Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(3): 113925, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38460128

ABSTRACT

Salmonella Typhimurium (S.Tm) utilizes the chemotaxis receptor Tsr to exploit gut inflammation. However, the characteristics of this exploitation and the mechanism(s) employed by the pathogen to circumvent antimicrobial effects of inflammation are poorly defined. Here, using different naturally occurring S.Tm strains (SL1344 and 14028) and competitive infection experiments, we demonstrate that type-three secretion system (T3SS)-2 virulence is indispensable for the beneficial effects of Tsr-directed chemotaxis. The removal of the 14028-specific prophage Gifsy3, encoding virulence effectors, results in the loss of the Tsr-mediated fitness advantage in that strain. Surprisingly, without T3SS-2 effector secretion, chemotaxis toward the gut epithelium using Tsr becomes disadvantageous for either strain. Our findings reveal that luminal neutrophils recruited as a result of NLRC4 inflammasome activation locally counteract S.Tm cells exploiting the byproducts of the host immune response. This work highlights a mechanism by which S.Tm exploitation of gut inflammation for colonization relies on the coordinated effects of chemotaxis and T3SS activities.


Subject(s)
Bacterial Proteins , Chemotaxis , Humans , Virulence , Salmonella typhimurium , Inflammation
2.
Science ; 382(6676): eadj3502, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38096285

ABSTRACT

The human gut microbiome plays an important role in resisting colonization of the host by pathogens, but we lack the ability to predict which communities will be protective. We studied how human gut bacteria influence colonization of two major bacterial pathogens, both in vitro and in gnotobiotic mice. Whereas single species alone had negligible effects, colonization resistance greatly increased with community diversity. Moreover, this community-level resistance rested critically upon certain species being present. We explained these ecological patterns through the collective ability of resistant communities to consume nutrients that overlap with those used by the pathogen. Furthermore, we applied our findings to successfully predict communities that resist a novel target strain. Our work provides a reason why microbiome diversity is beneficial and suggests a route for the rational design of pathogen-resistant communities.


Subject(s)
Gastrointestinal Microbiome , Host-Pathogen Interactions , Klebsiella Infections , Klebsiella pneumoniae , Salmonella Infections , Salmonella typhimurium , Animals , Humans , Mice , Nutrients/metabolism , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/growth & development , Klebsiella pneumoniae/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Symbiosis , Germ-Free Life , Klebsiella Infections/microbiology , Salmonella Infections/microbiology , Escherichia coli/genetics , Escherichia coli/metabolism
3.
PLoS Biol ; 21(8): e3002253, 2023 08.
Article in English | MEDLINE | ID: mdl-37651408

ABSTRACT

Salmonella Typhimurium elicits gut inflammation by the costly expression of HilD-controlled virulence factors. This inflammation alleviates colonization resistance (CR) mediated by the microbiota and thereby promotes pathogen blooms. However, the inflamed gut-milieu can also select for hilD mutants, which cannot elicit or maintain inflammation, therefore causing a loss of the pathogen's virulence. This raises the question of which conditions support the maintenance of virulence in S. Typhimurium. Indeed, it remains unclear why the wild-type hilD allele is dominant among natural isolates. Here, we show that microbiota transfer from uninfected or recovered hosts leads to rapid clearance of hilD mutants that feature attenuated virulence, and thereby contributes to the preservation of the virulent S. Typhimurium genotype. Using mouse models featuring a range of microbiota compositions and antibiotic- or inflammation-inflicted microbiota disruptions, we found that irreversible disruption of the microbiota leads to the accumulation of hilD mutants. In contrast, in models with a transient microbiota disruption, selection for hilD mutants was prevented by the regrowing microbiota community dominated by Lachnospirales and Oscillospirales. Strikingly, even after an irreversible microbiota disruption, microbiota transfer from uninfected donors prevented the rise of hilD mutants. Our results establish that robust S. Typhimurium gut colonization hinges on optimizing its manipulation of the host: A transient and tempered microbiota perturbation is favorable for the pathogen to both flourish in the inflamed gut and also minimize loss of virulence. Moreover, besides conferring CR, the microbiota may have the additional consequence of maintaining costly enteropathogen virulence mechanisms.


Subject(s)
Microbiota , Salmonella typhimurium , Animals , Mice , Virulence/genetics , Salmonella typhimurium/genetics , Virulence Factors/genetics , Inflammation
4.
Cell Host Microbe ; 31(7): 1140-1153.e3, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37348498

ABSTRACT

Antibiotic resistance plasmids can be disseminated between different Enterobacteriaceae in the gut. Here, we investigate how closely related Enterobacteriaceae populations with similar nutrient needs can co-bloom in the same gut and thereby facilitate plasmid transfer. Using different strains of Salmonella Typhimurium (S.Tm SL1344 and ATCC14028) and mouse models of Salmonellosis, we show that the bloom of one strain (i.e., recipient) from very low numbers in a gut pre-occupied by the other strain (i.e., donor) depends on strain-specific utilization of a distinct carbon source, galactitol or arabinose. Galactitol-dependent growth of the recipient S.Tm strain promotes plasmid transfer between non-isogenic strains and between E. coli and S.Tm. In mice stably colonized by a defined microbiota (OligoMM12), galactitol supplementation similarly facilitates co-existence of two S.Tm strains and promotes plasmid transfer. Our work reveals a metabolic strategy used by Enterobacteriaceae to expand in a pre-occupied gut and provides promising therapeutic targets for resistance plasmids spread.


Subject(s)
Escherichia coli , Salmonella Infections , Animals , Mice , Escherichia coli/genetics , Plasmids/genetics , Salmonella typhimurium/genetics , Galactitol , Anti-Bacterial Agents
5.
J Bacteriol ; 204(12): e0034722, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36383016

ABSTRACT

The global rise of drug-resistant bacteria is of great concern. Conjugative transfer of antibiotic resistance plasmids contributes to the emerging resistance crisis. Despite substantial progress in understanding the molecular basis of conjugation in vitro, the in vivo dynamics of intra- and interspecies conjugative plasmid transfer are much less understood. In this study, we focused on the streptomycin resistance-encoding mobilizable plasmid pRSF1010SL1344 (P3) of Salmonella enterica serovar Typhimurium strain SL1344. We show that P3 is mobilized by interacting with the conjugation machinery of the conjugative plasmid pCol1B9SL1344 (P2) of SL1344. Thereby, P3 can be transferred into a broad range of relevant environmental and clinical bacterial isolates in vitro and in vivo. Our data suggest that S. Typhimurium persisters in host tissues can serve as P3 reservoirs and foster transfer of both P2 and P3 once they reseed the gut lumen. This adds to our understanding of resistance plasmid transfer in ecologically relevant niches, including the mammalian gut. IMPORTANCE S. Typhimurium is a globally abundant bacterial species that rapidly occupies new niches and survives unstable environmental conditions. As an enteric pathogen, S. Typhimurium interacts with a broad range of bacterial species residing in the mammalian gut. High abundance of bacteria in the gut lumen facilitates conjugation and spread of plasmid-carried antibiotic resistance genes. By studying the transfer dynamics of the P3 plasmid in vitro and in vivo, we illustrate the impact of S. Typhimurium-mediated antibiotic resistance spread via conjugation to relevant environmental and clinical bacterial isolates. Plasmids are among the most critical vehicles driving antibiotic resistance spread. Further understanding of the dynamics and drivers of antibiotic resistance transfer is needed to develop effective solutions for slowing down the emerging threat of multidrug-resistant bacterial pathogens.


Subject(s)
Salmonella enterica , Salmonella typhimurium , Animals , Salmonella typhimurium/genetics , Serogroup , Conjugation, Genetic , Plasmids/genetics , Salmonella enterica/genetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Mammals/genetics
6.
Nat Commun ; 13(1): 1939, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35410999

ABSTRACT

Intestinal inflammation fuels the transmission of Salmonella Typhimurium (S.Tm). However, a substantial fitness cost is associated with virulence expression. Mutations inactivating transcriptional virulence regulators generate attenuated variants profiting from inflammation without enduring virulence cost. Such variants interfere with the transmission of fully virulent clones. Horizontal transfer of functional regulatory genes (HGT) into attenuated variants could nevertheless favor virulence evolution. To address this hypothesis, we cloned hilD, coding for the master regulator of virulence, into a conjugative plasmid that is highly transferrable during intestinal colonization. The resulting mobile hilD allele allows virulence to emerge from avirulent populations, and to be restored in attenuated mutants competing against virulent clones within-host. However, mutations inactivating the mobile hilD allele quickly arise. The stability of virulence mediated by HGT is strongly limited by its cost, which depends on the hilD expression level, and by the timing of transmission. We conclude that robust evolution of costly virulence expression requires additional selective forces such as narrow population bottlenecks during transmission.


Subject(s)
Gene Expression Regulation, Bacterial , Salmonella typhimurium , Bacterial Proteins/metabolism , Gene Transfer, Horizontal , Humans , Inflammation , Salmonella typhimurium/metabolism , Transcription Factors/metabolism , Virulence/genetics
7.
Elife ; 102021 12 07.
Article in English | MEDLINE | ID: mdl-34872631

ABSTRACT

Many plasmids encode antibiotic resistance genes. Through conjugation, plasmids can be rapidly disseminated. Previous work identified gut luminal donor/recipient blooms and tissue-lodged plasmid-bearing persister cells of the enteric pathogen Salmonella enterica serovar Typhimurium (S.Tm) that survive antibiotic therapy in host tissues, as factors promoting plasmid dissemination among Enterobacteriaceae. However, the buildup of tissue reservoirs and their contribution to plasmid spread await experimental demonstration. Here, we asked if re-seeding-plasmid acquisition-invasion cycles by S.Tm could serve to diversify tissue-lodged plasmid reservoirs, and thereby promote plasmid spread. Starting with intraperitoneal mouse infections, we demonstrate that S.Tm cells re-seeding the gut lumen initiate clonal expansion. Extended spectrum beta-lactamase (ESBL) plasmid-encoded gut luminal antibiotic degradation by donors can foster recipient survival under beta-lactam antibiotic treatment, enhancing transconjugant formation upon re-seeding. S.Tm transconjugants can subsequently re-enter host tissues introducing the new plasmid into the tissue-lodged reservoir. Population dynamics analyses pinpoint recipient migration into the gut lumen as rate-limiting for plasmid transfer dynamics in our model. Priority effects may be a limiting factor for reservoir formation in host tissues. Overall, our proof-of-principle data indicates that luminal antibiotic degradation and shuttling between the gut lumen and tissue-resident reservoirs can promote the accumulation and spread of plasmids within a host over time.


Subject(s)
Drug Resistance, Bacterial/genetics , Plasmids/genetics , Salmonella typhimurium/genetics , Animals , Conjugation, Genetic , Gene Transfer, Horizontal , Mice , Mice, 129 Strain , Plasmids/physiology , Salmonella Infections/drug therapy , Salmonella Infections/microbiology , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism , beta-Lactams/metabolism , beta-Lactams/pharmacology
8.
Cell Microbiol ; 23(12): e13400, 2021 12.
Article in English | MEDLINE | ID: mdl-34800311

ABSTRACT

The pathogenic fungus Cryptococcus neoformans must overcome iron limitation to cause disease in mammalian hosts. Previously, we reported a screen for insertion mutants with poor growth on haem as the sole iron source. In this study, we characterised one such mutant and found that the defective gene encoded a Vam6/Vps39/TRAP1 domain-containing protein required for robust growth on haem, an important iron source in host tissue. We designated this protein Vps3 based on reciprocal best matches with the corresponding protein in Saccharomyces cerevisiae. C. neoformans encodes a second Vam6/Vps39/TRAP1 domain-containing protein designated Vam6/Vlp1, and we found that this protein is also required for robust growth on haem as well as on inorganic iron sources. This protein is predicted to be a component of the homotypic fusion and vacuole protein sorting complex involved in endocytosis. Further characterisation of the vam6Δ and vps3Δ mutants revealed perturbed trafficking of iron acquisition functions (e.g., the high affinity iron permease Cft1) and impaired processing of the transcription factor Rim101, a regulator of haem and iron acquisition. The vps3Δ and vam6Δ mutants also had pleiotropic phenotypes including loss of virulence in a mouse model of cryptococcosis, reduced virulence factor elaboration and increased susceptibility to stress, indicating pleiotropic roles for Vps3 and Vam6 beyond haem use in C. neoformans. TAKE AWAYS: Two Vam6/Vps39/TRAP1-domain proteins, Vps3 and Vam6, support the growth of Cryptococcus neoformans on haem. Loss of Vps3 and Vam6 influences the trafficking and expression of iron uptake proteins. Loss of Vps3 or Vam6 eliminates the ability of C. neoformans to cause disease in a mouse model of cryptococcosis.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Animals , Cryptococcus neoformans/genetics , Fungal Proteins/genetics , Iron , Mice , Vacuoles , Virulence
9.
Methods Mol Biol ; 2357: 253-272, 2021.
Article in English | MEDLINE | ID: mdl-34590264

ABSTRACT

A previously unappreciated link between persisters and the emergence and spread of antibiotic resistance has been recently established. The bulk of this research has been conducted in vitro, but some studies are beginning to elucidate the importance of persister reservoirs in both antibiotic treatment failure and the spread of antibiotic resistance using in vivo models. In order to further this research, careful analyses of the mechanisms of persister reservoir formation as well as the dynamics of persister survival and postantibiotic regrowth are of importance. Here, we present a mouse model to quantitatively study Salmonella persisters in vivo. By using neutral unique sequence barcodes, we describe the quantitative analysis of rare events (aka bottlenecks) associated with persister reservoir formation, survival, and reseeding of the gut lumen. This provides quantitative data for persister-fueled plasmid transfer in vivo. Although this chapter describes analysis of Salmonella persisters in a mouse model, these concepts can be applied to any experimental system provided that tractable experimental systems are present.


Subject(s)
Salmonella Infections , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Mice , Plasmids/genetics , Population Density , Salmonella/genetics
10.
Nat Microbiol ; 6(7): 830-841, 2021 07.
Article in English | MEDLINE | ID: mdl-34045711

ABSTRACT

The ability of gut bacterial pathogens to escape immunity by antigenic variation-particularly via changes to surface-exposed antigens-is a major barrier to immune clearance1. However, not all variants are equally fit in all environments2,3. It should therefore be possible to exploit such immune escape mechanisms to direct an evolutionary trade-off. Here, we demonstrate this phenomenon using Salmonella enterica subspecies enterica serovar Typhimurium (S.Tm). A dominant surface antigen of S.Tm is its O-antigen: a long, repetitive glycan that can be rapidly varied by mutations in biosynthetic pathways or by phase variation4,5. We quantified the selective advantage of O-antigen variants in the presence and absence of O-antigen-specific immunoglobulin A and identified a set of evolutionary trajectories allowing immune escape without an associated fitness cost in naive mice. Through the use of rationally designed oral vaccines, we induced immunoglobulin A responses blocking all of these trajectories. This selected for Salmonella mutants carrying deletions of the O-antigen polymerase gene wzyB. Due to their short O-antigen, these evolved mutants were more susceptible to environmental stressors (detergents or complement) and predation (bacteriophages) and were impaired in gut colonization and virulence in mice. Therefore, a rationally induced cocktail of intestinal antibodies can direct an evolutionary trade-off in S.Tm. This lays the foundations for the exploration of mucosal vaccines capable of setting evolutionary traps as a prophylactic strategy.


Subject(s)
Immunoglobulin A/immunology , Intestines/immunology , Salmonella Infections/prevention & control , Salmonella Vaccines/immunology , Salmonella typhimurium/immunology , Administration, Oral , Animals , Antibodies, Bacterial/immunology , Antigenic Variation , Bacterial Proteins/genetics , Evolution, Molecular , Genetic Fitness , Hexosyltransferases/genetics , Immune Evasion , Immunity, Mucosal , Intestines/microbiology , Mice , Mutation , O Antigens/genetics , O Antigens/immunology , Salmonella Infections/microbiology , Salmonella Vaccines/administration & dosage , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Virulence
11.
Mucosal Immunol ; 14(3): 615-629, 2021 05.
Article in English | MEDLINE | ID: mdl-33731826

ABSTRACT

The gut epithelium is a critical protective barrier. Its NAIP/NLRC4 inflammasome senses infection by Gram-negative bacteria, including Salmonella Typhimurium (S.Tm) and promotes expulsion of infected enterocytes. During the first ~12-24 h, this reduces mucosal S.Tm loads at the price of moderate enteropathy. It remained unknown how this NAIP/NLRC4-dependent tradeoff would develop during subsequent infection stages. In NAIP/NLRC4-deficient mice, S.Tm elicited severe enteropathy within 72 h, characterized by elevated mucosal TNF (>20 pg/mg) production from bone marrow-derived cells, reduced regeneration, excessive enterocyte loss, and a collapse of the epithelial barrier. TNF-depleting antibodies prevented this destructive pathology. In hosts proficient for epithelial NAIP/NLRC4, a heterogeneous enterocyte death response with both apoptotic and pyroptotic features kept S.Tm loads persistently in check, thereby preventing this dire outcome altogether. Our results demonstrate that immediate and selective removal of infected enterocytes, by locally acting epithelium-autonomous NAIP/NLRC4, is required to avoid a TNF-driven inflammatory hyper-reaction that otherwise destroys the epithelial barrier.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Enterocytes/immunology , Inflammation/immunology , Intestinal Mucosa/pathology , Neuronal Apoptosis-Inhibitory Protein/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Cells, Cultured , Cytotoxicity, Immunologic , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Apoptosis-Inhibitory Protein/genetics , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
ISME J ; 15(3): 862-878, 2021 03.
Article in English | MEDLINE | ID: mdl-33149210

ABSTRACT

Horizontal gene transfer, mediated by conjugative plasmids, is a major driver of the global rise of antibiotic resistance. However, the relative contributions of factors that underlie the spread of plasmids and their roles in conjugation in vivo are unclear. To address this, we investigated the spread of clinical Extended Spectrum Beta-Lactamase (ESBL)-producing plasmids in the absence of antibiotics in vitro and in the mouse intestine. We hypothesised that plasmid properties would be the primary determinants of plasmid spread and that bacterial strain identity would also contribute. We found clinical Escherichia coli strains natively associated with ESBL-plasmids conjugated to three distinct E. coli strains and one Salmonella enterica serovar Typhimurium strain. Final transconjugant frequencies varied across plasmid, donor, and recipient combinations, with qualitative consistency when comparing transfer in vitro and in vivo in mice. In both environments, transconjugant frequencies for these natural strains and plasmids covaried with the presence/absence of transfer genes on ESBL-plasmids and were affected by plasmid incompatibility. By moving ESBL-plasmids out of their native hosts, we showed that donor and recipient strains also modulated transconjugant frequencies. This suggests that plasmid spread in the complex gut environment of animals and humans can be predicted based on in vitro testing and genetic data.


Subject(s)
Escherichia coli , Salmonella enterica , Animals , Anti-Bacterial Agents/pharmacology , Conjugation, Genetic , Escherichia coli/genetics , Gene Transfer, Horizontal , Mice , Plasmids/genetics , Salmonella enterica/genetics , beta-Lactamases/genetics
13.
Nat Rev Microbiol ; 18(9): 479-490, 2020 09.
Article in English | MEDLINE | ID: mdl-32461608

ABSTRACT

Antibiotic treatment failure is of growing concern. Genetically encoded resistance is key in driving this process. However, there is increasing evidence that bacterial antibiotic persistence, a non-genetically encoded and reversible loss of antibiotic susceptibility, contributes to treatment failure and emergence of resistant strains as well. In this Review, we discuss the evolutionary forces that may drive the selection for antibiotic persistence. We review how some aspects of antibiotic persistence have been directly selected for whereas others result from indirect selection in disparate ecological contexts. We then discuss the consequences of antibiotic persistence on pathogen evolution. Persisters can facilitate the evolution of antibiotic resistance and virulence. Finally, we propose practical means to prevent persister formation and how this may help to slow down the evolution of virulence and resistance in pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/genetics , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/physiology , Evolution, Molecular
14.
Mucosal Immunol ; 13(3): 530-544, 2020 05.
Article in English | MEDLINE | ID: mdl-31953493

ABSTRACT

Inflammasomes can prevent systemic dissemination of enteropathogenic bacteria. As adapted pathogens including Salmonella Typhimurium (S. Tm) have evolved evasion strategies, it has remained unclear when and where inflammasomes restrict their dissemination. Bacterial population dynamics establish that the NAIP/NLRC4 inflammasome specifically restricts S. Tm migration from the gut to draining lymph nodes. This is solely attributable to NAIP/NLRC4 within intestinal epithelial cells (IECs), while S. Tm evades restriction by phagocyte NAIP/NLRC4. NLRP3 and Caspase-11 also fail to restrict S. Tm mucosa traversal, migration to lymph nodes, and systemic pathogen growth. The ability of IECs (not phagocytes) to mount a NAIP/NLRC4 defense in vivo is explained by particularly high NAIP/NLRC4 expression in IECs and the necessity for epithelium-invading S. Tm to express the NAIP1-6 ligands-flagella and type-III-secretion-system-1. Imaging reveals both ligands to be promptly downregulated following IEC-traversal. These results highlight the importance of intestinal epithelial NAIP/NLRC4 in blocking bacterial dissemination in vivo, and explain why this constitutes a uniquely evasion-proof defense against the adapted enteropathogen S. Tm.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Pathogen-Associated Molecular Pattern Molecules , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Animals , Caspases/metabolism , Disease Models, Animal , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Organ Specificity/immunology , Phagocytes/immunology , Phagocytes/metabolism , Salmonella Infections/metabolism
15.
Nat Microbiol ; 4(12): 2164-2174, 2019 12.
Article in English | MEDLINE | ID: mdl-31591555

ABSTRACT

The microbiota confers colonization resistance, which blocks Salmonella gut colonization1. As diet affects microbiota composition, we studied whether food composition shifts enhance susceptibility to infection. Shifting mice to diets with reduced fibre or elevated fat content for 24 h boosted Salmonella Typhimurium or Escherichia coli gut colonization and plasmid transfer. Here, we studied the effect of dietary fat. Colonization resistance was restored within 48 h of return to maintenance diet. Salmonella gut colonization was also boosted by two oral doses of oleic acid or bile salts. These pathogen blooms required Salmonella's AcrAB/TolC-dependent bile resistance. Our data indicate that fat-elicited bile promoted Salmonella gut colonization. Both E. coli and Salmonella show much higher bile resistance than the microbiota. Correspondingly, competitive E. coli can be protective in the fat-challenged gut. Diet shifts and fat-elicited bile promote S. Typhimurium gut infections in mice lacking E. coli in their microbiota. This mouse model may be useful for studying pathogen-microbiota-host interactions, the protective effect of E. coli, to analyse the spread of resistance plasmids and assess the impact of food components on the infection process.


Subject(s)
Dietary Fats/administration & dosage , Escherichia coli/physiology , Gastrointestinal Microbiome , Microbial Interactions , Salmonella typhimurium/physiology , Animal Feed , Animals , Bile Acids and Salts/administration & dosage , Female , Host-Pathogen Interactions , Male , Mice , Mice, Inbred C57BL , Oleic Acids/administration & dosage
16.
Nature ; 573(7773): 276-280, 2019 09.
Article in English | MEDLINE | ID: mdl-31485077

ABSTRACT

The emergence of antibiotic-resistant bacteria through mutations or the acquisition of genetic material such as resistance plasmids represents a major public health issue1,2. Persisters are subpopulations of bacteria that survive antibiotics by reversibly adapting their physiology3-10, and can promote the emergence of antibiotic-resistant mutants11. We investigated whether persisters can also promote the spread of resistance plasmids. In contrast to mutations, the transfer of resistance plasmids requires the co-occurrence of both a donor and a recipient bacterial strain. For our experiments, we chose the facultative intracellular entero-pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) and Escherichia coli, a common member of the microbiota12. S. Typhimurium forms persisters that survive antibiotic therapy in several host tissues. Here we show that tissue-associated S. Typhimurium persisters represent long-lived reservoirs of plasmid donors or recipients. The formation of reservoirs of S. Typhimurium persisters requires Salmonella pathogenicity island (SPI)-1 and/or SPI-2 in gut-associated tissues, or SPI-2 at systemic sites. The re-seeding of these persister bacteria into the gut lumen enables the co-occurrence of donors with gut-resident recipients, and thereby favours plasmid transfer between various strains of Enterobacteriaceae. We observe up to 99% transconjugants within two to three days of re-seeding. Mathematical modelling shows that rare re-seeding events may suffice for a high frequency of conjugation. Vaccination reduces the formation of reservoirs of persisters after oral infection with S. Typhimurium, as well as subsequent plasmid transfer. We conclude that-even without selection for plasmid-encoded resistance genes-small reservoirs of pathogen persisters can foster the spread of promiscuous resistance plasmids in the gut.


Subject(s)
Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Gastrointestinal Microbiome/genetics , Gene Transfer, Horizontal , Intestinal Mucosa/microbiology , Plasmids/genetics , Salmonella typhimurium/genetics , Animals , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Mice , Models, Theoretical , Salmonella typhimurium/drug effects , Vaccination
17.
Front Microbiol ; 8: 2044, 2017.
Article in English | MEDLINE | ID: mdl-29104568

ABSTRACT

To understand how bacteria evolve and adapt to their environment, it can be relevant to monitor phenotypic changes that occur in a population. Single cell level analyses and sorting of mutant cells according to a particular phenotypic readout can constitute efficient strategies. However, when the phenotype of interest is expressed heterogeneously in ancestral isogenic populations of cells, single cell level sorting approaches are not optimal. Phenotypic heterogeneity can for instance make no-expression mutant cells indistinguishable from a subpopulation of wild-type cells transiently not expressing the phenotype. The analysis of clonal populations (e.g., isolated colonies), in which the average phenotype is measured, can circumvent this issue. Indeed, no-expression mutants form negative populations while wild-type clones form populations in which average expression of the phenotype yields a positive signal. We present here an optimized colony immunoblot protocol and a semi-automated image analysis pipeline (ImageJ macro) allowing for rapid detection of clones harboring mutations that affect the heterogeneous (i.e., bimodal) expression of the Type Three Secretion System-1 (TTSS-1) in Salmonella enterica serovar Typhimurium. We show that this protocol can efficiently differentiate clones expressing TTSS-1 at various levels in mixed populations. We were able to detect the emergence of hilC mutants in which the proportion of cells expressing TTSS-1 was reduced compared to the ancestor. We could also follow changes in the frequency of different mutants during long-term infections. This demonstrates that our protocol constitutes a tractable approach to assess semi-quantitatively the evolutionary dynamics of heterogeneous phenotypes, such as the expression of virulence genes, in bacterial populations.

18.
Nature ; 544(7651): 498-502, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28405025

ABSTRACT

Vaccine-induced high-avidity IgA can protect against bacterial enteropathogens by directly neutralizing virulence factors or by poorly defined mechanisms that physically impede bacterial interactions with the gut tissues ('immune exclusion'). IgA-mediated cross-linking clumps bacteria in the gut lumen and is critical for protection against infection by non-typhoidal Salmonella enterica subspecies enterica serovar Typhimurium (S. Typhimurium). However, classical agglutination, which was thought to drive this process, is efficient only at high pathogen densities (≥108 non-motile bacteria per gram). In typical infections, much lower densities (100-107 colony-forming units per gram) of rapidly dividing bacteria are present in the gut lumen. Here we show that a different physical process drives formation of clumps in vivo: IgA-mediated cross-linking enchains daughter cells, preventing their separation after division, and clumping is therefore dependent on growth. Enchained growth is effective at all realistic pathogen densities, and accelerates pathogen clearance from the gut lumen. Furthermore, IgA enchains plasmid-donor and -recipient clones into separate clumps, impeding conjugative plasmid transfer in vivo. Enchained growth is therefore a mechanism by which IgA can disarm and clear potentially invasive species from the intestinal lumen without requiring high pathogen densities, inflammation or bacterial killing. Furthermore, our results reveal an untapped potential for oral vaccines in combating the spread of antimicrobial resistance.


Subject(s)
Antibody Affinity , Immunoglobulin A/immunology , Intestines/immunology , Intestines/microbiology , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology , Animals , Bacterial Adhesion , Bacterial Vaccines , Cecum/immunology , Cecum/microbiology , Colony Count, Microbial , Conjugation, Genetic , Female , Humans , Male , Mice , Plasmids/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/prevention & control , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity
19.
Science ; 355(6330): 1211-1215, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28302859

ABSTRACT

Bacteriophage transfer (lysogenic conversion) promotes bacterial virulence evolution. There is limited understanding of the factors that determine lysogenic conversion dynamics within infected hosts. A murine Salmonella Typhimurium (STm) diarrhea model was used to study the transfer of SopEΦ, a prophage from STm SL1344, to STm ATCC14028S. Gut inflammation and enteric disease triggered >55% lysogenic conversion of ATCC14028S within 3 days. Without inflammation, SopEΦ transfer was reduced by up to 105-fold. This was because inflammation (e.g., reactive oxygen species, reactive nitrogen species, hypochlorite) triggers the bacterial SOS response, boosts expression of the phage antirepressor Tum, and thereby promotes free phage production and subsequent transfer. Mucosal vaccination prevented a dense intestinal STm population from inducing inflammation and consequently abolished SopEΦ transfer. Vaccination may be a general strategy for blocking pathogen evolution that requires disease-driven transfer of temperate bacteriophages.


Subject(s)
Diarrhea/microbiology , Diarrhea/pathology , Enteritis/microbiology , Lysogeny , Salmonella Phages/pathogenicity , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/virology , Animals , Disease Models, Animal , Enteritis/prevention & control , Inflammation/microbiology , Inflammation/prevention & control , Intestines/microbiology , Mice , Mice, Inbred C57BL , SOS Response, Genetics , Salmonella Phages/genetics , Vaccination , Viral Proteins/metabolism
20.
Cell Microbiol ; 19(6)2017 06.
Article in English | MEDLINE | ID: mdl-28061020

ABSTRACT

The pathogenic fungus Cryptococcus neoformans delivers virulence factors such as capsule polysaccharide to the cell surface to cause disease in vertebrate hosts. In this study, we screened for mutants sensitive to the secretion inhibitor brefeldin A to identify secretory pathway components that contribute to virulence. We identified an ortholog of the cell division control protein 50 (Cdc50) family of the noncatalytic subunit of type IV P-type ATPases (flippases) that establish phospholipid asymmetry in membranes and function in vesicle-mediated trafficking. We found that a cdc50 mutant in C. neoformans was defective for survival in macrophages, attenuated for virulence in mice and impaired in iron acquisition. The mutant also showed increased sensitivity to drugs associated with phospholipid metabolism (cinnamycin and miltefosine), the antifungal drug fluconazole and curcumin, an iron chelator that accumulates in the endoplasmic reticulum. Cdc50 is expected to function with catalytic subunits of flippases, and we previously documented the involvement of the flippase aminophospholipid translocases (Apt1) in virulence factor delivery. A comparison of phenotypes with mutants defective in genes encoding candidate flippases (designated APT1, APT2, APT3, and APT4) revealed similarities primarily between cdc50 and apt1 suggesting a potential functional interaction. Overall, these results highlight the importance of membrane composition and homeostasis for the ability of C. neoformans to cause disease.


Subject(s)
Cell Membrane/metabolism , Cryptococcus neoformans/pathogenicity , Fungal Proteins/genetics , Iron/metabolism , P-type ATPases/metabolism , Phospholipid Transfer Proteins/genetics , Animals , Antifungal Agents/pharmacology , Bacteriocins/pharmacology , Brefeldin A/pharmacology , Cell Division/physiology , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/metabolism , Curcumin/pharmacology , Endoplasmic Reticulum/metabolism , Female , Fluconazole/pharmacology , Fungal Polysaccharides/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , P-type ATPases/genetics , Peptides, Cyclic/pharmacology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...