Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-28785546

ABSTRACT

The FeoABC ferrous transporter is a wide-spread bacterial system. While the feoABC locus is regulated by a number of factors in the bacteria studied, we have previously found that regulation of feoABC in Yersinia pestis appears to be unique. None of the non-iron responsive transcriptional regulators that control expression of feoABC in other bacteria do so in Y. pestis. Another unique factor is the iron and Fur regulation of the Y. pestis feoABC locus occurs during microaerobic but not aerobic growth. Here we show that this unique iron-regulation is not due to a unique aspect of the Y. pestis Fur protein but to DNA sequences that regulate transcription. We have used truncations, alterations, and deletions of the feoA::lacZ reporter to assess the mechanism behind the failure of iron to repress transcription under aerobic conditions. These studies plus EMSAs and DNA sequence analysis have led to our proposal that the feoABC locus has two promoters: an upstream P1 promoter whose expression is relatively iron-independent but repressed under microaerobic conditions and the known downstream Fur-regulated P2 promoter. In addition, we have identified two regions that bind Y. pestis protein(s), although we have not identified these protein(s) or their function. Finally we used iron uptake assays to demonstrate that both FeoABC and YfeABCD transport ferrous iron in an energy-dependent manner and also use ferric iron as a substrate for uptake.


Subject(s)
Gene Expression Regulation, Bacterial , Iron/metabolism , Membrane Transport Proteins/metabolism , Promoter Regions, Genetic , Trace Elements/metabolism , Yersinia pestis/genetics , Yersinia pestis/metabolism , Artificial Gene Fusion , Biological Transport, Active , DNA Mutational Analysis , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Genes, Reporter , Genetic Loci , Membrane Transport Proteins/genetics , Sequence Analysis, DNA , Yersinia pestis/growth & development , beta-Galactosidase/analysis , beta-Galactosidase/genetics
2.
Metallomics ; 9(6): 757-772, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28540946

ABSTRACT

A number of bacterial pathogens require the ZnuABC Zinc (Zn2+) transporter and/or a second Zn2+ transport system to overcome Zn2+ sequestration by mammalian hosts. Previously we have shown that in addition to ZnuABC, Yersinia pestis possesses a second Zn2+ transporter that involves components of the yersiniabactin (Ybt), siderophore-dependent iron transport system. Synthesis of the Ybt siderophore and YbtX, a member of the major facilitator superfamily, are both critical components of the second Zn2+ transport system. Here we demonstrate that a ybtX znu double mutant is essentially avirulent in mouse models of bubonic and pneumonic plague while a ybtX mutant retains high virulence in both plague models. While sequestration of host Zn is a key nutritional immunity factor, excess Zn appears to have a significant antimicrobial role in controlling intracellular bacterial survival. Here, we demonstrate that ZntA, a Zn2+ exporter, plays a role in resistance to Zn toxicity in vitro, but that a zntA zur double mutant retains high virulence in both pneumonic and bubonic plague models and survival in macrophages. We also confirm that Ybt does not directly bind Zn2+in vitro under the conditions tested. However, we detect a significant increase in Zn2+-binding ability of filtered supernatants from a Ybt+ strain compared to those from a strain unable to produce the siderophore, supporting our previously published data that Ybt biosynthetic genes are involved in the production of a secreted Zn-binding molecule (zincophore). Our data suggest that Ybt or a modified Ybt participate in or promote Zn-binding activity in culture supernatants and is involved in Zn acquisition in Y. pestis.


Subject(s)
Bacterial Proteins/metabolism , Plague/pathology , Virulence Factors/metabolism , Yersinia pestis/pathogenicity , Zinc/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bacterial Proteins/genetics , Cells, Cultured , Female , Gene Expression Regulation, Bacterial , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mutation , Plague/microbiology , Virulence , Virulence Factors/genetics
3.
Metallomics ; 7(6): 965-78, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25891079

ABSTRACT

Yersinia pestis, the causative agent of bubonic, septicemic and pneumonic plague, encodes a multitude of Fe transport systems. Some of these are defective due to frameshift or IS element insertions, while others are functional in vitro but have no established role in causing infections. Indeed only 3 Fe transporters (Ybt, Yfe and Feo) have been shown to be important in at least one form of plague. The yersiniabactin (Ybt) system is essential in the early dermal/lymphatic stages of bubonic plague, irrelevant in the septicemic stage, and critical in pneumonic plague. Two Mn transporters have been characterized (Yfe and MntH). These two systems play a role in bubonic plague but the double yfe mntH mutant is fully virulent in a mouse model of pneumonic plague. The same in vivo phenotype occurs with a mutant lacking two (Yfe and Feo) of four ferrous transporters. A role for the Ybt siderophore in Zn acquisition has been revealed. Ybt-dependent Zn acquisition uses a transport system completely independent of the Fe-Ybt uptake system. Together Ybt components and ZnuABC play a critical role in Zn acquisition in vivo. Single mutants in either system retain high virulence in a mouse model of septicemic plague while the double mutant is completely avirulent.


Subject(s)
Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Metals/metabolism , Transition Elements/metabolism , Yersinia pestis/physiology , Animals , Humans , Plague/metabolism , Plague/microbiology
4.
Mol Microbiol ; 93(4): 759-75, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24979062

ABSTRACT

Bacterial pathogens must overcome host sequestration of zinc (Zn(2+) ), an essential micronutrient, during the infectious disease process. While the mechanisms to acquire chelated Zn(2+) by bacteria are largely undefined, many pathogens rely upon the ZnuABC family of ABC transporters. Here we show that in Yersinia pestis, irp2, a gene encoding the synthetase (HMWP2) for the siderophore yersiniabactin (Ybt) is required for growth under Zn(2+) -deficient conditions in a strain lacking ZnuABC. Moreover, growth stimulation with exogenous, purified apo-Ybt provides evidence that Ybt may serve as a zincophore for Zn(2+) acquisition. Studies with the Zn(2+) -dependent transcriptional reporter znuA::lacZ indicate that the ability to synthesize Ybt affects the levels of intracellular Zn(2+) . However, the outer membrane receptor Psn and TonB as well as the inner membrane (IM) ABC transporter YbtPQ, which are required for Fe(3+) acquisition by Ybt, are not needed for Ybt-dependent Zn(2+) uptake. In contrast, the predicted IM protein YbtX, a member of the Major Facilitator Superfamily, was essential for Ybt-dependent Zn(2+) uptake. Finally, we show that the ZnuABC system and the Ybt synthetase HMWP2, presumably by Ybt synthesis, both contribute to the development of a lethal infection in a septicaemic plague mouse model.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Phenols/metabolism , Plague/microbiology , Thiazoles/metabolism , Virulence Factors/metabolism , Yersinia pestis/metabolism , Zinc/metabolism , Animals , Disease Models, Animal , Mice , Plague/pathology , Sepsis/microbiology , Sepsis/pathology , Virulence
5.
Infect Immun ; 80(11): 3880-91, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22927049

ABSTRACT

The Yfe/Sit and Feo transport systems are important for the growth of a variety of bacteria. In Yersinia pestis, single mutations in either yfe or feo result in reduced growth under static (limited aeration), iron-chelated conditions, while a yfe feo double mutant has a more severe growth defect. These growth defects were not observed when bacteria were grown under aerobic conditions or in strains capable of producing the siderophore yersiniabactin (Ybt) and the putative ferrous transporter FetMP. Both fetP and a downstream locus (flp for fet linked phenotype) were required for growth of a yfe feo ybt mutant under static, iron-limiting conditions. An feoB mutation alone had no effect on the virulence of Y. pestis in either bubonic or pneumonic plague models. An feo yfe double mutant was still fully virulent in a pneumonic plague model but had an ∼90-fold increase in the 50% lethal dose (LD(50)) relative to the Yfe(+) Feo(+) parent strain in a bubonic plague model. Thus, Yfe and Feo, in addition to Ybt, play an important role in the progression of bubonic plague. Finally, we examined the factors affecting the expression of the feo operon in Y. pestis. Under static growth conditions, the Y. pestis feo::lacZ fusion was repressed by iron in a Fur-dependent manner but not in cells grown aerobically. Mutations in feoC, fnr, arcA, oxyR, or rstAB had no significant effect on transcription of the Y. pestis feo promoter. Thus, the factor(s) that prevents repression by Fur under aerobic growth conditions remains to be identified.


Subject(s)
Bacterial Proteins/genetics , Membrane Transport Proteins/genetics , Plague/genetics , Virulence/genetics , Yersinia pestis/genetics , Animals , Bacterial Proteins/metabolism , Biological Transport , Gene Expression Regulation, Bacterial , Iron/metabolism , Iron Deficiencies , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Plague/microbiology , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity
7.
Microbiology (Reading) ; 158(Pt 3): 804-815, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22222497

ABSTRACT

Yersinia pestis has a flea-mammal-flea transmission cycle, and is a zoonotic pathogen that causes the systemic diseases bubonic and septicaemic plague in rodents and humans, as well as pneumonic plague in humans and non-human primates. Bubonic and pneumonic plague are quite different diseases that result from different routes of infection. Manganese (Mn) acquisition is critical for the growth and pathogenesis of a number of bacteria. The Yfe/Sit and/or MntH systems are the two prominent Mn transporters in Gram-negative bacteria. Previously we showed that the Y. pestis Yfe system transports Fe and Mn. Here we demonstrate that a mutation in yfe or mntH did not significantly affect in vitro aerobic growth under Mn-deficient conditions. A yfe mntH double mutant did exhibit a moderate growth defect which was alleviated by supplementation with Mn. No short-term energy-dependent uptake of (54)Mn was observed in this double mutant. Like the yfeA promoter, the mntH promoter was repressed by both Mn and Fe via Fur. Sequences upstream of the Fur binding sequence in the yfeA promoter converted an iron-repressible promoter to one that is also repressed by Mn and Fe. To our knowledge, this is the first report identifying cis promoter elements needed to alter cation specificities involved in transcriptional repression. Finally, the Y. pestis yfe mntH double mutant had an ~133-fold loss of virulence in a mouse model of bubonic plague but no virulence loss in the pneumonic plague model. This suggests that Mn availability, bacterial Mn requirements or Mn transporters used by Y. pestis are different in the lungs (pneumonic plague) compared with systemic disease.


Subject(s)
Bacterial Proteins/metabolism , Cation Transport Proteins/metabolism , Gene Expression Regulation, Bacterial , Membrane Transport Proteins/metabolism , Repressor Proteins/metabolism , Virulence Factors/metabolism , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , Animals , Artificial Gene Fusion , Bacterial Proteins/genetics , Cation Transport Proteins/genetics , Disease Models, Animal , Gene Deletion , Genes, Reporter , Humans , Manganese/metabolism , Membrane Transport Proteins/genetics , Mice , Plague/microbiology , Plague/pathology , Promoter Regions, Genetic , Survival Analysis , Virulence , Virulence Factors/genetics , Yersinia pestis/genetics , Yersinia pestis/growth & development , beta-Galactosidase/analysis , beta-Galactosidase/genetics
8.
Microbes Infect ; 13(10): 808-17, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21609780

ABSTRACT

Yersiniabactin (Ybt) is a siderophore-dependent iron uptake system encoded on a pathogenicity island that is widespread among pathogenic bacteria including the Yersiniae. While biosynthesis of the siderophore has been elucidated, the secretion mechanism and a few components of the uptake/utilization pathway are unidentified. ybt genes are transcriptionally repressed by Fur but activated by YbtA, likely in combination with the siderophore itself. The Ybt system is essential for the ability of Yersinia pestis to cause bubonic plague and important in pneumonic plague as well. However, the ability to cause fatal septicemic plague is independent of Ybt.


Subject(s)
Iron/metabolism , Phenols/metabolism , Plague/microbiology , Plague/pathology , Thiazoles/metabolism , Virulence Factors/metabolism , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , Gene Expression Regulation, Bacterial , Humans , Virulence Factors/genetics , Yersinia pestis/genetics
9.
Mol Microbiol ; 79(2): 533-51, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21219468

ABSTRACT

Cyclic di-GMP (c-di-GMP) is a signalling molecule that governs the transition between planktonic and biofilm states. Previously, we showed that the diguanylate cyclase HmsT and the putative c-di-GMP phosphodiesterase HmsP inversely regulate biofilm formation through control of HmsHFRS-dependent poly-ß-1,6-N-acetylglucosamine synthesis. Here, we systematically examine the functionality of the genes encoding putative c-di-GMP metabolic enzymes in Yersinia pestis. We determine that, in addition to hmsT and hmsP, only the gene y3730 encodes a functional enzyme capable of synthesizing c-di-GMP. The seven remaining genes are pseudogenes or encode proteins that do not function catalytically or are not expressed. Furthermore, we show that HmsP has c-di-GMP-specific phosphodiesterase activity. We report that a mutant incapable of c-di-GMP synthesis is unaffected in virulence in plague mouse models. Conversely, an hmsP mutant, unable to degrade c-di-GMP, is defective in virulence by a subcutaneous route of infection due to poly-ß-1,6-N-acetylglucosamine overproduction. This suggests that c-di-GMP signalling is not only dispensable but deleterious for Y. pestis virulence. Our results show that a key event in the evolution of Y. pestis from the ancestral Yersinia pseudotuberculosis was a significant reduction in the complexity of its c-di-GMP signalling network likely resulting from the different disease cycles of these human pathogens.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Bacterial Proteins/metabolism , Biofilms/growth & development , Cyclic GMP/analogs & derivatives , Signal Transduction , Virulence Factors/metabolism , Yersinia pestis/pathogenicity , Animals , Cyclic GMP/metabolism , Disease Models, Animal , Humans , Mice , Plague/microbiology , Plague/pathology , Virulence , Yersinia pestis/enzymology , Yersinia pestis/metabolism
10.
Vaccine ; 29(2): 274-82, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-20979987

ABSTRACT

We evaluated the ability of Yersinia pestis antigens HmuR, Psn and modified forms of LcrV delivered by live attenuated Salmonella strains to stimulate a protective immune response against subcutaneous or intranasal challenge with Y. pestis CO92. LcrV196 is a previously described truncated protein that includes aa 131-326 of LcrV and LcrV5214 has been modified to replace five key amino acids required for interaction with the TLR2 receptor. Psn is the outer membrane receptor for the siderophore, yersiniabactin, and the bacteriocin, pesticin. Mice immunized with Salmonella synthesizing Psn, LcrV196 or LcrV5214 developed serum IgG responses to the respective Yersinia antigen and were protected against pneumonic challenge with Y. pestis. Immunization with Salmonella synthesizing Psn or LcrV196 was sufficient to afford nearly full protection against bubonic challenge, while immunization with the strain synthesizing LcrV5214 was not protective. Immunization with Salmonella synthesizing HmuR, an outer membrane protein involved in heme acquisition in Y. pestis, was poorly immunogenic and did not elicit a protective response against either challenge route. These findings indicate that both Psn and LcrV196 delivered by Salmonella provide protection against both bubonic and pneumonic plague.


Subject(s)
Bacterial Proteins/immunology , Plague Vaccine/immunology , Plague/prevention & control , Yersinia pestis/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Disease Models, Animal , Female , Genetic Vectors , Mice , Mice, Inbred BALB C , Plague/immunology , Plague Vaccine/administration & dosage , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Salmonella Vaccines/genetics , Survival Analysis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Yersinia pestis/genetics
11.
Infect Immun ; 78(12): 5163-77, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20855510

ABSTRACT

Little is known about Zn homeostasis in Yersinia pestis, the plague bacillus. The Znu ABC transporter is essential for zinc (Zn) uptake and virulence in a number of bacterial pathogens. Bioinformatics analysis identified ZnuABC as the only apparent high-affinity Zn uptake system in Y. pestis. Mutation of znuACB caused a growth defect in Chelex-100-treated PMH2 growth medium, which was alleviated by supplementation with submicromolar concentrations of Zn. Use of transcriptional reporters confirmed that Zur mediated Zn-dependent repression and that it can repress gene expression in response to Zn even in the absence of Znu. Virulence testing in mouse models of bubonic and pneumonic plague found only a modest increase in survival in low-dose infections by the znuACB mutant. Previous studies of cluster 9 (C9) transporters suggested that Yfe, a well-characterized C9 importer for manganese (Mn) and iron in Y. pestis, might function as a second, high-affinity Zn uptake system. Isothermal titration calorimetry revealed that YfeA, the solute-binding protein component of Yfe, binds Mn and Zn with comparably high affinities (dissociation constants of 17.8 ± 4.4 nM and 6.6 ± 1.2 nM, respectively), although the complete Yfe transporter could not compensate for the loss of Znu in in vitro growth studies. Unexpectedly, overexpression of Yfe interfered with the znu mutant's ability to grow in low concentrations of Zn, while excess Zn interfered with the ability of Yfe to import iron at low concentrations; these results suggest that YfeA can bind Zn in the bacterial cell but that Yfe is incompetent for transport of the metal. In addition to Yfe, we have now eliminated MntH, FetMP, Efe, Feo, a substrate-binding protein, and a putative nickel transporter as the unidentified, secondary Zn transporter in Y. pestis. Unlike other bacterial pathogens, Y. pestis does not require Znu for high-level infectivity and virulence; instead, it appears to possess a novel class of transporter, which can satisfy the bacterium's Zn requirements under in vivo metal-limiting conditions. Our studies also underscore the need for bacterial cells to balance binding and transporter specificities within the periplasm in order to maintain transition metal homeostasis.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Plague/microbiology , Yersinia pestis/pathogenicity , Zinc/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Cloning, Molecular , Gene Expression Regulation, Bacterial/physiology , Mice , Middle East , Mutation , Virulence/genetics , Virulence/physiology , Yersinia pestis/genetics , Yersinia pestis/growth & development , Yersinia pestis/physiology , Zinc/physiology
12.
Environ Microbiol ; 12(7): 2034-47, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20406298

ABSTRACT

We previously showed that mutations in the genes encoding the two main biosynthetic enzymes responsible for polyamine production, arginine decarboxylase (SpeA) and ornithine decarboxylase (SpeC) cause a loss of biofilm formation in Yersinia pestis. In Y. pestis the development of a biofilm is dependent on 6 Hms (hemin storage) proteins (HmsH, F, R, S, T and P) grouped into 3 operons; hmsHFRS, hmsT and hmsP. In this article we show that polyamines are necessary to maintain the levels of key Hms proteins. In the absence of polyamines there is an approximately 93%, approximately 43% and approximately 90% reduction in protein levels of HmsR, HmsS and HmsT respectively. Overexpression of hmsR and hmsT from plasmids alone can restore biofilm formation to a SpeA(-)SpeC(-) mutant. Addition of exogenous putrescine also restores normal levels of HmsR, HmsS, HmsT and biofilm production. Analyses using transcriptional reporters and quantitative RT-PCR indicate that the initiation of transcription and mRNA stability are not reduced by polyamine deficiency. Instead, translational reporters indicate that polyamines function at least in part by modulating the translation of HmsR and HmsT. Although construction of a consensus Shine-Dalgarno sequence upstream of hmsT modestly reduced the stimulation of translation by putrescine, additional mechanisms likely contribute to the polyamine-dependent expression of HmsT. Finally, we have shown that polyamines play a role in bubonic plague.


Subject(s)
Bacterial Proteins/biosynthesis , Biofilms/growth & development , Gene Expression Regulation, Bacterial/drug effects , Polyamines/metabolism , Yersinia pestis/physiology , Animals , Disease Models, Animal , Gene Expression Profiling , Genes, Reporter , Mice , Plague/microbiology , RNA Stability , Reverse Transcriptase Polymerase Chain Reaction , Virulence , Yersinia pestis/growth & development , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
13.
Microbiology (Reading) ; 156(Pt 7): 2226-2238, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20413552

ABSTRACT

Synthesis of the siderophore yersiniabactin (Ybt) proceeds by a mixed nonribosomal peptide synthetase/polyketide synthase mechanism. Transcription of ybt genes encoding biosynthetic and transport functions is repressed under excess iron conditions by Fur, but is also activated by Ybt via the transcriptional regulator YbtA. While mutations in most biosynthetic genes and ybtA negate transcription activation from the regulated promoters, three biosynthetic mutations do not reduce this transcriptional activation. Here we show that two of these mutants, one lacking the putative type II thioesterase (TE) YbtT and the other with a mutation in the TE domain of HMWP1, produce reduced levels of authentic Ybt that are capable of signalling activity. Alanine substitutions in two residues of YbtT that are essential for catalytic activity in other type II TEs reduced the ability of Yersinia pestis to grow under iron-chelated conditions. The third mutant, which lacks the salicylate synthase YbtS, did not make authentic Ybt but did produce a signalling molecule. Finally, a Delta pgm strain of Y. pestis, which lacks essential Ybt biosynthetic genes, also produced a signalling molecule that can activate transcription of ybt genes. The non-Ybt signal molecules from these two mutants are likely separate compounds. While these compounds are not biologically relevant to normal Ybt regulation, a comparison of the structures of Ybt and other signalling molecules will help in determining the chemical structures recognized as a Ybt signal.


Subject(s)
Bacterial Proteins/genetics , Phenols/metabolism , Siderophores/biosynthesis , Thiazoles/metabolism , Transcriptional Activation , Yersinia pestis/genetics , Yersinia pestis/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial
14.
Infect Immun ; 78(5): 2045-52, 2010 May.
Article in English | MEDLINE | ID: mdl-20160020

ABSTRACT

Iron acquisition from the host is an important step in the pathogenic process. While Yersinia pestis has multiple iron transporters, the yersiniabactin (Ybt) siderophore-dependent system plays a major role in iron acquisition in vitro and in vivo. In this study, we determined that the Ybt system is required for the use of iron bound by transferrin and lactoferrin and examined the importance of the Ybt system for virulence in mouse models of bubonic and pneumonic plague. Y. pestis mutants unable to either transport Ybt or synthesize the siderophore were both essentially avirulent via subcutaneous injection (bubonic plague model). Surprisingly, via intranasal instillation (pneumonic plague model), we saw a difference in the virulence of Ybt biosynthetic and transport mutants. Ybt biosynthetic mutants displayed an approximately 24-fold-higher 50% lethal dose (LD(50)) than transport mutants. In contrast, under iron-restricted conditions in vitro, a Ybt transport mutant had a more severe growth defect than the Ybt biosynthetic mutant. Finally, a Delta pgm mutant had a greater loss of virulence than the Ybt biosynthetic mutant, indicating that the 102-kb pgm locus encodes a virulence factor, in addition to Ybt, that plays a role in the pathogenesis of pneumonic plague.


Subject(s)
Iron/metabolism , Phenols/metabolism , Plague/microbiology , Plague/pathology , Thiazoles/metabolism , Virulence Factors/metabolism , Yersinia pestis/pathogenicity , Animals , Female , Lethal Dose 50 , Mice , Survival Analysis , Virulence , Virulence Factors/deficiency
15.
Biometals ; 23(2): 275-94, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20049509

ABSTRACT

Although Yersinia pestis epidemic biovars and Yersinia pseudotuberculosis are recently diverged, highly related species, they cause different diseases via disparate transmission routes. Since iron transport systems are important for iron acquisition from hosts and for survival in the environment, we have analyzed potential iron transport systems encoded by epidemic and non-epidemic or endemic strains of Y. pestis as well as two virulent Y. pseudotuberculosis strains. Computational biology analysis of these genomes showed a high degree of identity/similarity among 16 proven or possible iron/heme transporters identified. Of these, 7 systems were essentially the same in all seven genomes analyzed. The remaining 9 loci had 2-6 genetic variations among these genomes. Two untested, potential siderophore-dependent systems appear intact in Y. pseudotuberculosis but are disrupted or absent in all the endemic Y. pestis strains as well as the epidemic strains from the antiqua and mediaevalis biovars. Only one of these two loci are obviously disrupted in Y. pestis CO92 (epidemic orientalis biovar). Experimental studies failed to identify a role for hemin uptake systems in the virulence of pneumonic plague and suggest that Y. pestis CO92 does not make a siderophore other than Ybt.


Subject(s)
Bacterial Proteins/metabolism , Iron/metabolism , Siderophores/metabolism , Yersinia pestis/metabolism , Yersinia pseudotuberculosis/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bacterial Proteins/genetics , Hemin/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Structure , Plague/microbiology , Siderophores/chemistry , Siderophores/genetics , Yersinia pestis/genetics , Yersinia pestis/pathogenicity , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/pathogenicity
16.
Microbiology (Reading) ; 156(Pt 5): 1424-1438, 2010 May.
Article in English | MEDLINE | ID: mdl-20093287

ABSTRACT

The Yersinia pestis Hms(+) phenotype is a manifestation of biofilm formation that causes adsorption of Congo red and haemin at 26 degrees C but not at 37 degrees C. This phenotype is required for blockage of the proventricular valve of the oriental rat flea and plays a role in transmission of bubonic plague from fleas to mammals. Genes responsible for this phenotype are located in three separate operons, hmsHFRS, hmsT and hmsP. HmsH and HmsF are outer membrane (OM) proteins, while the other four Hms proteins are located in the inner membrane. According to the Hidden Markov Method-based predictor, HmsH has a large N terminus in the periplasm, a beta-barrel structure with 16 beta-strands that traverse the OM, eight surface-exposed loops, and seven short turns connecting the beta-strands on the periplasmic side. Here, we demonstrate that HmsH is a heat-modifiable protein, a characteristic of other beta-barrel proteins, thereby supporting the bioinformatics analysis. Alanine scanning mutagenesis was used to identify conserved amino acids in the HmsH-like family that are critical for the function of HmsH in biofilm formation. Of 23 conserved amino acids mutated, four residues affected HmsH function and three likely caused protein instability. We used formaldehyde cross-linking to demonstrate that HmsH interacts with HmsF but not with HmsR, HmsS, HmsT or HmsP. Loss-of-function HmsH variants with single alanine substitutions retained their beta-structure and interaction with HmsF. Finally, using a polar hmsH : : mini-kan mutant, we demonstrated that biofilm development is not important for the pathogenesis of bubonic or pneumonic plague in mice.


Subject(s)
Bacterial Outer Membrane Proteins/physiology , Biofilms/growth & development , Yersinia pestis/physiology , Alanine , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Conserved Sequence , Hemin/genetics , Hemin/metabolism , Mice , Molecular Sequence Data , Mutagenesis , Plague/genetics , Plague/microbiology , Plague/transmission , Protein Conformation , Siphonaptera/microbiology , Yersinia pestis/genetics
17.
Vaccine ; 27(39): 5363-70, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19596407

ABSTRACT

The use of live recombinant attenuated Salmonella vaccines (RASV) synthesizing Yersinia proteins is a promising approach for controlling infection by Yersinia species. In this study, we constructed attenuated Salmonella strains which synthesize a truncated form of LcrV, LcrV196 and evaluated the immune response and protective efficacy elicited by these strains in mice against two other major species of Yersinia: Yersinia pseudotuberculosis and Yersinia enterocolitica. Surprisingly, we found that the RASV strain alone was sufficient to afford nearly full protection against challenge with Y. pseudotuberculosis, indicating the likelihood that Salmonella produces immunogenic cross-protective antigens. In contrast, lcrV196 expression was required for protection against challenge with Y. enterocolitica strain 8081, but was not sufficient to achieve significant protection against challenge with Y. enterocolitica strain WA, which expressed a divergent form of lcrV. Nevertheless, we are encouraged by these findings to continue pursuing our long-term goal of developing a single vaccine to protect against all three human pathogenic species of Yersinia.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Pore Forming Cytotoxic Proteins/immunology , Salmonella typhimurium/immunology , Yersinia Infections/prevention & control , Administration, Oral , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cross Reactions , DNA, Bacterial/genetics , Female , Mice , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Yersinia Infections/immunology , Yersinia enterocolitica/immunology , Yersinia pestis/immunology , Yersinia pseudotuberculosis/immunology
18.
Adv Exp Med Biol ; 603: 178-91, 2007.
Article in English | MEDLINE | ID: mdl-17966414

ABSTRACT

Gram-negative bacteria predominantly use two types of quorum sensing (QS) systems--LuxI-LuxR, responsible for synthesis of N-acylhomoserine lactones (AHL or AI-1 signal molecule), and LuxS, which makes furanones (AI-2 signal molecule). We showed that LuxS and two LuxI-LuxR (YtbIR and YpsIR) systems are functional in Y. pestis. Four different AHL molecules were detected in Y. pestis extracts using TLC bioassays. Our data suggest that YtbIR is responsible for the production of long chain AHLs. Confocal laser scanning microscopy showed that biofilm formation is decreased in an ytbIR ypsIR luxS mutant. Two-dimensional gel electrophoresis revealed altered levels of protein expression in a Y. pestis triple QS mutant at 26 degrees C and 37 degrees C.


Subject(s)
Quorum Sensing/genetics , Quorum Sensing/physiology , Yersinia pestis/genetics , Yersinia pestis/physiology , Acyl-Butyrolactones/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Base Sequence , Biofilms/growth & development , Carbon-Sulfur Lyases/genetics , Carbon-Sulfur Lyases/physiology , DNA Primers/genetics , DNA, Bacterial/genetics , Gene Expression , Genes, Bacterial , Genes, Reporter , Homoserine/analogs & derivatives , Homoserine/biosynthesis , Lac Operon , Lactones , Plasmids/genetics
19.
Adv Exp Med Biol ; 603: 387-99, 2007.
Article in English | MEDLINE | ID: mdl-17966435

ABSTRACT

The use of live recombinant Salmonella attenuated vaccine (RASV) encoding Yersinia proteins is a promising new approach for the vaccination against Yersinia pestis. We have tested the efficacy of 2 proteins, Psn and a portion of LcrV in protecting mice against virulent Yersinia pestis challenge. To remove the immunosuppressive properties of LcrV protein, the lcrV gene, without the TLR2 receptor sequence, was cloned into a beta-lactamase secretion vector. Immunizations were performed with RSAV expressing LcrV or Psn. Challenge with a virulent Y. pestis strain was performed 4 weeks after the last immunization. Our results show that the truncated LcrV protein delivered by RASV is sufficient to afford a full protective immune response in a mouse model of bubonic plague and the Psn protein afforded partial protection in a non-optimized system. This finding should facilitate the design and development of a new generation of vaccines against Y. pestis.


Subject(s)
Plague Vaccine/administration & dosage , Plague/prevention & control , Yersinia pestis/immunology , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Base Sequence , DNA, Bacterial/genetics , Female , Genes, Bacterial , Mice , Mice, Inbred BALB C , Plague/immunology , Plague Vaccine/genetics , Plague Vaccine/immunology , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Yersinia pestis/genetics
20.
Microbiology (Reading) ; 153(Pt 7): 2332-2341, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17600077

ABSTRACT

Yersinia pestis genomes contain genes homologous to the aerobactin receptor (iutA) and biosynthetic genes (iucABCD) as well as the ferric hydroxamate uptake system (fhuCDB) of Escherichia coli. However, iucA is disrupted by a frameshift mutation. An E. coli strain carrying the cloned Y. pestis aerobactin region was unable to produce aerobactin, but could use the siderophore as an iron source. Repair of the frameshift mutation in iucA did not allow aerobactin production in E. coli or Y. pestis. In contrast, a Y. pestis strain with a plasmid encoding the iucABCD-iutA genes from Shigella flexneri or pColV-K30 did produce and secrete the siderophore. In addition, Yersinia pseudotuberculosis PB1, which encodes the iucABCD-iutA locus without the Y. pestis-specific frameshift mutation, also failed to produce aerobactin. The Y. pestis fhuCDB operon, encoding an ABC transporter for a range of hydroxamate siderophores, was able to complement a strain of E. coli with a transposon insertion in fhuC, allowing utilization of aerobactin and ferrichrome. Y. pestis KIM6, a strain deficient in the production of the siderophore yersiniabactin, was able to use both the ferrichrome and the aerobactin siderophores as a source of iron. Mutations in iutA or the fhu operon abolished the ability of KIM6 to use aerobactin. Mutations in the fhu operon, but not in iutA, affected the ability of KIM6 to use ferrichrome. This demonstrates that Y. pestis uses both ferrichrome and aerobactin, but has lost the ability to synthesize aerobactin.


Subject(s)
Biological Transport , Ferric Compounds/metabolism , Hydroxamic Acids/metabolism , Yersinia pestis/genetics , Yersinia pestis/physiology , Bacterial Outer Membrane Proteins/metabolism , Genes, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL
...