Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Helicobacter ; 24(4): e12587, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31062466

ABSTRACT

BACKGROUND: Helicobacter pylori represents an interesting model of bacterial pathogenesis given that most infections are asymptomatic, while a minority of infections cause severe gastric disease. H pylori strain B128 7.13 is used extensively to understand H pylori pathophysiology. Due to extensive restriction-modification systems, the fact that only some H pylori strains are naturally transformable, the inability of common plasmid and transposon vectors to replicate in this bacterium, as well as the limited number of antibiotic cassettes that are functional in H pylori, there are relatively few genetic tools for the mutagenesis of this bacterium. MATERIALS AND METHODS: Here, we use PacBio and Illumina sequencing to reveal the complete genome sequence of H pylori B128 7.13. Furthermore, we describe a system to generate markerless and scarless mutations on the H pylori chromosome using the counter-selection marker, galactokinase from Escherichia coli. RESULTS: We show that this mutagenesis strategy can be used to generate in-frame insertions, gene deletions, and multiple independent mutations in B128 7.13. Using the closed genome as a reference, we also report the absence of second site chromosomal mutations and/or rearrangements in our mutagenized strains. We compare the genome sequence of H pylori B128 7.13 with a closely related strain, H pylori B8, and reveal one notable region of difference, which is a 1430 bp insertion encoding a H pylori-specific DUF874 family protein of unknown function. CONCLUSIONS: This article reports the closed genome of the important H pylori B128 7.13 strain and a mutagenesis method that can be adopted by researchers as an alternative strategy to generate isogenic mutants of H pylori in order to further our understanding of this bacterium.


Subject(s)
Genetic Techniques , Genome, Bacterial , Helicobacter pylori/genetics , Base Sequence , Helicobacter Infections/microbiology , Helicobacter pylori/classification , Helicobacter pylori/isolation & purification , Humans , Mutagenesis , Mutation , Whole Genome Sequencing
2.
Infect Immun ; 83(4): 1384-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25624355

ABSTRACT

The evolution of pathogenic bacteria is a multifaceted and complex process, which is strongly influenced by the horizontal acquisition of genetic elements and their subsequent expression in their new hosts. A well-studied example is the RegA regulon of the enteric pathogen Citrobacter rodentium. The RegA regulatory protein is a member of the AraC/XylS superfamily, which coordinates the expression of a gene repertoire that is necessary for full pathogenicity of this murine pathogen. Upon stimulation by an exogenous, gut-associated signal, namely, bicarbonate ions, RegA activates the expression of a series of genes, including virulence factors, such as autotransporters, fimbriae, a dispersin-like protein, and the grlRA operon on the locus of enterocyte effacement pathogenicity island. Interestingly, the genes encoding RegA homologues are distributed across the genus Escherichia, encompassing pathogenic and nonpathogenic subtypes. In this study, we carried out a series of bioinformatic, transcriptional, and functional analyses of the RegA regulons of these bacteria. Our results demonstrated that regA has been horizontally transferred to Escherichia spp. and C. rodentium. Comparative studies of two RegA homologues, namely, those from C. rodentium and E. coli SMS-3-5, a multiresistant environmental strain of E. coli, showed that the two regulators acted similarly in vitro but differed in terms of their abilities to activate the virulence of C. rodentium in vivo, which evidently was due to their differential activation of grlRA. Our data indicate that RegA from C. rodentium has strain-specific adaptations that facilitate infection of its murine host. These findings shed new light on the development of virulence by C. rodentium and on the evolution of virulence-regulatory genes of bacterial pathogens in general.


Subject(s)
AraC Transcription Factor/genetics , Bacterial Proteins/genetics , Citrobacter rodentium/genetics , Citrobacter rodentium/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Animals , Biological Evolution , Escherichia coli/pathogenicity , Gene Expression Regulation, Bacterial , Gene Transfer, Horizontal , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/genetics , Phylogeny , Repressor Proteins/genetics , Virulence Factors/genetics
3.
PLoS Pathog ; 9(1): e1003117, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23326233

ABSTRACT

The Type II Secretion System (T2SS) is a molecular machine that drives the secretion of fully-folded protein substrates across the bacterial outer membrane. A key element in the machinery is the secretin: an integral, multimeric outer membrane protein that forms the secretion pore. We show that three distinct forms of T2SSs can be distinguished based on the sequence characteristics of their secretin pores. Detailed comparative analysis of two of these, the Klebsiella-type and Vibrio-type, showed them to be further distinguished by the pilotin that mediates their transport and assembly into the outer membrane. We have determined the crystal structure of the novel pilotin AspS from Vibrio cholerae, demonstrating convergent evolution wherein AspS is functionally equivalent and yet structurally unrelated to the pilotins found in Klebsiella and other bacteria. AspS binds to a specific targeting sequence in the Vibrio-type secretins, enhances the kinetics of secretin assembly, and homologs of AspS are found in all species of Vibrio as well those few strains of Escherichia and Shigella that have acquired a Vibrio-type T2SS.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Secretion Systems/physiology , Porins/metabolism , Secretin/metabolism , Vibrio cholerae/metabolism , Amino Acid Sequence , Bacterial Outer Membrane Proteins/chemistry , Binding Sites/physiology , Computational Biology , Crystallization , Evolution, Molecular , Gene Expression Regulation, Bacterial , Klebsiella/physiology , Molecular Sequence Data , Phylogeny , Porins/chemistry , Protein Binding , Species Specificity , Vibrio cholerae/genetics
4.
Helicobacter ; 20(4): 269-83, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25669590

ABSTRACT

BACKGROUND: Multiple studies have established the importance of the tol-pal gene cluster in bacterial cell membrane integrity and outer membrane vesicle (OMV) formation in Escherichia coli. In contrast, the functions of Tol-Pal proteins in pathogenic organisms, including those of the Epsilonproteobacteria, remain poorly if at all defined. The aim of this study was to characterize the roles of two key components of the Tol-Pal system, TolB and Pal, in OMV formation in the pathogenic bacterium, Helicobacter pylori. METHODS: H. pylori ΔtolB, Δpal and ΔtolBpal mutants, as well as complemented strains, were generated and assessed for changes in morphology and OMV production by scanning electron microscopy and enzyme-linked immunoassay (ELISA), respectively. The protein content and pro-inflammatory properties of OMVs were determined by mass spectroscopy and interleukin-8 (IL-8) ELISA on culture supernatants from OMV-stimulated cells, respectively. RESULTS: H. pylori ΔtolB and Δpal bacteria exhibited aberrant cell morphology and/or flagella biosynthesis. Importantly, the disruption of H. pylori tolB but not pal resulted in a significant increase in OMV production. The OMVs from H. pylori ΔtolB and Δpal bacteria harbored many of the major outer membrane and virulence proteins observed in wild-type (WT) OMVs. Interestingly, ΔtolB, Δpal and ΔtolBpal OMVs induced significantly higher levels of IL-8 production by host cells, compared with WT OMVs. CONCLUSIONS: This work demonstrates that TolB and Pal are important for membrane integrity in H. pylori. Moreover, it shows how H. pylori tolB-pal genes may be manipulated to develop "hypervesiculating" strains for vaccine purposes.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Interleukin-8/metabolism , Periplasmic Proteins/metabolism , Cell Membrane/physiology , Enzyme-Linked Immunospot Assay , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Mass Spectrometry , Microscopy, Electron, Scanning , Periplasmic Proteins/genetics
5.
Infect Immun ; 81(4): 1078-89, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23340312

ABSTRACT

AraC-like regulators play a key role in the expression of virulence factors in enteric pathogens, such as enteropathogenic Escherichia coli (EPEC), enterotoxigenic E. coli, enteroaggregative E. coli, and Citrobacter rodentium. Bioinformatic analysis of the genome of rabbit-specific EPEC (REPEC) strain E22 (O103:H2) revealed the presence of a gene encoding an AraC-like regulatory protein, RegR, which shares 71% identity to the global virulence regulator, RegA, of C. rodentium. Microarray analysis demonstrated that RegR exerts 25- to 400-fold activation on transcription of several genes encoding putative virulence-associated factors, including a fimbrial operon (SEF14), a serine protease, and an autotransporter adhesin. These observations were confirmed by proteomic analysis of secreted and heat-extracted surface-associated proteins. The mechanism of RegR-mediated activation was investigated by using its most highly upregulated gene target, sefA. Transcriptional analyses and electrophoretic mobility shift assays showed that RegR activates the expression of sefA by binding to a region upstream of the sefA promoter, thereby relieving gene silencing by the global regulatory protein H-NS. Moreover, RegR was found to contribute significantly to virulence in a rabbit infection experiment. Taken together, our findings indicate that RegR controls the expression of a series of accessory adhesins that significantly enhance the virulence of REPEC strain E22.


Subject(s)
Bacterial Proteins/metabolism , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Gene Expression Regulation, Bacterial , Regulon , Transcription Factors/metabolism , Virulence Factors/biosynthesis , Animals , Bacterial Proteins/genetics , Computational Biology , DNA, Bacterial/metabolism , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Proteins/analysis , Fimbriae Proteins/biosynthesis , Fimbriae Proteins/genetics , Gene Expression Profiling , Microarray Analysis , Promoter Regions, Genetic , Protein Binding , Proteome/analysis , Rabbits , Transcription Factors/genetics , Transcription, Genetic , Virulence Factors/genetics
6.
Infect Immun ; 80(6): 2042-52, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22451516

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) is a major cause of diarrhea in infants in developing countries. We have identified a functional type II secretion system (T2SS) in EPEC that is homologous to the pathway responsible for the secretion of heat-labile enterotoxin by enterotoxigenic E. coli. The wild-type EPEC T2SS was able to secrete a heat-labile enterotoxin reporter, but an isogenic T2SS mutant could not. We showed that the major substrate of the T2SS in EPEC is SslE, an outer membrane lipoprotein (formerly known as YghJ), and that a functional T2SS is essential for biofilm formation by EPEC. T2SS and SslE mutants were arrested at the microcolony stage of biofilm formation, suggesting that the T2SS is involved in the development of mature biofilms and that SslE is a dominant effector of biofilm development. Moreover, the T2SS was required for virulence, as infection of rabbits with a rabbit-specific EPEC strain carrying a mutation in either the T2SS or SslE resulted in significantly reduced intestinal colonization and milder disease.


Subject(s)
Biofilms/growth & development , Enteropathogenic Escherichia coli/physiology , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Virulence Factors/metabolism , Animals , Cell Membrane , Enteropathogenic Escherichia coli/cytology , Enteropathogenic Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Mutation , Rabbits , Substrate Specificity , Virulence , Virulence Factors/genetics
7.
Appl Environ Microbiol ; 78(15): 5083-92, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22582067

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a lethal human intestinal pathogen that causes hemorrhagic colitis and the hemolytic-uremic syndrome. EHEC is transmitted by the fecal-oral route and has a lower infectious dose than most other enteric bacterial pathogens in that fewer than 100 CFU are able to cause disease. This low infectious dose has been attributed to the ability of EHEC to survive in the acidic environment of the human stomach. In silico analysis of the genome of EHEC O157:H7 strain EDL933 revealed a gene, patE, for a putative AraC-like regulatory protein within the prophage island, CP-933H. Transcriptional analysis in E. coli showed that the expression of patE is induced during stationary phase. Data from microarray assays demonstrated that PatE activates the transcription of genes encoding proteins of acid resistance pathways. In addition, PatE downregulated the expression of a number of genes encoding heat shock proteins and the type III secretion pathway of EDL933. Transcriptional analysis and electrophoretic mobility shift assays suggested that PatE also activates the transcription of the gene for the acid stress chaperone hdeA by binding to its promoter region. Finally, assays of acid tolerance showed that increasing the expression of PatE in EHEC greatly enhanced the ability of the bacteria to survive in different acidic environments. Together, these findings indicate that EHEC strain EDL933 carries a prophage-encoded regulatory system that contributes to acid resistance.


Subject(s)
Adaptation, Biological/genetics , AraC Transcription Factor/genetics , Escherichia coli O157/physiology , Gastric Acid/metabolism , Metabolic Networks and Pathways/genetics , Trans-Activators/metabolism , Transcriptional Activation/physiology , AraC Transcription Factor/physiology , Electrophoretic Mobility Shift Assay , Genomics/methods , Humans , Metabolic Networks and Pathways/physiology , Microarray Analysis , Trans-Activators/genetics
8.
J Bacteriol ; 193(7): 1777-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21278290

ABSTRACT

We identified several promoters responsible for the expression of regA, which encodes a global virulence regulator in Citrobacter rodentium. Expression of some of the promoters was strongly autoactivated by RegA in conjunction with bicarbonate. Biochemical and mutational analyses were used to determine the consensus sequence of the RegA-binding sites.


Subject(s)
Bacterial Proteins/metabolism , Citrobacter rodentium/metabolism , Citrobacter rodentium/pathogenicity , Gene Expression Regulation, Bacterial/physiology , Transcription, Genetic , Bacterial Proteins/genetics , Base Sequence , Citrobacter rodentium/genetics , DNA, Bacterial/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Virulence
9.
Front Microbiol ; 12: 651488, 2021.
Article in English | MEDLINE | ID: mdl-33815340

ABSTRACT

Enterotoxigenic E. coli (ETEC) is a common cause of diarrhea in children in low- and middle-income countries, and in travelers to these countries. ETEC is also an important cause of morbidity and premature mortality in piglets, calves, goat kids and lambs. The major virulence determinants of ETEC are enterotoxins and colonization factors, which enable the pathogen to colonize the small intestine and deliver enterotoxins, such as the heat-stable enterotoxins, STp and STh, to epithelial cells. Because most ETEC strains are host-specific, there are few convenient animal models to investigate the pathogenesis of ETEC infections or to evaluate specific anti-ETEC interventions, such as drugs and vaccines. An exception is ETEC strains bearing F41 pili, which mediate intestinal colonization of various young animals, including neonatal mice, to cause disease and in some cases death. In this study, we used the archetypal F41-producing bovine ETEC strain, B41 (O101:NM; K99, F41, STp) to validate and further explore the contribution of F41 and STp to bacterial virulence. By using targeted gene deletion and trans-complementation studies, augmented by whole genome sequencing, and in vitro and animal studies of virulence, we established that F41 mediates colonization of the mouse intestine and is essential for bacterial virulence. In addition, we showed for the first time that STp is as important as F41 for virulence. Together, these findings validate the use of neonatal mice to study the pathogenesis of F41-bearing ETEC and to investigate possible specific anti-ETEC interventions including vaccines that target heat-stable enterotoxins.

10.
J Bacteriol ; 192(14): 3722-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20472788

ABSTRACT

The locus for enterocyte effacement (LEE) is the virulence hallmark of the attaching-and-effacing (A/E) intestinal pathogens, namely, enteropathogenic Escherichia coli, enterohemorrhagic E. coli, and Citrobacter rodentium. The LEE carries more than 40 genes that are arranged in several operons, e.g., LEE1 to LEE5. Expression of the various transcriptional units is subject to xenogeneic silencing by the histone-like protein H-NS. The LEE1-encoded regulator, Ler, plays a key role in relieving this repression at several major LEE promoters, including LEE2 to LEE5. To achieve appropriate intracellular concentrations of Ler in different environments, A/E pathogens have evolved a sophisticated regulatory network to control ler expression. For example, the LEE-encoded GrlA and GrlR proteins work as activator and antiactivator, respectively, of ler transcription. Thus, control of the transcriptional activities of the LEE1 (ler) promoter and the grlRA operon determines the rate of transcription of all of the LEE-encoded virulence factors. To date, only a single promoter has been identified for the grlRA operon. In this study, we showed that the non-LEE-encoded AraC-like regulatory protein RegA of C. rodentium directly stimulates transcription of the grlRA promoter by binding to an upstream region in the presence of bicarbonate ions. In addition, in vivo and in vitro transcription assays revealed a sigma(70) promoter that is specifically responsible for transcription of grlA. Expression from this promoter was strongly repressed by H-NS and its paralog StpA but was activated by Ler. DNase I footprinting demonstrated that Ler binds to a region upstream of the grlA promoter, whereas H-NS interacts specifically with a region extending from the grlA core promoter into its coding sequence. Together, these findings provide new insights into the environmental regulation and differential expressions of the grlR and grlA genes of C. rodentium.


Subject(s)
Bacterial Proteins/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Gene Expression Regulation, Bacterial/physiology , Operon/genetics , Bacterial Proteins/genetics , Base Sequence , Chromosome Mapping , Citrobacter rodentium/pathogenicity , DNA, Bacterial , Gene Expression Regulation, Enzymologic/physiology , Molecular Sequence Data , Sodium Bicarbonate , Transcription Initiation Site , Transcription, Genetic , Virulence
11.
Immunol Cell Biol ; 80(6): 574-83, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12406392

ABSTRACT

The 65 kilodalton heat shock protein (Hsp65) from mycobacterial species elicits immune responses and in some cases protective immunity. Here we have used a DNA sublibrary approach to identify antigenic fragments of Mycobacterium avium Hsp65 and a synthetic peptide approach to delineate CD4+ T cell determinants. A panel of Hsp65 reactive CD4+ T cell clones was established from lymph node cells obtained from BALB/c mice immunized with recombinant Hsp65. The clones were tested for proliferative reactivity against the products of the DNA sublibrary of the hsp65 gene. A T cell epitope, restricted by the I-Ad molecule, was identified within the C-terminal region of Hsp65 and the minimal epitope (amino acid residues 489-503) delineated using overlapping peptides spanning the C-terminal fragment. Additionally, the CD4+ T cell clone recognizing this epitope also responded to native Hsp65 present in M. avium lysates by both proliferation and cytokine production, indicating that the epitope was present and processed similarly both in the native and the recombinant forms of Hsp65. This sequence identified in BALB/c mice (Hsp65 489-503) is identical in other mycobacteria, notably M. tuberculosis, M. bovis and M. leprae, suggesting the epitope may have wider application in murine models of other mycobacterial infections.


Subject(s)
Bacterial Proteins , Chaperonins/metabolism , Histocompatibility Antigens Class II/metabolism , Mycobacterium avium/metabolism , Peptides/metabolism , Amino Acid Motifs , Amino Acid Sequence , CD4-Positive T-Lymphocytes/immunology , Chaperonin 60 , Chaperonins/genetics , Chaperonins/immunology , Epitopes , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL