Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 22(4): e3002597, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38684033

ABSTRACT

Intestinal epithelial cells (IECs) play pivotal roles in nutrient uptake and in the protection against gut microorganisms. However, certain enteric pathogens, such as Salmonella enterica serovar Typhimurium (S. Tm), can invade IECs by employing flagella and type III secretion systems (T3SSs) with cognate effector proteins and exploit IECs as a replicative niche. Detection of flagella or T3SS proteins by IECs results in rapid host cell responses, i.e., the activation of inflammasomes. Here, we introduce a single-cell manipulation technology based on fluidic force microscopy (FluidFM) that enables direct bacteria delivery into the cytosol of single IECs within a murine enteroid monolayer. This approach allows to specifically study pathogen-host cell interactions in the cytosol uncoupled from preceding events such as docking, initiation of uptake, or vacuole escape. Consistent with current understanding, we show using a live-cell inflammasome reporter that exposure of the IEC cytosol to S. Tm induces NAIP/NLRC4 inflammasomes via its known ligands flagellin and T3SS rod and needle. Injected S. Tm mutants devoid of these invasion-relevant ligands were able to grow in the cytosol of IECs despite the absence of T3SS functions, suggesting that, in the absence of NAIP/NLRC4 inflammasome activation and the ensuing cell death, no effector-mediated host cell manipulation is required to render the epithelial cytosol growth-permissive for S. Tm. Overall, the experimental system to introduce S. Tm into single enteroid cells enables investigations into the molecular basis governing host-pathogen interactions in the cytosol with high spatiotemporal resolution.


Subject(s)
Calcium-Binding Proteins , Cytosol , Flagellin , Host-Pathogen Interactions , Inflammasomes , Salmonella typhimurium , Type III Secretion Systems , Cytosol/metabolism , Cytosol/microbiology , Animals , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/metabolism , Type III Secretion Systems/metabolism , Inflammasomes/metabolism , Mice , Flagellin/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Neuronal Apoptosis-Inhibitory Protein/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Mice, Inbred C57BL , CARD Signaling Adaptor Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , Single-Cell Analysis/methods , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Salmonella Infections/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism
2.
Nat Microbiol ; 9(4): 1103-1116, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38503975

ABSTRACT

Microbiomes feature recurrent compositional structures under given environmental conditions. However, these patterns may conceal diverse underlying population dynamics that require intrastrain resolution. Here we developed a genomic tagging system, termed wild-type isogenic standardized hybrid (WISH)-tags, that can be combined with quantitative polymerase chain reaction and next-generation sequencing for microbial strain enumeration. We experimentally validated the performance of 62 tags and showed that they can be differentiated with high precision. WISH-tags were introduced into model and non-model bacterial members of the mouse and plant microbiota. Intrastrain priority effects were tested using one species of isogenic barcoded bacteria in the murine gut and the Arabidopsis phyllosphere, both with and without microbiota context. We observed colonization resistance against late-arriving strains of Salmonella Typhimurium in the mouse gut, whereas the phyllosphere accommodated Sphingomonas latecomers in a manner proportional to their presence at the late inoculation timepoint. This demonstrates that WISH-tags are a resource for deciphering population dynamics underlying microbiome assembly across biological systems.


Subject(s)
Microbiota , Animals , Mice , Microbiota/genetics , Salmonella typhimurium/genetics , Bacteria , Population Dynamics
3.
Bioinformatics ; 40(2)2024 02 01.
Article in English | MEDLINE | ID: mdl-38341646

ABSTRACT

MOTIVATION: DNA barcoding has become a powerful tool for assessing the fitness of strains in a variety of studies, including random transposon mutagenesis screens, attenuation of site-directed mutants, and population dynamics of isogenic strain pools. However, the statistical analysis, visualization, and contextualization of the data resulting from such experiments can be complex and require bioinformatic skills. RESULTS: Here, we developed mBARq, a user-friendly tool designed to simplify these steps for diverse experimental setups. The tool is seamlessly integrated with an intuitive web app for interactive data exploration via the STRING and KEGG databases to accelerate scientific discovery. AVAILABILITY AND IMPLEMENTATION: The tool is implemented in Python. The source code is freely available (https://github.com/MicrobiologyETHZ/mbarq) and the web app can be accessed at: https://microbiomics.io/tools/mbarq-app.


Subject(s)
DNA Barcoding, Taxonomic , Software , DNA , Computational Biology
4.
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
5.
PLoS Pathog ; 19(6): e1011235, 2023 06.
Article in English | MEDLINE | ID: mdl-37384776

ABSTRACT

Recruitment of neutrophils into and across the gut mucosa is a cardinal feature of intestinal inflammation in response to enteric infections. Previous work using the model pathogen Salmonella enterica serovar Typhimurium (S.Tm) established that invasion of intestinal epithelial cells by S.Tm leads to recruitment of neutrophils into the gut lumen, where they can reduce pathogen loads transiently. Notably, a fraction of the pathogen population can survive this defense, re-grow to high density, and continue triggering enteropathy. However, the functions of intraluminal neutrophils in the defense against enteric pathogens and their effects on preventing or aggravating epithelial damage are still not fully understood. Here, we address this question via neutrophil depletion in different mouse models of Salmonella colitis, which differ in their degree of enteropathy. In an antibiotic pretreated mouse model, neutrophil depletion by an anti-Ly6G antibody exacerbated epithelial damage. This could be linked to compromised neutrophil-mediated elimination and reduced physical blocking of the gut-luminal S.Tm population, such that the pathogen density remained high near the epithelial surface throughout the infection. Control infections with a ssaV mutant and gentamicin-mediated elimination of gut-luminal pathogens further supported that neutrophils are protecting the luminal surface of the gut epithelium. Neutrophil depletion in germ-free and gnotobiotic mice hinted that the microbiota can modulate the infection kinetics and ameliorate epithelium-disruptive enteropathy even in the absence of neutrophil-protection. Together, our data indicate that the well-known protective effect of the microbiota is augmented by intraluminal neutrophils. After antibiotic-mediated microbiota disruption, neutrophils are central for maintaining epithelial barrier integrity during acute Salmonella-induced gut inflammation, by limiting the sustained pathogen assault on the epithelium in a critical window of the infection.


Subject(s)
Neutrophils , Salmonella Infections , Animals , Mice , Salmonella typhimurium , Epithelial Cells , Anti-Bacterial Agents , Inflammation , Epithelium , Intestinal Mucosa
6.
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
7.
PLoS Pathog ; 16(5): e1008503, 2020 05.
Article in English | MEDLINE | ID: mdl-32365138

ABSTRACT

Salmonella enterica serovar Typhimurium (S.Tm) infections of cultured cell lines have given rise to the ruffle model for epithelial cell invasion. According to this model, the Type-Three-Secretion-System-1 (TTSS-1) effectors SopB, SopE and SopE2 drive an explosive actin nucleation cascade, resulting in large lamellipodia- and filopodia-containing ruffles and cooperative S.Tm uptake. However, cell line experiments poorly recapitulate many of the cell and tissue features encountered in the host's gut mucosa. Here, we employed bacterial genetics and multiple imaging modalities to compare S.Tm invasion of cultured epithelial cell lines and the gut absorptive epithelium in vivo in mice. In contrast to the prevailing ruffle-model, we find that absorptive epithelial cell entry in the mouse gut occurs through "discreet-invasion". This distinct entry mode requires the conserved TTSS-1 effector SipA, involves modest elongation of local microvilli in the absence of expansive ruffles, and does not favor cooperative invasion. Discreet-invasion preferentially targets apicolateral hot spots at cell-cell junctions and shows strong dependence on local cell neighborhood. This proof-of-principle evidence challenges the current model for how S.Tm can enter gut absorptive epithelial cells in their intact in vivo context.


Subject(s)
Bacterial Adhesion , Intestinal Mucosa/microbiology , Salmonella Infections , Salmonella typhimurium , Type I Secretion Systems/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Dogs , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Salmonella Infections/genetics , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Type I Secretion Systems/genetics
8.
Cell Host Microbe ; 27(6): 922-936.e6, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32416061

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.


Subject(s)
Aspartic Acid/metabolism , Fumarates/metabolism , Gastrointestinal Microbiome/physiology , Malates/metabolism , Salmonella/metabolism , Administration, Oral , Animals , Aspartic Acid/administration & dosage , Bacterial Proteins/metabolism , Citric Acid Cycle , Disease Models, Animal , Escherichia coli/metabolism , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Intestines/microbiology , Malates/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mutagenesis , RNA, Ribosomal, 16S/genetics , Salmonella/genetics , Salmonella/growth & development , Salmonella typhimurium , Sequence Analysis, DNA , Succinic Acid
9.
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
10.
PLoS One ; 13(10): e0206214, 2018.
Article in English | MEDLINE | ID: mdl-30359438

ABSTRACT

BACKGROUND AND AIMS: Lactulose is a common food ingredient and widely used as a treatment for constipation or hepatic encephalopathy and a substrate for hydrogen breath tests. Lactulose is fermented by the colon microbiota resulting in the production of hydrogen (H2). H2 is a substrate for enteropathogens including Salmonella Typhimurium (S. Typhimurium) and increased H2 production upon lactulose ingestion might favor the growth of H2-consuming enteropathogens. We aimed to analyze effects of single-dose lactulose ingestion on the growth of intrinsic Escherichia coli (E. coli), which can be efficiently quantified by plating and which share most metabolic requirements with S. Typhimurium. METHODS: 32 healthy volunteers (18 females, 14 males) were recruited. Participants were randomized for single-dose ingestion of 50 g lactulose or 50 g sucrose (controls). After ingestion, H2 in expiratory air and symptoms were recorded. Stool samples were acquired at days -1, 1 and 14. We analyzed 16S microbiota composition and abundance and characteristics of E. coli isolates. RESULTS: Lactulose ingestion resulted in diarrhea in 14/17 individuals. In 14/17 individuals, H2-levels in expiratory air increased by ≥20 ppm within 3 hours after lactulose challenge. H2-levels correlated with the number of defecations within 6 hours. E. coli was detectable in feces of all subjects (2 x 10(2)-10(9) CFU/g). However, the number of E. coli colony forming units (CFU) on selective media did not differ between any time point before or after challenge with sucrose or lactulose. The microbiota composition also remained stable upon lactulose exposure. CONCLUSION: Ingestion of a single dose of 50 g lactulose does not significantly alter E. coli density in stool samples of healthy volunteers. 50 g lactulose therefore seems unlikely to sufficiently alter growth conditions in the intestine for a significant predisposition to infection with H2-consuming enteropathogens such as S. Typhimurium (www.clinicaltrials.gov NCT02397512).


Subject(s)
Gastrointestinal Microbiome/drug effects , Lactulose/pharmacology , Microbial Viability/drug effects , Adult , Colon/drug effects , Colon/microbiology , Diarrhea/chemically induced , Diarrhea/microbiology , Dietary Sucrose/pharmacology , Dose-Response Relationship, Drug , Feces/microbiology , Female , Healthy Volunteers , Humans , Male , Middle Aged , Young Adult
11.
Cell Host Microbe ; 21(4): 443-454, 2017 Apr 12.
Article in English | MEDLINE | ID: mdl-28407482

ABSTRACT

Despite decades of research, efficient therapies for most enteropathogenic bacteria are still lacking. In this review, we focus on Salmonella enterica Typhimurium (S. Typhimurium), a frequent cause of acute, self-limiting food-borne diarrhea and a model that has revealed key principles of enteropathogen infection. We review the steps of gut infection and the mucosal innate-immune defenses limiting pathogen burdens, and we discuss how inflammation boosts gut luminal S. Typhimurium growth. We also discuss how S. Typhimurium-induced inflammation accelerates the transfer of plasmids and phages, which may promote the transmission of antibiotic resistance and facilitate emergence of pathobionts and pathogens with enhanced virulence. The targeted manipulation of the microbiota and vaccination might offer strategies to prevent this evolution. As gut luminal microbes impact various aspects of the host's physiology, improved strategies for preventing enteropathogen infection and disease-inflicted DNA exchange may be of broad interest well beyond the acute infection.


Subject(s)
Diarrhea/microbiology , Diarrhea/pathology , Gene Transfer, Horizontal , Host-Pathogen Interactions , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/pathogenicity , Immunity, Innate , Interspersed Repetitive Sequences , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development
12.
Cell Host Microbe ; 20(2): 238-49, 2016 Aug 10.
Article in English | MEDLINE | ID: mdl-27453483

ABSTRACT

Salmonella Typhimurium (S.Tm) causes acute enteropathy resolving after 4-7 days. Strikingly, antibiotic therapy does not accelerate disease resolution. We screened for factors blocking remission using a S.Tm enterocolitis model. The antibiotic ciprofloxacin clears pathogen stool loads within 3-24 hr, while gut pathology resolves more slowly (ψ50: ∼48 hr, remission: 6-9 days). This delayed resolution is mediated by an interferon-γ (IFN-γ)-dependent response that is triggered during acute infection and continues throughout therapy. Specifically, IFN-γ production by mucosal T and NK cells retards disease resolution by maintaining signaling through the transcriptional regulator STAT1 and boosting expression of inflammatory mediators like IL-1ß, TNF, and iNOS. Additionally, sustained IFN-γ fosters phagocyte accumulation and hampers antimicrobial defense mediated by IL-22 and the lectin REGIIIß. These findings reveal a role for IFN-γ in delaying resolution of intestinal inflammation and may inform therapies for acute Salmonella enteropathy, chronic inflammatory bowel diseases, or disease resolution during antibiotic treatment.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Enterocolitis/pathology , Gastrointestinal Tract/pathology , Interferon-gamma/metabolism , Salmonella Infections/pathology , Salmonella typhimurium/growth & development , Animals , Disease Models, Animal , Enterocolitis/drug therapy , Enterocolitis/immunology , Enterocolitis/microbiology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Immunologic Factors/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Killer Cells, Natural/immunology , Mice, Inbred C57BL , Phagocytes/immunology , Salmonella Infections/drug therapy , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/drug effects , Signal Transduction , T-Lymphocytes/immunology
13.
Cell Host Microbe ; 17(5): 716-25, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25920978

ABSTRACT

Gene inactivation by transposon insertion or allelic exchange is a powerful approach to probe gene function. Unfortunately, many microbes, including Chlamydia, are not amenable to routine molecular genetic manipulations. Here we describe an arrayed library of chemically induced mutants of the genetically intransigent pathogen Chlamydia trachomatis, in which all mutations have been identified by whole-genome sequencing, providing a platform for reverse genetic applications. An analysis of possible loss-of-function mutations in the collection uncovered plasticity in the central metabolic properties of this obligate intracellular pathogen. We also describe the use of the library in a forward genetic screen that identified InaC as a bacterial factor that binds host ARF and 14-3-3 proteins and modulates F-actin assembly and Golgi redistribution around the pathogenic vacuole. This work provides a robust platform for reverse and forward genetic approaches in Chlamydia and should serve as a valuable resource to the community.


Subject(s)
Chlamydia trachomatis/genetics , Genetics, Microbial/methods , Genome, Bacterial , Molecular Biology/methods , Mutagenesis , Mutation , Reverse Genetics/methods , Genetic Markers , Genetic Testing , Sequence Analysis, DNA
14.
Methods Mol Biol ; 1197: 347-58, 2014.
Article in English | MEDLINE | ID: mdl-25172291

ABSTRACT

Our understanding of how most microbes "work" is hindered by the lack of molecular genetic and recombinant DNA tools to manipulate their genomes. We devised an approach to perform genetic analysis in one such microbe, the obligate intracellular bacterial pathogen Chlamydia trachomatis. Comprehensive libraries of clone-purified mutants with distinct plaque morphologies were generated through chemical mutagenesis. Whole-genome sequencing (WGS) was then employed to identify the underlying genetic lesions and to draw correlations between mutated gene(s) and a common phenotype. Taking advantage of the ability of Chlamydia to exchange DNA in co-infection settings, we then generated recombinant strains after co-infection of mammalian cells with mutant and wild type bacteria. In this manner, causal relationships between genotypes and phenotypes were established. The pairing of chemically induced gene variation and WGS to establish correlative genotype-phenotype associations should be broadly applicable to a large list of medically and environmentally important microorganisms currently not amenable to genetic analysis.


Subject(s)
Chlamydia trachomatis/genetics , Chlamydia trachomatis/pathogenicity , Mutagenesis , Virulence/genetics , Genotype , Phenotype
15.
PLoS One ; 9(6): e100763, 2014.
Article in English | MEDLINE | ID: mdl-24955832

ABSTRACT

Chlamydia trachomatis is an obligate intracellular bacterial pathogen and the second leading cause of sexually transmitted infections in the US. Infections cause significant morbidity and can lead to serious reproductive sequelae, including an epidemiological link to increased rates of reproductive cancers. One of the overt changes that infected cells exhibit is the development of genomic instability leading to multinucleation. Here we demonstrate that the induction of multinucleation is not conserved equally across chlamydial species; C. trachomatis L2 caused high levels of multinucleation, C. muridarum intermediate levels, and C. caviae had very modest effects on multinucleation. Our data show that at least two effector pathways together cause genomic instability during infection leading to multinucleation. We find that the highly conserved chlamydial protease CPAF is a key effector for one of these pathways. CPAF secretion is required for the loss of centrosome duplication regulation as well as inducing early mitotic exit. The second effector pathway involves the induction of centrosome position errors. This function is not conserved in three chlamydial species tested. Together these two pathways contribute to the induction of high levels of genomic instability and multinucleation seen in C. trachomatis infections.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia trachomatis/physiology , Giant Cells/microbiology , Giant Cells/pathology , Signal Transduction , 3T3 Cells , Animals , Centrosome , Chromosome Segregation , DNA, Bacterial/metabolism , Endopeptidases/metabolism , Fluorescent Antibody Technique , Griseofulvin/pharmacology , HeLa Cells , Humans , Mice , Mitotic Index , Models, Biological , Mutation , Species Specificity , Spindle Apparatus/metabolism
16.
Pathog Dis ; 71(3): 336-51, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24838663

ABSTRACT

The secreted Chlamydia protease CPAF cleaves a defined set of mammalian and Chlamydia proteins in vitro. As a result, this protease has been proposed to modulate a range of bacterial and host cellular functions. However, it has recently come into question the extent to which many of its identified substrates constitute bona fide targets of proteolysis in infected host cell rather than artifacts of postlysis degradation. Here, we clarify the role played by CPAF in cellular models of infection by analyzing Chlamydia trachomatis mutants deficient for CPAF activity. Using reverse genetic approaches, we identified two C. trachomatis strains possessing nonsense, loss-of-function mutations in cpa (CT858) and a third strain containing a mutation in type II secretion (T2S) machinery that inhibited CPAF activity by blocking zymogen secretion and subsequent proteolytic maturation into the active hydrolase. HeLa cells infected with T2S(-) or CPAF(-) C. trachomatis mutants lacked detectable in vitro CPAF proteolytic activity and were not defective for cellular traits that have been previously attributed to CPAF activity, including resistance to staurosporine-induced apoptosis, Golgi fragmentation, altered NFκB-dependent gene expression, and resistance to reinfection. However, CPAF-deficient mutants did display impaired generation of infectious elementary bodies (EBs), indicating an important role for this protease in the full replicative potential of C. trachomatis. In addition, we provide compelling evidence in live cells that CPAF-mediated protein processing of at least two host protein targets, vimentin filaments and the nuclear envelope protein lamin-associated protein-1 (LAP1), occurs rapidly after the loss of the inclusion membrane integrity, but before loss of plasma membrane permeability and cell lysis. CPAF-dependent processing of host proteins correlates with a loss of inclusion membrane integrity, and so we propose that CPAF plays a role late in infection, possibly during the stages leading to the dismantling of the infected cell prior to the release of EBs during cell lysis.


Subject(s)
Chlamydia trachomatis/enzymology , Host-Pathogen Interactions , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chlamydia trachomatis/genetics , Chlorocebus aethiops , Epithelial Cells/microbiology , HeLa Cells , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Peptide Hydrolases/deficiency , Protein Processing, Post-Translational , Proteolysis , Vero Cells
17.
J Vis Exp ; (80): e50636, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24192560

ABSTRACT

Chlamydia trachomatis, the etiological agent of sexually transmitted diseases and ocular infections, remains poorly characterized due to its intractability to experimental transformation with recombinant DNA. We developed an approach to perform genetic analysis in C. trachomatis despite the lack of molecular genetic tools. Our method involves: i.) chemical mutagenesis to rapidly generate comprehensive libraries of genetically-defined mutants with distinct phenotypes; ii.) whole-genome sequencing (WGS) to map the underlying genetic lesions and to find associations between mutated gene(s) and a common phenotype; iii.) generation of recombinant strains through co-infection of mammalian cells with mutant and wild type bacteria. Accordingly, we were able to establish causal relationships between genotypes and phenotypes. The coupling of chemically-induced gene variation and WGS to establish correlative genotype-phenotype associations should be broadly applicable to the large list of medically and environmentally important microorganisms currently intractable to genetic analysis.


Subject(s)
Chlamydia trachomatis/genetics , Genome, Bacterial , Genomics/methods , Sequence Analysis, DNA/methods , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Mutagenesis
18.
J Bacteriol ; 195(18): 4221-30, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23852872

ABSTRACT

Salicylidene acylhydrazides (SAHs) inhibit the type III secretion system (T3S) of Yersinia and other Gram-negative bacteria. In addition, SAHs restrict the growth and development of Chlamydia species. However, since the inhibition of Chlamydia growth by SAH is suppressed by the addition of excess iron and since SAHs have an iron-chelating capacity, their role as specific T3S inhibitors is unclear. We investigated here whether SAHs exhibit a function on C. trachomatis that goes beyond iron chelation. We found that the iron-saturated SAH INP0341 (IS-INP0341) specifically affects C. trachomatis infectivity with reduced generation of infectious elementary body (EB) progeny. Selection and isolation of spontaneous SAH-resistant mutant strains revealed that mutations in hemG suppressed the reduced infectivity caused by IS-INP0341 treatment. Structural modeling of C. trachomatis HemG predicts that the acquired mutations are located in the active site of the enzyme, suggesting that IS-INP0341 inhibits this domain of HemG and that protoporphyrinogen oxidase (HemG) and heme metabolism are important for C. trachomatis infectivity.


Subject(s)
Bacterial Proteins/genetics , Chlamydia trachomatis/drug effects , Chlamydia trachomatis/genetics , Hydrazines/pharmacology , Mutation , Protoporphyrinogen Oxidase/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Chlamydia trachomatis/enzymology , Chlamydia trachomatis/pathogenicity , Drug Resistance, Bacterial , HeLa Cells , Heme/metabolism , Humans , Iron/metabolism , Iron/pharmacology , Models, Molecular , Molecular Sequence Data , Protoporphyrinogen Oxidase/chemistry , Protoporphyrinogen Oxidase/metabolism
19.
Proc Natl Acad Sci U S A ; 109(4): 1263-8, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22232666

ABSTRACT

Chlamydia trachomatis, a pathogen responsible for diseases of significant clinical and public health importance, remains poorly characterized because of its intractability to routine molecular genetic manipulation. We have developed a combinatorial approach to rapidly generate a comprehensive library of genetically defined mutants. Chemical mutagenesis, coupled with whole-genome sequencing (WGS) and a system for DNA exchange within infected cells, was used to generate Chlamydia mutants with distinct phenotypes, map the underlying genetic lesions, and generate isogenic strains. As a result, we identified mutants with altered glycogen metabolism, including an attenuated strain defective for type II secretion. The coupling of chemically induced gene variation and WGS to establish genotype-phenotype associations should be broadly applicable to the large list of medically and environmentally important microorganisms currently intractable to genetic analysis.


Subject(s)
Chlamydia trachomatis/genetics , Chlamydia trachomatis/pathogenicity , Gene Library , Genetic Techniques , Phenotype , Virulence Factors/genetics , Animals , Base Sequence , Chlorocebus aethiops , Genomics/methods , Genotype , HeLa Cells , Humans , Immunohistochemistry , Microscopy, Electron, Transmission , Molecular Sequence Data , Mutagenesis/genetics , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA/methods , Vero Cells
20.
Proc Natl Acad Sci U S A ; 108(25): 10284-9, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21628561

ABSTRACT

Lipopolysaccharides (LPS) and lipooligosaccharides (LOS) are the main lipid components of bacterial outer membranes and are essential for cell viability in most Gram-negative bacteria. Here we show that small molecule inhibitors of LpxC [UDP-3-O-(R-3-hydroxymyristoyl)-GlcNAc deacetylase], the enzyme that catalyzes the first committed step in the biosynthesis of lipid A, block the synthesis of LOS in the obligate intracellular bacterial pathogen Chlamydia trachomatis. In the absence of LOS, Chlamydia remains viable and establishes a pathogenic vacuole ("inclusion") that supports robust bacterial replication. However, bacteria grown under these conditions were no longer infectious. In the presence of LpxC inhibitors, replicative reticulate bodies accumulated in enlarged inclusions but failed to express selected late-stage proteins and transition to elementary bodies, a Chlamydia developmental form that is required for invasion of mammalian cells. These findings suggest the presence of an outer membrane quality control system that regulates Chlamydia developmental transition to infectious elementary bodies and highlights the potential application of LpxC inhibitors as unique class of antichlamydial agents.


Subject(s)
Chlamydia trachomatis/pathogenicity , Inclusion Bodies/metabolism , Lipopolysaccharides/biosynthesis , Amidohydrolases/antagonists & inhibitors , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chlamydia Infections , Chlamydia trachomatis/cytology , Chlamydia trachomatis/physiology , HeLa Cells , Humans , Lipid A/biosynthesis , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...