novel platform for elucidating spatial and temporal modulation of EHEC survival and virulence along the GIT, and provide new insights into EHEC pathogenesis.
Collapse
|
126
|
Jacobson A, Lam L, Rajendram M, Tamburini F, Honeycutt J, Pham T, Van Treuren W, Pruss K, Stabler SR, Lugo K, Bouley DM, Vilches-Moure JG, Smith M, Sonnenburg JL, Bhatt AS, Huang KC, Monack D. A Gut Commensal-Produced Metabolite Mediates Colonization Resistance to Salmonella Infection. Cell Host Microbe 2018; 24:296-307.e7. [PMID: 30057174 PMCID: PMC6223613 DOI: 10.1016/j.chom.2018.07.002] [Citation(s) in RCA: 334] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/10/2018] [Accepted: 07/05/2018] [Indexed: 02/06/2023]
Abstract
The intestinal microbiota provides colonization resistance against pathogens, limiting pathogen expansion and transmission. These microbiota-mediated mechanisms were previously identified by observing loss of colonization resistance after antibiotic treatment or dietary changes, which severely disrupt microbiota communities. We identify a microbiota-mediated mechanism of colonization resistance against Salmonella enterica serovar Typhimurium (S. Typhimurium) by comparing high-complexity commensal communities with different levels of colonization resistance. Using inbred mouse strains with different infection dynamics and S. Typhimurium intestinal burdens, we demonstrate that Bacteroides species mediate colonization resistance against S. Typhimurium by producing the short-chain fatty acid propionate. Propionate directly inhibits pathogen growth in vitro by disrupting intracellular pH homeostasis, and chemically increasing intestinal propionate levels protects mice from S. Typhimurium. In addition, administering susceptible mice Bacteroides, but not a propionate-production mutant, confers resistance to S. Typhimurium. This work provides mechanistic understanding into the role of individualized microbial communities in host-to-host variability of pathogen transmission.
Collapse
Affiliation(s)
- Amanda Jacobson
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Lilian Lam
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Manohary Rajendram
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Fiona Tamburini
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Jared Honeycutt
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Trung Pham
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Will Van Treuren
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Kali Pruss
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | | | - Kyler Lugo
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Donna M Bouley
- Department of Comparative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jose G Vilches-Moure
- Department of Comparative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mark Smith
- Department of CHEM-H, Stanford University, Stanford, CA 94305, USA
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158 USA
| | - Ami S Bhatt
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Kerwyn Casey Huang
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158 USA
| | - Denise Monack
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
127
|
Sicard JF, Vogeleer P, Le Bihan G, Rodriguez Olivera Y, Beaudry F, Jacques M, Harel J. N-Acetyl-glucosamine influences the biofilm formation of Escherichia coli. Gut Pathog 2018; 10:26. [PMID: 29977346 PMCID: PMC6013987 DOI: 10.1186/s13099-018-0252-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Background The intestinal mucous layer is a physical barrier that limits the contact between bacteria and host epithelial cells. There is growing evidence that microbiota-produced metabolites can also be specifically sensed by gut pathogens as signals to induce or repress virulence genes. Many E. coli, including adherent and invasive (AIEC) strains, can form biofilm. This property can promote their intestinal colonization and resistance to immune mechanisms. We sought to evaluate the impact of mucus-derived sugars on biofilm formation of E. coli. Results We showed that the mucin sugar N-acetyl-glucosamine (NAG) can reduce biofilm formation of AIEC strain LF82. We demonstrated that the inactivation of the regulatory protein NagC, by addition of NAG or by mutation of nagC gene, reduced the biofilm formation of LF82 in static condition. Interestingly, real-time monitoring of biofilm formation of LF82 using microfluidic system showed that the mutation of nagC impairs the early process of biofilm development of LF82. Thus, NAG sensor NagC is involved in the early steps of biofilm formation of AIEC strain LF82 under both static and dynamic conditions. Its implication is partly due to the activation of type 1 fimbriae. NAG can also influence biofilm formation of other intestinal E. coli strains. Conclusions This study highlights how catabolism can be involved in biofilm formation of E. coli. Mucus-derived sugars can influence virulence properties of pathogenic E. coli and this study will help us better understand the mechanisms used to prevent colonization of the intestinal mucosa by pathogens. Electronic supplementary material The online version of this article (10.1186/s13099-018-0252-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jean-Félix Sicard
- 1Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2 Canada
| | - Philippe Vogeleer
- 1Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2 Canada
| | - Guillaume Le Bihan
- 1Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2 Canada
| | - Yaindrys Rodriguez Olivera
- 1Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2 Canada
| | - Francis Beaudry
- 1Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2 Canada
| | - Mario Jacques
- 2Regroupement de Recherche Pour un Lait de Qualité Optimale (Op+Lait), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2 Canada
| | - Josée Harel
- 1Groupe de Recherche sur les Maladies Infectieuses en Production Animale, Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2 Canada
| |
Collapse
|
128
|
Agamennone V, Roelofs D, van Straalen NM, Janssens TKS. Antimicrobial activity in culturable gut microbial communities of springtails. J Appl Microbiol 2018; 125:740-752. [PMID: 29723440 DOI: 10.1111/jam.13899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/01/2018] [Accepted: 04/25/2018] [Indexed: 01/11/2023]
Abstract
AIMS The rise of antibiotic resistance pushes the pharmaceutical industry to continually search for substances with new structures and novel mechanisms of action. Many environmental niches are still to be explored as sources of antimicrobials. In this paper, we assess the antimicrobial potential of gut microbes of springtails, soil invertebrates which live in a microbe-dominated environment and are known to be tolerant to entomopathogenic micro-organisms. METHODS AND RESULTS Bacteria isolated from the guts of five springtail species were tested for inhibitory activity against different microbial pathogens. We identified 46 unique isolates belonging to 17 genera and 15 families. Thirty-five of these isolates (76%) showed inhibitory activity, and 18 inhibited both bacterial and fungal pathogens. One isolate was active against all the pathogens tested. CONCLUSIONS We demonstrated a range of antimicrobial activities in bacteria isolated from the guts of springtails, indicative of complex interactions within the gut community, possibly relating to nutrition or defence against pathogens. SIGNIFICANCE AND IMPACT OF THE STUDY Our results suggest that a large proportion of cultivatable microbes associated with Collembola have a potential for antimicrobial production. We propose that soil invertebrates and their associated microbes are interesting targets for drug discovery.
Collapse
Affiliation(s)
- V Agamennone
- Department of Ecological Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - D Roelofs
- Department of Ecological Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - N M van Straalen
- Department of Ecological Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - T K S Janssens
- Department of Ecological Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Microlife Solutions, Amsterdam, The Netherlands
| |
Collapse
|
129
|
Bury S, Soundararajan M, Bharti R, von Bünau R, Förstner KU, Oelschlaeger TA. The Probiotic Escherichia coli Strain Nissle 1917 Combats Lambdoid Bacteriophages stx and λ. Front Microbiol 2018; 9:929. [PMID: 29896160 PMCID: PMC5987069 DOI: 10.3389/fmicb.2018.00929] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/20/2018] [Indexed: 01/21/2023] Open
Abstract
Shiga toxin (Stx) producing E. coli (STEC) such as Enterohemorrhagic E. coli (EHEC) are the major cause of foodborne illness in humans. In vitro studies showed the probiotic Escherichia coli strain Nissle 1917 (EcN) to efficiently inhibit the production of Stx. Life threatening EHEC strains as for example the serotype O104:H4, responsible for the great outbreak in 2011 in Germany, evolutionary developed from certain E. coli strains which got infected by stx2-encoding lambdoid phages turning the E. coli into lysogenic and subsequently Stx producing strains. Since antibiotics induce stx genes and Stx production, EHEC infected persons are not recommended to be treated with antibiotics. Therefore, EcN might be an alternative medication. However, because even commensal E. coli strains might be converted into Stx-producers after becoming host to a stx encoding prophage, we tested EcN for stx-phage genome integration. Our experiments revealed the resistance of EcN toward not only stx-phages but also against lambda-phages. This resistance was not based on the lack of or by mutated phage receptors. Rather it involved the expression of a phage repressor (pr) gene of a defective prophage in EcN which was able to partially protect E. coli K-12 strain MG1655 against stx and lambda phage infection. Furthermore, we observed EcN to inactivate phages and thereby to protect E. coli K-12 strains against infection by stx- as well as lambda-phages. Inactivation of lambda-phages was due to binding of lambda-phages to LamB of EcN whereas inactivation of stx-phages was caused by a thermostable protein of EcN. These properties together with its ability to inhibit Stx production make EcN a good candidate for the prevention of illness caused by EHEC and probably for the treatment of already infected people.
Collapse
Affiliation(s)
- Susanne Bury
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | | | - Richa Bharti
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | | | - Konrad U Förstner
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Tobias A Oelschlaeger
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
130
|
Kenny DJ, Balskus EP. Engineering chemical interactions in microbial communities. Chem Soc Rev 2018; 47:1705-1729. [PMID: 29210396 DOI: 10.1039/c7cs00664k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Microbes living within host-associated microbial communities (microbiotas) rely on chemical communication to interact with surrounding organisms. These interactions serve many purposes, from supplying the multicellular host with nutrients to antagonizing invading pathogens, and breakdown of chemical signaling has potentially negative consequences for both the host and microbiota. Efforts to engineer microbes to take part in chemical interactions represent a promising strategy for modulating chemical signaling within these complex communities. In this review, we discuss prominent examples of chemical interactions found within host-associated microbial communities, with an emphasis on the plant-root microbiota and the intestinal microbiota of animals. We then highlight how an understanding of such interactions has guided efforts to engineer microbes to participate in chemical signaling in these habitats. We discuss engineering efforts in the context of chemical interactions that enable host colonization, promote host health, and exclude pathogens. Finally, we describe prominent challenges facing this field and propose new directions for future engineering efforts.
Collapse
Affiliation(s)
- Douglas J Kenny
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA.
| | | |
Collapse
|
131
|
Abstract
Enteric pathogens with low infectious doses rely on the ability to orchestrate the expression of virulence and metabolism-associated genes in response to environmental cues for successful infection. Accordingly, the human pathogen enterohemorrhagic Escherichia coli (EHEC) employs a complex multifaceted regulatory network to link the expression of type III secretion system (T3SS) components to nutrient availability. While phosphorylation of histidine and aspartate residues on two-component system response regulators is recognized as an integral part of bacterial signaling, the involvement of phosphotyrosine-mediated control is minimally explored in Gram-negative pathogens. Our recent phosphotyrosine profiling study of E. coli identified 342 phosphorylated proteins, indicating that phosphotyrosine modifications in bacteria are more prevalent than previously anticipated. The present study demonstrates that tyrosine phosphorylation of a metabolite-responsive LacI/GalR family regulator, Cra, negatively affects T3SS expression under glycolytic conditions that are typical for the colonic lumen environment where production of the T3SS is unnecessary. Our data suggest that Cra phosphorylation affects T3SS expression by modulating the expression of ler, which encodes the major activator of EHEC virulence gene expression. Phosphorylation of the Cra Y47 residue diminishes DNA binding to fine-tune the expression of virulence-associated genes, including those of the locus of enterocyte effacement pathogenicity island that encode the T3SS, and thereby negatively affects the formation of attaching and effacing lesions. Our data indicate that tyrosine phosphorylation provides an additional mechanism to control the DNA binding of Cra and other LacI/GalR family regulators, including LacI and PurR. This study describes an initial effort to unravel the role of global phosphotyrosine signaling in the control of EHEC virulence potential. Enterohemorrhagic Escherichia coli (EHEC) causes outbreaks of hemorrhagic colitis and the potentially fatal hemolytic-uremic syndrome. Successful host colonization by EHEC relies on the ability to coordinate the expression of virulence factors in response to environmental cues. A complex network that integrates environmental signals at multiple regulatory levels tightly controls virulence gene expression. We demonstrate that EHEC utilizes a previously uncharacterized phosphotyrosine signaling pathway through Cra to fine-tune the expression of virulence-associated genes to effectively control T3SS production. This study demonstrates that tyrosine phosphorylation negatively affects the DNA-binding capacity of Cra, which affects the expression of genes related to virulence and metabolism. We demonstrate for the first time that phosphotyrosine-mediated control affects global transcription in EHEC. Our data provide insight into a hitherto unexplored regulatory level of the global network controlling EHEC virulence gene expression.
Collapse
|
132
|
Mullineaux-Sanders C, Suez J, Elinav E, Frankel G. Sieving through gut models of colonization resistance. Nat Microbiol 2018; 3:132-140. [PMID: 29358683 DOI: 10.1038/s41564-017-0095-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/08/2017] [Indexed: 12/24/2022]
Abstract
The development of innovative high-throughput genomics and metabolomics technologies has considerably expanded our understanding of the commensal microorganisms residing within the human body, collectively termed the microbiota. In recent years, the microbiota has been reported to have important roles in multiple aspects of human health, pathology and host-pathogen interactions. One function of commensals that has attracted particular interest is their role in protection against pathogens and pathobionts, a concept known as colonization resistance. However, pathogens are also able to sense and exploit the microbiota during infection. Therefore, obtaining a holistic understanding of colonization resistance mechanisms is essential for the development of microbiome-based and microbiome-targeting therapies for humans and animals. Achieving this is dependent on utilizing physiologically relevant animal models. In this Perspective, we discuss the colonization resistance functions of the gut microbiota and sieve through the advantages and limitations of murine models commonly used to study such mechanisms within the context of enteric bacterial infection.
Collapse
Affiliation(s)
- Caroline Mullineaux-Sanders
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Jotham Suez
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK.
| |
Collapse
|
133
|
Waldman AJ, Balskus EP. The Human Microbiota, Infectious Disease, and Global Health: Challenges and Opportunities. ACS Infect Dis 2018; 4:14-26. [PMID: 29207239 DOI: 10.1021/acsinfecdis.7b00232] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Despite significant advances in treating infectious diseases worldwide, morbidity and mortality associated with pathogen infection remains extraordinarily high and represents a critical scientific and global health challenge. Current strategies to combat these infectious agents include a combination of vaccines, small molecule drugs, increased hygiene standards, and disease-specific interventions. While these approaches have helped to drastically reduce the incidence and number of deaths associated with infection, continued investment in current strategies and the development of novel therapeutic approaches will be required to address these global health threats. Recently, human- and vector-associated microbiotas, the assemblages of microorganisms living on and within their hosts, have emerged as a potentially important factor mediating both infection risk and disease progression. These complex microbial communities are involved in intricate and dynamic interactions with both pathogens as well as the innate and adaptive immune systems of their hosts. Here, we discuss recent findings that have illuminated the importance of resident microbiotas in infectious disease, emphasizing opportunities for novel therapeutic intervention and future challenges for the field. Our discussion will focus on four major global health threats: tuberculosis, malaria, HIV, and enteric/diarrheal diseases. We hope this Perspective will highlight the many opportunities for chemists and chemical biologists in this field as well as inspire efforts to elucidate the mechanisms underlying established disease correlations, identify novel microbiota-based risk factors, and develop new therapeutic interventions.
Collapse
Affiliation(s)
- Abraham J. Waldman
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Emily P. Balskus
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
134
|
Cole BK, Scott E, Ilikj M, Bard D, Akins DR, Dyer DW, Chavez-Bueno S. Route of infection alters virulence of neonatal septicemia Escherichia coli clinical isolates. PLoS One 2017; 12:e0189032. [PMID: 29236742 PMCID: PMC5728477 DOI: 10.1371/journal.pone.0189032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 09/05/2017] [Indexed: 12/03/2022] Open
Abstract
Escherichia coli is the leading cause of Gram-negative neonatal septicemia in the United States. Invasion and passage across the neonatal gut after ingestion of maternal E. coli strains produce bacteremia. In this study, we compared the virulence properties of the neonatal E. coli bacteremia clinical isolate SCB34 with the archetypal neonatal E. coli meningitis strain RS218. Whole-genome sequencing data was used to compare the protein coding sequences among these clinical isolates and 33 other representative E. coli strains. Oral inoculation of newborn animals with either strain produced septicemia, whereas intraperitoneal injection caused septicemia only in pups infected with RS218 but not in those injected with SCB34. In addition to being virulent only through the oral route, SCB34 demonstrated significantly greater invasion and transcytosis of polarized intestinal epithelial cells in vitro as compared to RS218. Protein coding sequences comparisons highlighted the presence of known virulence factors that are shared among several of these isolates, and revealed the existence of proteins exclusively encoded in SCB34, many of which remain uncharacterized. Our study demonstrates that oral acquisition is crucial for the virulence properties of the neonatal bacteremia clinical isolate SCB34. This characteristic, along with its enhanced ability to invade and transcytose intestinal epithelium are likely determined by the specific virulence factors that predominate in this strain.
Collapse
Affiliation(s)
- Bryan K. Cole
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Edgar Scott
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Marko Ilikj
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - David Bard
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Darrin R. Akins
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - David W. Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Susana Chavez-Bueno
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
135
|
Chiu L, Bazin T, Truchetet ME, Schaeverbeke T, Delhaes L, Pradeu T. Protective Microbiota: From Localized to Long-Reaching Co-Immunity. Front Immunol 2017; 8:1678. [PMID: 29270167 PMCID: PMC5725472 DOI: 10.3389/fimmu.2017.01678] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/15/2017] [Indexed: 12/17/2022] Open
Abstract
Resident microbiota do not just shape host immunity, they can also contribute to host protection against pathogens and infectious diseases. Previous reviews of the protective roles of the microbiota have focused exclusively on colonization resistance localized within a microenvironment. This review shows that the protection against pathogens also involves the mitigation of pathogenic impact without eliminating the pathogens (i.e., “disease tolerance”) and the containment of microorganisms to prevent pathogenic spread. Protective microorganisms can have an impact beyond their niche, interfering with the entry, establishment, growth, and spread of pathogenic microorganisms. More fundamentally, we propose a series of conceptual clarifications in support of the idea of a “co-immunity,” where an organism is protected by both its own immune system and components of its microbiota.
Collapse
Affiliation(s)
- Lynn Chiu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| | - Thomas Bazin
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Hepato-Gastroenterology, Bordeaux Hospital University Center, Pessac, France
| | | | - Thierry Schaeverbeke
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Rheumatology, Bordeaux Hospital University Center, Bordeaux, France
| | - Laurence Delhaes
- Department of Parasitology and Mycology, Bordeaux Hospital University Center, Bordeaux, France.,University of Bordeaux, INSERM, Cardio-Thoracic Research Centre of Bordeaux, U1045, Bordeaux, France
| | - Thomas Pradeu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| |
Collapse
|
136
|
Vahjen W, Cuisiniere T, Zentek J. Protective effects of indigenous Escherichia coli against a pathogenic E. coli challenge strain in pigs. Benef Microbes 2017; 8:779-783. [PMID: 28969446 DOI: 10.3920/bm2017.0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
To investigate the inhibitory effect of indigenous enterobacteria on pathogenic Escherichia coli, a challenge trial with postweaning pigs was conducted. A pathogenic E. coli strain was administered to all animals and their health was closely monitored thereafter. Faecal samples were taken from three healthy and three diarrhoeic animals. Samples were cultivated on MacConkey agar and isolates were subcultured. A soft agar overlay assay was used to determine the inhibitory activity of the isolates. A total of 1,173 enterobacterial isolates were screened for their ability to inhibit the E. coli challenge strain. Colony forming units of enterobacteria on MacConkey agar were not different between healthy and diarrhoeic animals in the original samples. Furthermore, numbers of isolates per animal were also not significantly different between healthy (482 isolates) and diarrhoeic animals (691 isolates). A total of 43 isolates (3.7%) with inhibitory activity against the pathogenic E. coli challenge strain were detected. All inhibitory isolates were identified as E. coli via MALDI-TOF. The isolates belonged to the phylotypes A, C and E. Many isolates (67.4%) were commensal E. coli without relevant porcine pathogenic factors, but toxin- and fimbrial genes (stx2e, fae, estIb, elt1a, fas, fan) were detected in 14 inhibitory isolates. Healthy animals showed significantly (P=0.003) more inhibitory isolates (36 of 482 isolates; 7.5%) than diseased animals (7 of 691 isolates; 1.0%). There were no significant correlations regarding phylotype or pathogenic factors between healthy and diseased animals. This study has shown that a small proportion of indigenous E. coli is able to inhibit in vitro growth of a pathogenic E. coli strain in pigs. Furthermore, healthy animals possess significantly more inhibitory E. coli strains than diarrhoeic animals. The inhibition of pathogenic E. coli by specific indigenous E. coli strains may be an underlying principle for the containment of pathogenic E. coli in pigs.
Collapse
Affiliation(s)
- W Vahjen
- 1 Institute of Animal Nutrition, Freie Universitaet Berlin, Koenigin-Luise-Str. 49, 14195 Berlin, Germany
| | - T Cuisiniere
- 2 Ecolle de Biologie Industrielle, 13 Boulevard de L'hautil, 95092 Cergy-Pontoise, France
| | - J Zentek
- 1 Institute of Animal Nutrition, Freie Universitaet Berlin, Koenigin-Luise-Str. 49, 14195 Berlin, Germany
| |
Collapse
|
137
|
Al-Saedi F, Vaz DP, Stones DH, Krachler AM. 3-Sulfogalactosyl-dependent adhesion of Escherichia coli HS multivalent adhesion molecule is attenuated by sulfatase activity. J Biol Chem 2017; 292:19792-19803. [PMID: 28982977 PMCID: PMC5712619 DOI: 10.1074/jbc.m117.817908] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Bacterial adhesion to host receptors is an early and essential step in bacterial colonization, and the nature of adhesin–receptor interactions determines bacterial localization and thus the outcome of these interactions. Here, we determined the host receptors for the multivalent adhesion molecule (MAM) from the gut commensal Escherichia coli HS (MAMHS), which contains an array of seven mammalian cell entry domains. The MAMHS adhesin interacted with a range of host receptors, through recognition of a shared 3-O-sulfogalactosyl moiety. This functional group is also found in mucin, a component of the intestinal mucus layer and thus one of the prime adherence targets for commensal E. coli. Mucin gels impeded the motility of E. coli by acting as a physical barrier, and the barrier effect was enhanced by specific interactions between mucin and MAMHS in a sulfation-dependent manner. Desulfation of mucin by pure sulfatase or the sulfatase-producing commensal Bacteroides thetaiotaomicron decreased binding of E. coli to mucin and increased the attachment of bacteria to the epithelial surface via interactions with surface-localized sulfated lipid and protein receptors. Together, our results demonstrate that the E. coli adhesin MAMHS facilitates retention of a gut commensal by attachment to mucin. They further suggest that the amount of sulfatase secreted by mucin-foraging bacteria such as B. thetaiotaomicron, inhabiting the same niche, may affect the capacity of the mucus barrier to retain commensal E. coli.
Collapse
Affiliation(s)
- Fitua Al-Saedi
- From the Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston B15 2TT Birmingham, United Kingdom and
| | - Diana Pereira Vaz
- From the Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston B15 2TT Birmingham, United Kingdom and.,the Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, Texas 77030
| | - Daniel H Stones
- From the Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston B15 2TT Birmingham, United Kingdom and
| | - Anne Marie Krachler
- the Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, Texas 77030
| |
Collapse
|
138
|
Zebrafish ( Danio rerio) as a Vertebrate Model Host To Study Colonization, Pathogenesis, and Transmission of Foodborne Escherichia coli O157. mSphere 2017; 2:mSphere00365-17. [PMID: 28959735 PMCID: PMC5607324 DOI: 10.1128/mspheredirect.00365-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022] Open
Abstract
Foodborne infections with enterohemorrhagic Escherichia coli (EHEC) are a major cause of diarrheal illness in humans and can lead to severe complications such as hemolytic uremic syndrome. Cattle and other ruminants are the main reservoir of EHEC, which enters the food chain through contaminated meat, dairy, or vegetables. Here, we describe the establishment of a vertebrate model for foodborne EHEC infection, using larval zebrafish (Danio rerio) as a host and the protozoan prey Paramecium caudatum as a vehicle. We follow pathogen release from the vehicle, intestinal colonization, microbe-host interactions, and microbial gene induction within a live vertebrate host, in real time, throughout the course of infection. We demonstrate that foodborne EHEC colonizes the gastrointestinal tract faster and establishes a higher burden than waterborne infection. Expression of the locus of enterocyte effacement (LEE), a key EHEC virulence factor, was observed early during infection, mainly at sites that experience fluid shear, and required tight control to enable successful host colonization. EHEC infection led to strain- and LEE-dependent mortality in the zebrafish host. Despite the presence of the endogenous microbiota limiting EHEC colonization levels, EHEC colonization and virulence can be studied either under gnotobiotic conditions or against the backdrop of an endogenous (and variable) host microbiota. Finally, we show that the model can be used for investigation of factors affecting shedding and transmission of bacteria to naive hosts. Overall, this constitutes a useful model, which ideally complements the strengths of existing EHEC vertebrate models. IMPORTANCE Enterohemorrhagic Escherichia coli (EHEC) is a foodborne pathogen which can cause diarrhea, vomiting, and, in some cases, severe complications such as kidney failure in humans. Up to 30% of cattle are colonized with EHEC, which can enter the food chain through contaminated meat, dairy, and vegetables. In order to control infections and stop transmission, it is important to understand what factors allow EHEC to colonize its hosts, cause virulence, and aid transmission. Since this cannot be systematically studied in humans, it is important to develop animal models of infection and transmission. We developed a model which allows us to study foodborne infection in zebrafish, a vertebrate host that is transparent and genetically tractable. Our results show that foodborne infection is more efficient than waterborne infection and that the locus of enterocyte effacement is a key virulence determinant in the zebrafish model. It is induced early during infection, and loss of tight LEE regulation leads to a decreased bacterial burden and decreased host mortality. Overall, the zebrafish model allows us to study foodborne infection, including pathogen release from the food vehicle and gene regulation and its context of host-microbe interactions, as well as environmental shedding and transmission to naive hosts.
Collapse
|
139
|
Sicard JF, Le Bihan G, Vogeleer P, Jacques M, Harel J. Interactions of Intestinal Bacteria with Components of the Intestinal Mucus. Front Cell Infect Microbiol 2017; 7:387. [PMID: 28929087 PMCID: PMC5591952 DOI: 10.3389/fcimb.2017.00387] [Citation(s) in RCA: 307] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022] Open
Abstract
The human gut is colonized by a variety of large amounts of microbes that are collectively called intestinal microbiota. Most of these microbial residents will grow within the mucus layer that overlies the gut epithelium and will act as the first line of defense against both commensal and invading microbes. This mucus is essentially formed by mucins, a family of highly glycosylated protein that are secreted by specialize cells in the gut. In this Review, we examine how commensal members of the microbiota and pathogenic bacteria use mucus to their advantage to promote their growth, develop biofilms and colonize the intestine. We also discuss how mucus-derived components act as nutrient and chemical cues for adaptation and pathogenesis of bacteria and how bacteria can influence the composition of the mucus layer.
Collapse
Affiliation(s)
- Jean-Félix Sicard
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Guillaume Le Bihan
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Philippe Vogeleer
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Mario Jacques
- Regroupement de Recherche Pour un Lait de Qualité Optimale (Op+Lait), Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Josée Harel
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| |
Collapse
|
140
|
Clavel T, Neto JCG, Lagkouvardos I, Ramer-Tait AE. Deciphering interactions between the gut microbiota and the immune system via microbial cultivation and minimal microbiomes. Immunol Rev 2017; 279:8-22. [PMID: 28856739 PMCID: PMC5657458 DOI: 10.1111/imr.12578] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The community of microorganisms in the mammalian gastrointestinal tract, referred to as the gut microbiota, influences host physiology and immunity. The last decade of microbiome research has provided significant advancements for the field and highlighted the importance of gut microbes to states of both health and disease. Novel molecular techniques have unraveled the tremendous diversity of intestinal symbionts that potentially influence the host, many proof-of-concept studies have demonstrated causative roles of gut microbial communities in various pathologies, and microbiome-based approaches are beginning to be implemented in the clinic for diagnostic purposes or for personalized treatments. However, several challenges for the field remain: purely descriptive reports outnumbering mechanistic studies and slow translation of experimental results obtained in animal models into the clinics. Moreover, there is a dearth of knowledge regarding how gut microbes, including novel species that have yet to be identified, impact host immune responses. The sheer complexity of the gut microbial ecosystem makes it difficult, in part, to fully understand the microbiota-host networks that regulate immunity. In the present manuscript, we review key findings on the interactions between gut microbiota members and the immune system. Because culturing microbes allows performing functional studies, we have emphasized the impact of specific taxa or communities thereof. We also highlight underlying molecular mechanisms and discuss opportunities to implement minimal microbiome-based strategies.
Collapse
Affiliation(s)
- Thomas Clavel
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - João Carlos Gomes Neto
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Ilias Lagkouvardos
- ZIEL Institute for Food and Health, Core Facility Microbiome/NGS, Technical University of Munich, Germany
| | - Amanda E. Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
141
|
Kim S, Covington A, Pamer EG. The intestinal microbiota: Antibiotics, colonization resistance, and enteric pathogens. Immunol Rev 2017; 279:90-105. [PMID: 28856737 PMCID: PMC6026851 DOI: 10.1111/imr.12563] [Citation(s) in RCA: 460] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human gastrointestinal tract hosts a diverse network of microorganisms, collectively known as the microbiota that plays an important role in health and disease. For instance, the intestinal microbiota can prevent invading microbes from colonizing the gastrointestinal tract, a phenomenon known as colonization resistance. Perturbations to the microbiota, such as antibiotic administration, can alter microbial composition and result in the loss of colonization resistance. Consequently, the host may be rendered susceptible to colonization by a pathogen. This is a particularly relevant concern in the hospital setting, where antibiotic use and antibiotic-resistant pathogen exposure are more frequent. Many nosocomial infections arise from gastrointestinal colonization. Due to their resistance to antibiotics, treatment is often very challenging. However, recent studies have demonstrated that manipulating the commensal microbiota can prevent and treat various infections in the intestine. In this review, we discuss the members of the microbiota, as well as the mechanisms, that govern colonization resistance against specific pathogens. We also review the effects of antibiotics on the microbiota, as well as the unique epidemiology of immunocompromised patients that renders them a particularly high-risk population to intestinal nosocomial infections.
Collapse
Affiliation(s)
- Sohn Kim
- Immunology Program and Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - April Covington
- Immunology Program and Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric G Pamer
- Immunology Program and Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
142
|
Yin X, Lee B, Zaragoza J, Marco ML. Dietary perturbations alter the ecological significance of ingested Lactobacillus plantarum in the digestive tract. Sci Rep 2017; 7:7267. [PMID: 28779118 PMCID: PMC5544775 DOI: 10.1038/s41598-017-07428-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/23/2017] [Indexed: 12/18/2022] Open
Abstract
Host diet is a major determinant of the composition and function of the intestinal microbiome. Less understood is the importance of diet on ingested strains with probiotic significance. We investigated the population dynamics of exogenous Lactobacillus plantarum and its interactions with intestinal bacteria in mice undergoing switches between high-fat, high-sugar (HFHSD) and low-fat, plant-polysaccharide rich (LFPPD) diets. The survival and persistence of ingested L. plantarum WCFS1 was significantly improved during mouse consumption of HFHSD and was negatively associated with the numbers of indigenous Lactobacillus species. Diet also rapidly changed the composition of the indigenous microbiota, but with some taxa differentially affected between HFHSD periods. L. plantarum was not integrated into indigenous bacterial community networks according to co-occurrence patterns but still conferred distinct effects on bacterial species diversity and microbiota stability largely in a diet-dependent manner. Metagenome predictions supported the premise that L. plantarum dampens the effects of diet on the microbiome. This strain also consistently altered the predicted genetic content in the distal gut by enriching for genes encoding glyosyltransferases and bile salt hydrolases. Our findings demonstrate the interactions between ingested, transient probiotic bacteria and intestinal bacterial communities and how they can differ depending on host diet.
Collapse
Affiliation(s)
- Xiaochen Yin
- Department of Food Science and Technology, University of California, Davis, USA.,Department of Plant Pathology, Univeristy of California, Davis, CA, USA
| | - Bokyung Lee
- Department of Food Science and Technology, University of California, Davis, USA.,Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Jose Zaragoza
- Department of Food Science and Technology, University of California, Davis, USA.,Bayer Crop Science, West Sacramento, CA, USA
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, USA.
| |
Collapse
|
143
|
Galloway-Peña J, Brumlow C, Shelburne S. Impact of the Microbiota on Bacterial Infections during Cancer Treatment. Trends Microbiol 2017; 25:992-1004. [PMID: 28728967 DOI: 10.1016/j.tim.2017.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/06/2017] [Accepted: 06/15/2017] [Indexed: 02/08/2023]
Abstract
Patients being treated for cancer are at high risk for infectious complications, generally due to colonizing organisms that gain access to sterile sites via disrupted epithelial barriers. There is an emerging understanding that the ability of bacterial pathogens, including multidrug-resistant organisms, to colonize and subsequently infect humans is largely dependent on protective bacterial species present in the microbiome. Thus, herein we review recent studies demonstrating strong correlations between the microbiome of the oncology patient and infections occurring during chemotherapy. An increased knowledge of the interplay between potential pathogens, protective commensals, and the host immune system may facilitate the development of novel biomarkers or therapeutics that could help ameliorate the toll that infections take during the treatment of cancer.
Collapse
Affiliation(s)
- Jessica Galloway-Peña
- The Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chelcy Brumlow
- The Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samuel Shelburne
- The Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
144
|
Lewis BB, Pamer EG. Microbiota-Based Therapies for Clostridium difficile and Antibiotic-Resistant Enteric Infections. Annu Rev Microbiol 2017; 71:157-178. [PMID: 28617651 DOI: 10.1146/annurev-micro-090816-093549] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bacterial pathogens are increasingly antibiotic resistant, and development of clinically effective antibiotics is lagging. Curing infections increasingly requires antimicrobials that are broader spectrum, more toxic, and more expensive, and mortality attributable to antibiotic-resistant pathogens is rising. The commensal microbiota, comprising microbes that colonize the mammalian gastrointestinal tract, can provide high levels of resistance to infection, and the contributions of specific bacterial species to resistance are being discovered and characterized. Microbiota-mediated mechanisms of colonization resistance and pathogen clearance include bactericidal activity, nutrient depletion, immune activation, and manipulation of the gut's chemical environment. Current research is focusing on development of microbiota-based therapies to reduce intestinal colonization with antibiotic-resistant pathogens, with the goal of reducing pathogen transmission and systemic dissemination.
Collapse
Affiliation(s)
- Brittany B Lewis
- Infectious Diseases Service, Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065; ,
| | - Eric G Pamer
- Infectious Diseases Service, Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065; ,
| |
Collapse
|
145
|
Mann R, Mediati DG, Duggin IG, Harry EJ, Bottomley AL. Metabolic Adaptations of Uropathogenic E. coli in the Urinary Tract. Front Cell Infect Microbiol 2017; 7:241. [PMID: 28642845 PMCID: PMC5463501 DOI: 10.3389/fcimb.2017.00241] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/24/2017] [Indexed: 12/31/2022] Open
Abstract
Escherichia coli ordinarily resides in the lower gastrointestinal tract in humans, but some strains, known as Uropathogenic E. coli (UPEC), are also adapted to the relatively harsh environment of the urinary tract. Infections of the urine, bladder and kidneys by UPEC may lead to potentially fatal bloodstream infections. To survive this range of conditions, UPEC strains must have broad and flexible metabolic capabilities and efficiently utilize scarce essential nutrients. Whole-organism (or "omics") methods have recently provided significant advances in our understanding of the importance of metabolic adaptation in the success of UPECs. Here we describe the nutritional and metabolic requirements for UPEC infection in these environments, and focus on particular metabolic responses and adaptations of UPEC that appear to be essential for survival in the urinary tract.
Collapse
Affiliation(s)
| | | | | | | | - Amy L. Bottomley
- Faculty of Science, The iThree Institute, University of Technology SydneyUltimo, NSW, Australia
| |
Collapse
|
146
|
Becattini S, Littmann ER, Carter RA, Kim SG, Morjaria SM, Ling L, Gyaltshen Y, Fontana E, Taur Y, Leiner IM, Pamer EG. Commensal microbes provide first line defense against Listeria monocytogenes infection. J Exp Med 2017; 214:1973-1989. [PMID: 28588016 PMCID: PMC5502438 DOI: 10.1084/jem.20170495] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/06/2017] [Accepted: 04/19/2017] [Indexed: 01/22/2023] Open
Abstract
Becattini et al. provide evidence that a diverse gut microbiota antagonizes the foodborne pathogen Listeria monocytogenes in the intestinal lumen, thereby reducing bloodstream invasion. Microbiota perturbation by antibiotic treatment increases susceptibility to listeriosis, with dramatic effects in immunocompromised hosts. Listeria monocytogenes is a foodborne pathogen that causes septicemia, meningitis and chorioamnionitis and is associated with high mortality. Immunocompetent humans and animals, however, can tolerate high doses of L. monocytogenes without developing systemic disease. The intestinal microbiota provides colonization resistance against many orally acquired pathogens, and antibiotic-mediated depletion of the microbiota reduces host resistance to infection. Here we show that a diverse microbiota markedly reduces Listeria monocytogenes colonization of the gut lumen and prevents systemic dissemination. Antibiotic administration to mice before low dose oral inoculation increases L. monocytogenes growth in the intestine. In immunodeficient or chemotherapy-treated mice, the intestinal microbiota provides nonredundant defense against lethal, disseminated infection. We have assembled a consortium of commensal bacteria belonging to the Clostridiales order, which exerts in vitro antilisterial activity and confers in vivo resistance upon transfer into germ free mice. Thus, we demonstrate a defensive role of the gut microbiota against Listeria monocytogenes infection and identify intestinal commensal species that, by enhancing resistance against this pathogen, represent potential probiotics.
Collapse
Affiliation(s)
- Simone Becattini
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Eric R Littmann
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Rebecca A Carter
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sohn G Kim
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sejal M Morjaria
- Infectious Diseases Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Lilan Ling
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Yangtsho Gyaltshen
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Emily Fontana
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ying Taur
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY.,Infectious Diseases Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ingrid M Leiner
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Eric G Pamer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY .,Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY.,Infectious Diseases Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
147
|
Borton MA, Sabag-Daigle A, Wu J, Solden LM, O’Banion BS, Daly RA, Wolfe RA, Gonzalez JF, Wysocki VH, Ahmer BMM, Wrighton KC. Chemical and pathogen-induced inflammation disrupt the murine intestinal microbiome. MICROBIOME 2017; 5:47. [PMID: 28449706 PMCID: PMC5408407 DOI: 10.1186/s40168-017-0264-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/11/2017] [Indexed: 05/06/2023]
Abstract
BACKGROUND Salmonella is one of the most significant food-borne pathogens to affect humans and agriculture. While it is well documented that Salmonella infection triggers host inflammation, the impacts on the gut environment are largely unknown. A CBA/J mouse model was used to evaluate intestinal responses to Salmonella-induced inflammation. In parallel, we evaluated chemically induced inflammation by dextran sodium sulfate (DSS) and a non-inflammation control. We profiled gut microbial diversity by sequencing 16S ribosomal ribonucleic acid (rRNA) genes from fecal and cecal samples. These data were correlated to the inflammation marker lipocalin-2 and short-chain fatty acid concentrations. RESULTS We demonstrated that inflammation, chemically or biologically induced, restructures the chemical and microbial environment of the gut over a 16-day period. We observed that the ten mice within the Salmonella treatment group had a variable Salmonella relative abundance, with three high responding mice dominated by >46% Salmonella at later time points and the remaining seven mice denoted as low responders. These low- and high-responding Salmonella groups, along with the chemical DSS treatment, established an inflammation gradient with chemical and low levels of Salmonella having at least 3 log-fold lower lipocalin-2 concentration than the high-responding Salmonella mice. Total short-chain fatty acid and individual butyrate concentrations each negatively correlated with inflammation levels. Microbial communities were also structured along this inflammation gradient. Low levels of inflammation, regardless of chemical or biological induction, enriched for Akkermansia spp. in the Verrucomicrobiaceae and members of the Bacteroidetes family S24-7. Relative to the control or low inflammation groups, high levels of Salmonella drastically decreased the overall microbial diversity, specifically driven by the reduction of Alistipes and Lachnospiraceae in the Bacteroidetes and Firmicutes phyla, respectively. Conversely, members of the Enterobacteriaceae and Lactobacillus were positively correlated to high levels of Salmonella-induced inflammation. CONCLUSIONS Our results show that enteropathogenic infection and intestinal inflammation are interrelated factors modulating gut homeostasis. These findings may prove informative with regard to prophylactic or therapeutic strategies to prevent disruption of microbial communities, or promote their restoration.
Collapse
Affiliation(s)
- Mikayla A. Borton
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210 USA
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210 USA
| | - Jikang Wu
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Lindsey M. Solden
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Bridget S. O’Banion
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Rebecca A. Daly
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Richard A. Wolfe
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Juan F. Gonzalez
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210 USA
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210 USA
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Brian M. M. Ahmer
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210 USA
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210 USA
| | - Kelly C. Wrighton
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| |
Collapse
|
148
|
Can't you hear me knocking: contact-dependent competition and cooperation in bacteria. Emerg Top Life Sci 2017; 1:75-83. [PMID: 29085916 DOI: 10.1042/etls20160019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Microorganisms are in constant competition for growth niches and environmental resources. In Gram-negative bacteria, contact-dependent growth inhibition (CDI) systems link the fate of one cell with its immediate neighbor through touch-dependent, receptor-mediated toxin delivery. Though discovered for their ability to confer a competitive growth advantage, CDI systems also play significant roles in inter-sibling cooperation, promoting both auto-aggregation and biofilm formation. In this review, we detail the mechanisms of CDI toxin delivery and consider how toxin exchange between isogenic sibling cells could regulate gene expression.
Collapse
|
149
|
Hwang IY, Koh E, Wong A, March JC, Bentley WE, Lee YS, Chang MW. Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models. Nat Commun 2017; 8:15028. [PMID: 28398304 PMCID: PMC5394271 DOI: 10.1038/ncomms15028] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/22/2017] [Indexed: 12/16/2022] Open
Abstract
Bacteria can be genetically engineered to kill specific pathogens or inhibit their virulence. We previously developed a synthetic genetic system that allows a laboratory strain of Escherichia coli to sense and kill Pseudomonas aeruginosa in vitro. Here, we generate a modified version of the system, including a gene encoding an anti-biofilm enzyme, and use the probiotic strain Escherichia coli Nissle 1917 as host. The engineered probiotic shows in vivo prophylactic and therapeutic activity against P. aeruginosa during gut infection in two animal models (Caenorhabditis elegans and mice). These findings support the further development of engineered microorganisms with potential prophylactic and therapeutic activities against gut infections. Bacteria can be engineered to kill specific pathogens. Here, the authors modify and optimize a synthetic genetic system in a probiotic strain of Escherichia coli, and show that the engineered probiotic can sense and kill the pathogen Pseudomonas aeruginosa in two animal models of gut infection.
Collapse
Affiliation(s)
- In Young Hwang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Elvin Koh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Adison Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - John C March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - William E Bentley
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| | - Yung Seng Lee
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore S 119074, Singapore
| | - Matthew Wook Chang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
150
|
Pereira FC, Berry D. Microbial nutrient niches in the gut. Environ Microbiol 2017; 19:1366-1378. [PMID: 28035742 PMCID: PMC5412925 DOI: 10.1111/1462-2920.13659] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 12/14/2022]
Abstract
The composition and function of the mammalian gut microbiota has been the subject of much research in recent years, but the principles underlying the assembly and structure of this complex community remain incompletely understood. Processes that shape the gut microbiota are thought to be mostly niche-driven, with environmental factors such as the composition of available nutrients largely determining whether or not an organism can establish. The concept that the nutrient landscape dictates which organisms can successfully colonize and persist in the gut was first proposed in Rolf Freter's nutrient niche theory. In a situation where nutrients are perfectly mixed and there is balanced microbial growth, Freter postulated that an organism can only survive if it is able to utilize one or a few limiting nutrients more efficiently than its competitors. Recent experimental work indicates, however, that nutrients in the gut vary in space and time. We propose that in such a scenario, Freter's nutrient niche theory must be expanded to account for the co-existence of microorganisms utilizing the same nutrients but in distinct sites or at different times, and that metabolic flexibility and mixed-substrate utilization are common strategies for survival in the face of ever-present nutrient fluctuations.
Collapse
Affiliation(s)
- Fátima C. Pereira
- Department of Microbiology and Ecosystem Science, Division of Microbial EcologyUniversity of ViennaAlthanstrasse 14Vienna1090Austria
| | - David Berry
- Department of Microbiology and Ecosystem Science, Division of Microbial EcologyUniversity of ViennaAlthanstrasse 14Vienna1090Austria
| |
Collapse
|