Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
mBio ; 9(5)2018 09 25.
Article in English | MEDLINE | ID: mdl-30254126

ABSTRACT

The intracellular human pathogen Shigella flexneri invades the colon epithelium, replicates to high cell density within the host cell, and then spreads to adjacent epithelial cells. When S. flexneri gains access to the host cytosol, the bacteria metabolize host cytosolic carbon using glycolysis and mixed acid fermentation, producing formate as a by-product. We show that S. flexneri infection results in the accumulation of formate within the host cell. Loss of pyruvate formate lyase (PFL; ΔpflB), which converts pyruvate to acetyl coenzyme A (CoA) and formate, eliminates S. flexneri formate production and reduces the ability of S. flexneri to form plaques in epithelial cell monolayers. This defect in PFL does not decrease the intracellular growth rate of S. flexneri; rather, it affects cell-to-cell spread. The S. flexneri ΔpflB mutant plaque defect is complemented by supplying exogenous formate; conversely, deletion of the S. flexneri formate dehydrogenase gene fdnG increases host cell formate accumulation and S. flexneri plaque size. Furthermore, exogenous formate increases plaque size of the wild-type (WT) S. flexneri strain and promotes S. flexneri cell-to-cell spread. We also demonstrate that formate increases the expression of S. flexneri virulence genes icsA and ipaJ Intracellular S. flexneriicsA and ipaJ expression is dependent on the presence of formate, and ipaJ expression correlates with S. flexneri intracellular density during infection. Finally, consistent with elevated ipaJ, we show that formate alters S. flexneri-infected host interferon- and tumor necrosis factor (TNF)-stimulated gene expression. We propose that Shigella-derived formate is an intracellular signal that modulates virulence in response to bacterial metabolism.IMPORTANCEShigella is an intracellular pathogen that invades the human host cell cytosol and exploits intracellular nutrients for growth, enabling the bacterium to create its own metabolic niche. For Shigella to effectively invade and replicate within the host cytoplasm, it must sense and adapt to changing environmental conditions; however, the mechanisms and signals sensed by S. flexneri are largely unknown. We have found that the secreted Shigella metabolism by-product formate regulates Shigella intracellular virulence gene expression and its ability to spread among epithelial cells. We propose that Shigella senses formate accumulation in the host cytosol as a way to determine intracellular Shigella density and regulate secreted virulence factors accordingly, enabling spatiotemporal regulation of effectors important for dampening the host immune response.


Subject(s)
Formates/pharmacology , Gene Expression Regulation, Bacterial , Shigella flexneri/drug effects , Virulence Factors/genetics , Acetyltransferases/genetics , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Carbon/metabolism , Cell Line , Cytosol/microbiology , DNA-Binding Proteins/genetics , Humans , Shigella flexneri/genetics , Shigella flexneri/pathogenicity , Transcription Factors/genetics
2.
J Bacteriol ; 198(20): 2810-7, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27481926

ABSTRACT

UNLABELLED: Manganese plays an important role in the cellular physiology and metabolism of bacterial species, including the human pathogen Vibrio cholerae The intracellular level of manganese ions is controlled through coordinated regulation of the import and export of this element. We have identified a putative manganese exporter (VC0022), named mneA (manganese exporter A), which is highly conserved among Vibrio spp. An mneA mutant exhibited sensitivity to manganese but not to other cations. Under high-manganese conditions, the mneA mutant showed an almost 50-fold increase in intracellular manganese levels and reduced intracellular iron relative to those of its wild-type parent, suggesting that the mutant's manganese sensitivity is due to the accumulation of toxic levels of manganese and reduced iron. Expression of mneA suppressed the manganese-sensitive phenotype of an Escherichia coli strain carrying a mutation in the nonhomologous manganese export gene, mntP, further supporting a manganese export function for V. cholerae MneA. The level of mneA mRNA was induced approximately 2.5-fold after addition of manganese to the medium, indicating regulation of this gene by manganese. This study offers the first insights into understanding manganese homeostasis in this important pathogen. IMPORTANCE: Bacterial cells control intracellular metal concentrations by coordinating acquisition in metal-limited environments with export in metal-excess environments. We identified a putative manganese export protein, MneA, in Vibrio cholerae An mneA mutant was sensitive to manganese, and this effect was specific to manganese. The mneA mutant accumulated high levels of intracellular manganese with a concomitant decrease in intracellular iron levels when grown in manganese-supplemented medium. Expression of mneA in trans suppressed the manganese sensitivity of an E. coli mntP mutant. This study is the first to investigate manganese export in V. cholerae.


Subject(s)
Bacterial Proteins/metabolism , Manganese/metabolism , Membrane Transport Proteins/metabolism , Vibrio cholerae/metabolism , Bacterial Proteins/genetics , Biological Transport , Gene Expression Regulation, Bacterial , Iron/metabolism , Membrane Transport Proteins/genetics , Mutation , Vibrio cholerae/genetics
3.
Infect Immun ; 84(2): 511-23, 2016 02.
Article in English | MEDLINE | ID: mdl-26644383

ABSTRACT

Vibrio cholerae, the causative agent of the severe diarrheal disease cholera, thrives in both marine environments and the human host. To do so, it must encode the tools necessary to acquire essential nutrients, including iron, under these vastly different conditions. A number of V. cholerae iron acquisition systems have been identified; however, the precise role of each system is not fully understood. To test the roles of individual systems, we generated a series of mutants in which only one of the four systems that support iron acquisition on unsupplemented LB agar, Feo, Fbp, Vct, and Vib, remains functional. Analysis of these mutants under different growth conditions showed that these systems are not redundant. The strain carrying only the ferrous iron transporter Feo grew well at acidic, but not alkaline, pH, whereas the ferric iron transporter Fbp promoted better growth at alkaline than at acidic pH. A strain defective in all four systems (null mutant) had a severe growth defect under aerobic conditions but accumulated iron and grew as well as the wild type in the absence of oxygen, suggesting the presence of an additional, unidentified iron transporter in V. cholerae. In support of this, the null mutant was only moderately attenuated in an infant mouse model of infection. While the null mutant used heme as an iron source in vitro, we demonstrate that heme is not available to V. cholerae in the infant mouse intestine.


Subject(s)
Cholera/microbiology , Heme/metabolism , Iron/metabolism , Vibrio cholerae/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cholera/metabolism , Disease Models, Animal , Humans , Hydrogen-Ion Concentration , Intestinal Mucosa/metabolism , Intestines/microbiology , Mice , Mutation , Oxygen/metabolism , Peptide Synthases/genetics , Peptide Synthases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Vibrio cholerae/genetics , Vibrio cholerae/growth & development
4.
Infect Immun ; 82(2): 660-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478081

ABSTRACT

The Vps/VacJ ABC transporter system is proposed to function in maintaining the lipid asymmetry of the outer membrane. Mutations in vps or vacJ in Shigella flexneri resulted in increased sensitivity to lysis by the detergent sodium dodecyl sulfate (SDS), and the vpsC mutant showed minor differences in its phospholipid profile compared to the wild type. vpsC mutants were unable to form plaques in cultured epithelial cells, but this was not due to a failure to invade, to replicate intracellularly, or to polymerize actin via IcsA for movement within epithelial cells. The addition of the outer membrane phospholipase gene pldA on a multicopy plasmid in a vpsC or vacJ mutant restored its resistance to SDS, suggesting a restoration of lipid asymmetry to the outer membrane. However, the pldA plasmid did not restore the mutant's ability to form plaques in tissue culture cells. Increased PldA levels also failed to restore the mutant's phospholipid profile to that of the wild type. We propose a dual function of the Vps/VacJ ABC transporter system in S. flexneri in both the maintenance of lipid asymmetry in the outer membrane and the intercellular spread of the bacteria between adjacent epithelial cells.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Shigella flexneri/physiology , ATP-Binding Cassette Transporters/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Cell Line , Epithelial Cells/microbiology , Humans , Membrane Proteins/genetics , Mutation , Phospholipids/analysis , Shigella flexneri/chemistry
5.
BMC Microbiol ; 12: 226, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-23035718

ABSTRACT

BACKGROUND: Glutamyl queuosine-tRNA(Asp) synthetase (GluQ-RS) is a paralog of the catalytic domain of glutamyl-tRNA synthetase and catalyzes the formation of glutamyl-queuosine on the wobble position of tRNA(Asp). Here we analyze the transcription of its gene in Shigella flexneri, where it is found downstream of dksA, which encodes a transcriptional regulator involved in stress responses. RESULTS: The genomic organization, dksA-gluQ-rs, is conserved in more than 40 bacterial species. RT-PCR assays show co-transcription of both genes without a significant change in transcript levels during growth of S. flexneri. However, mRNA levels of the intergenic region changed during growth, increasing at stationary phase, indicating an additional level of control over the expression of gluQ-rs gene. Transcriptional fusions with lacZ as a reporter gene only produced ß-galactosidase activity when the constructs included the dksA promoter, indicating that gluQ-rs do not have a separate promoter. Using bioinformatics, we identified a putative transcriptional terminator between dksA and gluQ-rs. Deletion or alteration of the predicted terminator resulted in increased expression of the lacZ reporter compared with cells containing the wild type terminator sequence. Analysis of the phenotype of a gluQ-rs mutant suggested that it may play a role in some stress responses, since growth of the mutant was impaired in the presence of osmolytes. CONCLUSIONS: The results presented here, show that the expression of gluQ-rs depends on the dksA promoter, and strongly suggest the presence and the functionality of a transcriptional terminator regulating its expression. Also, the results indicate a link between glutamyl-queuosine synthesis and stress response in Shigella flexneri.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Shigella flexneri/genetics , Shigella flexneri/metabolism , Transcription Factors/metabolism , Amino Acyl-tRNA Synthetases/genetics , Artificial Gene Fusion , Bacterial Proteins/genetics , Computational Biology , DNA Mutational Analysis , Gene Order , Genes, Reporter , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Synteny , Transcription Factors/genetics , Transcription Termination, Genetic , beta-Galactosidase/analysis , beta-Galactosidase/genetics
6.
mBio ; 2(3): e00069-11, 2011.
Article in English | MEDLINE | ID: mdl-21628502

ABSTRACT

UNLABELLED: Natural habitats vary in available nutrients and room for bacteria to grow, but successful colonization can lead to overcrowding and stress. Here we show that competing sibling colonies of Paenibacillus dendritiformis bacteria survive overcrowding by switching between two distinct vegetative phenotypes, motile rods and immotile cocci. Growing colonies of the rod-shaped bacteria produce a toxic protein, Slf, which kills cells of encroaching sibling colonies. However, sublethal concentrations of Slf induce some of the rods to switch to Slf-resistant cocci, which have distinct metabolic and resistance profiles, including resistance to cell wall antibiotics. Unlike dormant spores of P. dendritiformis, the cocci replicate. If cocci encounter conditions that favor rods, they secrete a signaling molecule that induces a switch to rods. Thus, in contrast to persister cells, P. dendritiformis bacteria adapt to changing environmental conditions by inducible and reversible phenotypic switching. IMPORTANCE: In favorable environments, species may face space and nutrient limits due to overcrowding. Bacteria provide an excellent model for analyzing principles underlying overcrowding and regulation of density in nature, since their population dynamics can be easily and accurately assessed under controlled conditions. We describe a newly discovered mechanism for survival of a bacterial population during overcrowding. When competing with sibling colonies, Paenibacillus dendritiformis produces a lethal protein (Slf) that kills cells at the interface of encroaching colonies. Slf also induces a small proportion of the cells to switch from motile, rod-shaped cells to nonmotile, Slf-resistant, vegetative cocci. When crowding is reduced and nutrients are no longer limiting, the bacteria produce a signal that induces cocci to switch back to motile rods, allowing the population to spread. Genes encoding components of this phenotypic switching pathway are widespread among bacterial species, suggesting that this survival mechanism is not unique to P. dendritiformis.


Subject(s)
Locomotion , Microbial Viability , Paenibacillus/cytology , Paenibacillus/physiology , Stress, Physiological , Anti-Bacterial Agents/metabolism , Bacterial Toxins/biosynthesis , Bacterial Toxins/toxicity , Gene Expression Regulation, Bacterial , Paenibacillus/drug effects , Paenibacillus/growth & development , Phenotype
7.
Infect Immun ; 77(5): 1992-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19289511

ABSTRACT

The sit-encoded iron transport system is present within pathogenicity islands in all Shigella spp. and some pathogenic Escherichia coli strains. The islands contain numerous insertion elements and sequences with homology to bacteriophage genes. The Shigella flexneri sit genes can be lost as a result of deletion within the island. The formation of deletions was dependent upon RecA and occurred at relatively high frequency. This suggests that the sit region is inherently unstable, yet sit genes are maintained in all of the clinical isolates tested. Characterization of the sitABCD genes in S. flexneri indicates that they encode a ferrous iron transport system, although the genes are induced aerobically. The sit genes provide a competitive advantage to S. flexneri growing within epithelial cells, and a sitA mutant is outcompeted by the wild type in cultured epithelial cells. The Sit system is also required for virulence in a mouse lung model. The sitA mutant was able to infect the mice and induce a protective immune response but was avirulent compared to its wild-type parent strain.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Iron/metabolism , Shigella flexneri/genetics , Shigella flexneri/pathogenicity , Virulence Factors/genetics , Virulence Factors/metabolism , Animals , Cell Line , Dysentery, Bacillary/microbiology , Epithelial Cells/microbiology , Female , Gene Deletion , Humans , Lung/microbiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred BALB C , Virulence
8.
J Bacteriol ; 190(17): 5953-62, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18586940

ABSTRACT

Vibrio cholerae uses a variety of strategies for obtaining iron in its diverse environments. In this study we report the identification of a novel iron utilization protein in V. cholerae, VciB. The vciB gene and its linked gene, vciA, were isolated in a screen for V. cholerae genes that permitted growth of an Escherichia coli siderophore mutant in low-iron medium. The vciAB operon encodes a predicted TonB-dependent outer membrane receptor, VciA, and a putative inner membrane protein, VciB. VciB, but not VciA, was required for growth stimulation of E. coli and Shigella flexneri strains in low-iron medium. Consistent with these findings, TonB was not needed for VciB-mediated growth. No growth enhancement was seen when vciB was expressed in an E. coli or S. flexneri strain defective for the ferrous iron transporter Feo. Supplying the E. coli feo mutant with a plasmid encoding either E. coli or V. cholerae Feo, or the S. flexneri ferrous iron transport system Sit, restored VciB-mediated growth; however, no stimulation was seen when either of the ferric uptake systems V. cholerae Fbp and Haemophilus influenzae Hit was expressed. These data indicate that VciB functions by promoting iron uptake via a ferrous, but not ferric, iron transport system. VciB-dependent iron accumulation via Feo was demonstrated directly in iron transport assays using radiolabeled iron. A V. cholerae vciB mutant did not exhibit any growth defects in either in vitro or in vivo assays, possibly due to the presence of other systems with overlapping functions in this pathogen.


Subject(s)
Bacterial Proteins/metabolism , Iron/metabolism , Membrane Transport Proteins/metabolism , Vibrio cholerae/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/physiology , Iron/pharmacokinetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , Operon/genetics , Polymerase Chain Reaction , Sequence Analysis, DNA , Vibrio cholerae/genetics , Vibrio cholerae/growth & development
9.
J Bacteriol ; 189(15): 5566-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17526712

ABSTRACT

A Shigella flexneri degP mutant, which was defective for plaque formation in Henle cell monolayers, had a reduced amount of IcsA detectable on the bacterial surface with antibody. However, the mutant secreted IcsA to the outer membrane at wild-type levels. This suggests that IcsA adopts an altered conformation in the outer membrane of the degP mutant with reduced exposure on the cell surface. IcsA is, therefore, unlikely to be accessible to actin-nucleating proteins within the eukaryotic cell cytoplasm, which is required for bacterial movement within the host cell and cell-to-cell spread. The degP mutant was somewhat more sensitive to detergents, antibiotics, and the antimicrobial peptide magainin, indicating that the degP phenotype was not limited to IcsA surface presentation. The plaque defect of the degP mutant, which is independent of DegP protease activity, was suppressed by overexpression of the periplasmic chaperone Skp but not by SurA. S. flexneri skp and surA mutants failed to form plaques in Henle cell monolayers and were defective in cell surface presentation and polar localization of IcsA. Therefore, the three periplasmic folding factors DegP, Skp, and SurA were all required for IcsA localization and plaque formation by S. flexneri.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Peptidylprolyl Isomerase/metabolism , Periplasmic Proteins/metabolism , Serine Endopeptidases/metabolism , Shigella flexneri/metabolism , Transcription Factors/metabolism , Bacterial Outer Membrane Proteins/analysis , Bacterial Proteins/genetics , Cell Line , Cell Membrane/chemistry , Heat-Shock Proteins/genetics , Humans , Lipopolysaccharides/metabolism , Molecular Chaperones/genetics , Mutation , Peptidylprolyl Isomerase/genetics , Periplasmic Proteins/genetics , Phenotype , Serine Endopeptidases/genetics , Shigella flexneri/chemistry , Shigella flexneri/genetics , Shigella flexneri/pathogenicity
10.
Infect Immun ; 73(12): 8167-78, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16299312

ABSTRACT

Regulation of iron uptake and utilization is critical for bacterial growth and for prevention of iron toxicity. In many bacterial species, this regulation depends on the iron-responsive master regulator Fur. In this study we report the effects of iron and Fur on gene expression in Vibrio cholerae. We show that Fur has both positive and negative regulatory functions, and we demonstrate Fur-independent regulation of gene expression by iron. Nearly all of the known iron acquisition genes were repressed by Fur under iron-replete conditions. In addition, genes for two newly identified iron transport systems, Feo and Fbp, were found to be negatively regulated by iron and Fur. Other genes identified in this study as being induced in low iron and in the fur mutant include those encoding superoxide dismutase (sodA), fumarate dehydratase (fumC), bacterioferritin (bfr), bacterioferritin-associated ferredoxin (bfd), and multiple genes of unknown function. Several genes encoding iron-containing proteins were repressed in low iron and in the fur mutant, possibly reflecting the need to reserve available iron for the most critical functions. Also repressed in the fur mutant, but independently of iron, were genes located in the V. cholerae pathogenicity island, encoding the toxin-coregulated pilus (TCP), and genes within the V. cholerae mega-integron. The fur mutant exhibited very weak autoagglutination, indicating a possible defect in expression or assembly of the TCP, a major virulence factor of V. cholerae. Consistent with this observation, the fur mutant competed poorly with its wild-type parental strain for colonization of the infant mouse gut.


Subject(s)
Bacterial Proteins/physiology , Gene Expression Regulation, Bacterial , Iron/metabolism , Repressor Proteins/physiology , Vibrio cholerae/genetics , Vibrio cholerae/pathogenicity , Bacterial Proteins/genetics , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Genes, Bacterial , Iron/pharmacology , Mutation , Oligonucleotide Array Sequence Analysis , Repressor Proteins/genetics , Up-Regulation , Vibrio cholerae/metabolism , Virulence/genetics , Virulence/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...