Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Clin Vaccine Immunol ; 22(12): 1255-68, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26446423

ABSTRACT

Earlier, we showed that the Δlpp ΔmsbB Δail triple mutant of Yersinia pestis CO92 with deleted genes encoding Braun lipoprotein (Lpp), an acyltransferase (MsbB), and the attachment invasion locus (Ail), respectively, was avirulent in a mouse model of pneumonic plague. In this study, we further evaluated the immunogenic potential of the Δlpp ΔmsbB Δail triple mutant and its derivative by different routes of vaccination. Mice were immunized via the subcutaneous (s.c.) or the intramuscular (i.m.) route with two doses (2 × 10(6) CFU/dose) of the above-mentioned triple mutant with 100% survivability of the animals. Upon subsequent pneumonic challenge with 70 to 92 50% lethal doses (LD(50)) of wild-type (WT) strain CO92, all of the mice survived when immunization occurred by the i.m. route. Since Ail has virulence and immunogenic potential, a mutated version of Ail devoid of its virulence properties was created, and the genetically modified ail replaced the native ail gene on the chromosome of the Δlpp ΔmsbB double mutant, creating a Δlpp ΔmsbB::ailL2 vaccine strain. This newly generated mutant was attenuated similarly to the Δlpp ΔmsbB Δail triple mutant when administered by the i.m. route and provided 100% protection to animals against subsequent pneumonic challenge. Not only were the two above-mentioned mutants cleared rapidly from the initial i.m. site of injection in animals with no histopathological lesions, the immunized mice did not exhibit any disease symptoms during immunization or after subsequent exposure to WT CO92. These two mutants triggered balanced Th1- and Th2-based antibody responses and cell-mediated immunity. A substantial increase in interleukin-17 (IL-17) from the T cells of vaccinated mice, a cytokine of the Th17 cells, further augmented their vaccine potential. Thus, the Δlpp ΔmsbB Δail and Δlpp ΔmsbB::ailL2 mutants represent excellent vaccine candidates for plague, with the latter mutant still retaining Ail immunogenicity but with a much diminished virulence potential.


Subject(s)
Mutation , Plague Vaccine/immunology , Plague/prevention & control , Yersinia pestis/genetics , Yersinia pestis/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Disease Models, Animal , Gene Deletion , Immunity, Cellular , Immunity, Humoral , Immunization , Injections, Intramuscular , Lipoproteins/immunology , Mice , Plague/immunology , Plague/microbiology , Plague Vaccine/administration & dosage , Plague Vaccine/genetics , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
2.
Microb Pathog ; 80: 27-38, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25697665

ABSTRACT

We recently characterized the Δlpp Δpla double in-frame deletion mutant of Yersinia pestis CO92 molecularly, biologically, and immunologically. While Braun lipoprotein (Lpp) activates toll-like receptor-2 to initiate an inflammatory cascade, plasminogen activator (Pla) protease facilitates bacterial dissemination in the host. The Δlpp Δpla double mutant was highly attenuated in evoking bubonic and pneumonic plague, was rapidly cleared from mouse organs, and generated humoral and cell-mediated immune responses to provide subsequent protection to mice against a lethal challenge dose of wild-type (WT) CO92. Here, we further characterized the Δlpp Δpla double mutant in two murine macrophage cell lines as well as in primary human monocyte-derived macrophages to gauge its potential as a live-attenuated vaccine candidate. We first demonstrated that the Δpla single and the Δlpp Δpla double mutant were unable to survive efficiently in murine and human macrophages, unlike WT CO92. We observed that the levels of Pla and its associated protease activity were not affected in the Δlpp single mutant, and, likewise, deletion of the pla gene from WT CO92 did not alter Lpp levels. Further, our study revealed that both Lpp and Pla contributed to the intracellular survival of WT CO92 via different mechanisms. Importantly, the ability of the Δlpp Δpla double mutant to be phagocytized by macrophages, to stimulate production of tumor necrosis factor-α and interleukin-6, and to activate the nitric oxide killing pathways of the host cells remained unaltered when compared to the WT CO92-infected macrophages. Finally, macrophages infected with either the WT CO92 or the Δlpp Δpla double mutant were equally efficient in their uptake of zymosan particles as determined by flow cytometric analysis. Overall, our data indicated that although the Δlpp Δpla double mutant of Y. pestis CO92 was highly attenuated, it retained the ability to elicit innate and subsequent acquired immune responses in the host similar to that of WT CO92, which are highly desirable in a live-attenuated vaccine candidate.


Subject(s)
Gene Deletion , Lipoproteins/deficiency , Macrophages, Alveolar/microbiology , Macrophages/microbiology , Peptide Hydrolases/deficiency , Plasminogen Activators/deficiency , Yersinia pestis/growth & development , Animals , Cells, Cultured , Humans , Immunity, Innate , Mice , Microbial Viability , Plague Vaccine , Vaccines, Attenuated , Virulence , Yersinia pestis/genetics
3.
Infect Immun ; 83(4): 1318-38, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605764

ABSTRACT

Previously, we showed that deletion of genes encoding Braun lipoprotein (Lpp) and MsbB attenuated Yersinia pestis CO92 in mouse and rat models of bubonic and pneumonic plague. While Lpp activates Toll-like receptor 2, the MsbB acyltransferase modifies lipopolysaccharide. Here, we deleted the ail gene (encoding the attachment-invasion locus) from wild-type (WT) strain CO92 or its lpp single and Δlpp ΔmsbB double mutants. While the Δail single mutant was minimally attenuated compared to the WT bacterium in a mouse model of pneumonic plague, the Δlpp Δail double mutant and the Δlpp ΔmsbB Δail triple mutant were increasingly attenuated, with the latter being unable to kill mice at a 50% lethal dose (LD50) equivalent to 6,800 LD50s of WT CO92. The mutant-infected animals developed balanced TH1- and TH2-based immune responses based on antibody isotyping. The triple mutant was cleared from mouse organs rapidly, with concurrent decreases in the production of various cytokines and histopathological lesions. When surviving animals infected with increasing doses of the triple mutant were subsequently challenged on day 24 with the bioluminescent WT CO92 strain (20 to 28 LD50s), 40 to 70% of the mice survived, with efficient clearing of the invading pathogen, as visualized in real time by in vivo imaging. The rapid clearance of the triple mutant, compared to that of WT CO92, from animals was related to the decreased adherence and invasion of human-derived HeLa and A549 alveolar epithelial cells and to its inability to survive intracellularly in these cells as well as in MH-S murine alveolar and primary human macrophages. An early burst of cytokine production in macrophages elicited by the triple mutant compared to WT CO92 and the mutant's sensitivity to the bactericidal effect of human serum would further augment bacterial clearance. Together, deletion of the ail gene from the Δlpp ΔmsbB double mutant severely attenuated Y. pestis CO92 to evoke pneumonic plague in a mouse model while retaining the required immunogenicity needed for subsequent protection against infection.


Subject(s)
Acyltransferases/genetics , Bacterial Outer Membrane Proteins/genetics , Lipoproteins/genetics , Plague/immunology , Virulence Factors/genetics , Yersinia pestis/pathogenicity , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/immunology , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Cell Line , Disease Models, Animal , Drug Resistance, Bacterial/genetics , Female , Gene Deletion , Gentamicins/pharmacology , HeLa Cells , Humans , Intracellular Space/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Plague/pathology , Yersinia pestis/genetics , Yersinia pestis/immunology
4.
Appl Environ Microbiol ; 80(14): 4162-83, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24795370

ABSTRACT

The genomes of 10 Aeromonas isolates identified and designated Aeromonas hydrophila WI, Riv3, and NF1 to NF4; A. dhakensis SSU; A. jandaei Riv2; and A. caviae NM22 and NM33 were sequenced and annotated. Isolates NF1 to NF4 were from a patient with necrotizing fasciitis (NF). Two environmental isolates (Riv2 and -3) were from the river water from which the NF patient acquired the infection. While isolates NF2 to NF4 were clonal, NF1 was genetically distinct. Outside the conserved core genomes of these 10 isolates, several unique genomic features were identified. The most virulent strains possessed one of the following four virulence factors or a combination of them: cytotoxic enterotoxin, exotoxin A, and type 3 and 6 secretion system effectors AexU and Hcp. In a septicemic-mouse model, SSU, NF1, and Riv2 were the most virulent, while NF2 was moderately virulent. These data correlated with high motility and biofilm formation by the former three isolates. Conversely, in a mouse model of intramuscular infection, NF2 was much more virulent than NF1. Isolates NF2, SSU, and Riv2 disseminated in high numbers from the muscular tissue to the visceral organs of mice, while NF1 reached the liver and spleen in relatively lower numbers on the basis of colony counting and tracking of bioluminescent strains in real time by in vivo imaging. Histopathologically, degeneration of myofibers with significant infiltration of polymorphonuclear cells due to the highly virulent strains was noted. Functional genomic analysis provided data that allowed us to correlate the highly infectious nature of Aeromonas pathotypes belonging to several different species with virulence signatures and their potential ability to cause NF.


Subject(s)
Aeromonas hydrophila/genetics , Fasciitis, Necrotizing/microbiology , Genes, Bacterial , Virulence Factors/genetics , Aeromonas hydrophila/isolation & purification , Aeromonas hydrophila/pathogenicity , Animals , Biofilms/growth & development , DNA, Bacterial/genetics , Disease Models, Animal , Enterotoxins/metabolism , Female , Fresh Water/microbiology , Genetic Association Studies , Gram-Negative Bacterial Infections/microbiology , Humans , Mice , Phylogeny , Plague/microbiology , Plasmids/genetics , Sequence Alignment , Sequence Analysis, DNA , Water Microbiology
5.
Infect Immun ; 82(6): 2485-503, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24686064

ABSTRACT

Currently, there is no FDA-approved vaccine against Yersinia pestis, the causative agent of bubonic and pneumonic plague. Since both humoral immunity and cell-mediated immunity are essential in providing the host with protection against plague, we developed a live-attenuated vaccine strain by deleting the Braun lipoprotein (lpp) and plasminogen-activating protease (pla) genes from Y. pestis CO92. The Δlpp Δpla double isogenic mutant was highly attenuated in evoking both bubonic and pneumonic plague in a mouse model. Further, animals immunized with the mutant by either the intranasal or the subcutaneous route were significantly protected from developing subsequent pneumonic plague. In mice, the mutant poorly disseminated to peripheral organs and the production of proinflammatory cytokines concurrently decreased. Histopathologically, reduced damage to the lungs and livers of mice infected with the Δlpp Δpla double mutant compared to the level of damage in wild-type (WT) CO92-challenged animals was observed. The Δlpp Δpla mutant-immunized mice elicited a humoral immune response to the WT bacterium, as well as to CO92-specific antigens. Moreover, T cells from mutant-immunized animals exhibited significantly higher proliferative responses, when stimulated ex vivo with heat-killed WT CO92 antigens, than mice immunized with the same sublethal dose of WT CO92. Likewise, T cells from the mutant-immunized mice produced more gamma interferon (IFN-γ) and interleukin-4. These animals had an increasing number of tumor necrosis factor alpha (TNF-α)-producing CD4(+) and CD8(+) T cells than WT CO92-infected mice. These data emphasize the role of TNF-α and IFN-γ in protecting mice against pneumonic plague. Overall, our studies provide evidence that deletion of the lpp and pla genes acts synergistically in protecting animals against pneumonic plague, and we have demonstrated an immunological basis for this protection.


Subject(s)
Lipoproteins/metabolism , Plague/microbiology , Plasminogen Activators/metabolism , Yersinia pestis/pathogenicity , Analysis of Variance , Animals , Antibodies, Bacterial/metabolism , Chemokines/metabolism , Colony Count, Microbial , Cytokines/metabolism , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Lipoproteins/genetics , Macrophages/microbiology , Mice , Plague/immunology , Plasminogen Activators/genetics , Virulence , Yersinia pestis/genetics , Yersinia pestis/immunology
6.
Astrobiology ; 13(9): 821-32, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23988036

ABSTRACT

Previously, we reported that there was no enhancement in the virulence potential (as measured by cell culture infections) of the bacterial pathogen Yersinia pestis (YP) following modeled microgravity/clinorotation growth. We have now further characterized the effects of clinorotation (CR) on YP growth kinetics, antibiotic sensitivity, cold growth, and YP's virulence potential in a murine model of infection. Surprisingly, none of the aforementioned phenotypes were altered. To better understand why CR did not enhance YP's virulence potential as it did for other bacterial pathogens, a YP ΔymoA isogenic mutant in the KIM/D27 background strain that is unable to produce the histone-like YmoA protein and influences DNA topography was used in both cell culture and murine models of infection. YmoA represses type three secretion system (T3SS) virulence gene expression in the yersiniae. Similar to our CR-grown parental YP strain data, the CR-grown ΔymoA mutant induced reduced HeLa cell cytotoxicity with concomitantly decreased Yersinia outer protein E (YopE) and low calcium response V (LcrV) antigen production and secretion. Important, however, were our findings that, although no significant differences were observed in survival of mice infected intraperitoneally with either normal gravity (NG)- or CR-grown parental YP, the ΔymoA mutant induced significantly more mortality in infected mice than did the parental strain following CR growth. Taken together, our data demonstrate that CR did enhance the virulence potential of the YP ΔymoA mutant in a murine infection model (relative to the CR-grown parental strain), despite inducing less HeLa cell rounding in our cell culture infection assay due to reduced T3SS activity. Therefore, CR, which induces a unique type of bacterial stress, might be enhancing YP's virulence potential in vivo through a T3SS-independent mechanism when the histone-like YmoA protein is absent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Cold Temperature , Mutation/genetics , Weightlessness , Yersinia pestis/growth & development , Yersinia pestis/pathogenicity , Animals , Antigens, Bacterial , Bacterial Proteins/metabolism , Drug Resistance, Microbial/drug effects , Female , HeLa Cells , Humans , Mice , Microbial Sensitivity Tests , Phenotype , Plague/microbiology , Plague/pathology , Pore Forming Cytotoxic Proteins/metabolism , Rotation , Virulence/drug effects , Yersinia pestis/drug effects
7.
PLoS Pathog ; 9(7): e1003495, 2013.
Article in English | MEDLINE | ID: mdl-23853602

ABSTRACT

Pneumonic plague is a highly virulent infectious disease with 100% mortality rate, and its causative organism Yersinia pestis poses a serious threat for deliberate use as a bioterror agent. Currently, there is no FDA approved vaccine against plague. The polymeric bacterial capsular protein F1, a key component of the currently tested bivalent subunit vaccine consisting, in addition, of low calcium response V antigen, has high propensity to aggregate, thus affecting its purification and vaccine efficacy. We used two basic approaches, structure-based immunogen design and phage T4 nanoparticle delivery, to construct new plague vaccines that provided complete protection against pneumonic plague. The NH2-terminal ß-strand of F1 was transplanted to the COOH-terminus and the sequence flanking the ß-strand was duplicated to eliminate polymerization but to retain the T cell epitopes. The mutated F1 was fused to the V antigen, a key virulence factor that forms the tip of the type three secretion system (T3SS). The F1mut-V protein showed a dramatic switch in solubility, producing a completely soluble monomer. The F1mut-V was then arrayed on phage T4 nanoparticle via the small outer capsid protein, Soc. The F1mut-V monomer was robustly immunogenic and the T4-decorated F1mut-V without any adjuvant induced balanced TH1 and TH2 responses in mice. Inclusion of an oligomerization-deficient YscF, another component of the T3SS, showed a slight enhancement in the potency of F1-V vaccine, while deletion of the putative immunomodulatory sequence of the V antigen did not improve the vaccine efficacy. Both the soluble (purified F1mut-V mixed with alhydrogel) and T4 decorated F1mut-V (no adjuvant) provided 100% protection to mice and rats against pneumonic plague evoked by high doses of Y. pestis CO92. These novel platforms might lead to efficacious and easily manufacturable next generation plague vaccines.


Subject(s)
Antigens, Bacterial/metabolism , Antigens, Viral/metabolism , Bacteriophage T4/immunology , Capsid/immunology , Plague/immunology , Vaccines, Virus-Like Particle/immunology , Yersinia pestis/virology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriophage T4/chemistry , Bacteriophage T4/metabolism , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Female , Mice , Mice, Inbred BALB C , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Particle Size , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plague/microbiology , Plague/prevention & control , Plague/virology , Plague Vaccine/chemistry , Plague Vaccine/immunology , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Protein Interaction Domains and Motifs , Random Allocation , Rats , Rats, Inbred BN , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Vaccines, Virus-Like Particle/chemistry , Yersinia pestis/immunology
8.
mBio ; 4(2): e00064-13, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23611906

ABSTRACT

UNLABELLED: Aeromonas hydrophila has increasingly been implicated as a virulent and antibiotic-resistant etiologic agent in various human diseases. In a previously published case report, we described a subject with a polymicrobial wound infection that included a persistent and aggressive strain of A. hydrophila (E1), as well as a more antibiotic-resistant strain of A. hydrophila (E2). To better understand the differences between pathogenic and environmental strains of A. hydrophila, we conducted comparative genomic and functional analyses of virulence-associated genes of these two wound isolates (E1 and E2), the environmental type strain A. hydrophila ATCC 7966(T), and four other isolates belonging to A. aquariorum, A. veronii, A. salmonicida, and A. caviae. Full-genome sequencing of strains E1 and E2 revealed extensive differences between the two and strain ATCC 7966(T). The more persistent wound infection strain, E1, harbored coding sequences for a cytotoxic enterotoxin (Act), a type 3 secretion system (T3SS), flagella, hemolysins, and a homolog of exotoxin A found in Pseudomonas aeruginosa. Corresponding phenotypic analyses with A. hydrophila ATCC 7966(T) and SSU as reference strains demonstrated the functionality of these virulence genes, with strain E1 displaying enhanced swimming and swarming motility, lateral flagella on electron microscopy, the presence of T3SS effector AexU, and enhanced lethality in a mouse model of Aeromonas infection. By combining sequence-based analysis and functional assays, we characterized an A. hydrophila pathotype, exemplified by strain E1, that exhibited increased virulence in a mouse model of infection, likely because of encapsulation, enhanced motility, toxin secretion, and cellular toxicity. IMPORTANCE: Aeromonas hydrophila is a common aquatic bacterium that has increasingly been implicated in serious human infections. While many determinants of virulence have been identified in Aeromonas, rapid identification of pathogenic versus nonpathogenic strains remains a challenge for this genus, as it is for other opportunistic pathogens. This paper demonstrates, by using whole-genome sequencing of clinical Aeromonas strains, followed by corresponding virulence assays, that comparative genomics can be used to identify a virulent subtype of A. hydrophila that is aggressive during human infection and more lethal in a mouse model of infection. This aggressive pathotype contained genes for toxin production, toxin secretion, and bacterial motility that likely enabled its pathogenicity. Our results highlight the potential of whole-genome sequencing to transform microbial diagnostics; with further advances in rapid sequencing and annotation, genomic analysis will be able to provide timely information on the identities and virulence potential of clinically isolated microorganisms.


Subject(s)
Aeromonas hydrophila/genetics , Aeromonas hydrophila/isolation & purification , Gram-Negative Bacterial Infections/microbiology , Virulence Factors/genetics , Wound Infection/microbiology , Aeromonas hydrophila/cytology , Aeromonas hydrophila/pathogenicity , Animals , Disease Models, Animal , Female , Flagella/physiology , Genome, Bacterial , Genotype , Humans , Locomotion , Mice , Microscopy, Electron , Molecular Sequence Data , Sequence Analysis, DNA
9.
Microbiology (Reading) ; 159(Pt 6): 1120-1135, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23519162

ABSTRACT

Aeromonas hydrophila, a Gram-negative bacterium, is an emerging human pathogen equipped with both a type 3 and a type 6 secretion system (T6SS). In this study, we evaluated the roles played by paralogous T6SS effector proteins, hemolysin co-regulated proteins (Hcp-1 and -2) and valine glycine repeat G (VgrG-1, -2 and -3) protein family members in A. hydrophila SSU pathogenesis by generating various combinations of deletion mutants of the their genes. In addition to their predicted roles as structural components and effector proteins of the T6SS, our data clearly demonstrated that paralogues of Hcp and VgrG also influenced bacterial motility, protease production and biofilm formation. Surprisingly, there was limited to no observed functional redundancy among and/or between the aforementioned T6SS effector paralogues in multiple assays. Our data indicated that Hcp and VgrG paralogues located within the T6SS cluster were more involved in forming T6SS structures, while the primary roles of Hcp-1 and VgrG-1, located outside of the T6SS cluster, were as T6SS effectors. In terms of influence on bacterial physiology, Hcp-1, but not Hcp-2, influenced bacterial motility and protease production, and in its absence, increases in both of the aforementioned activities were observed. Likewise, VgrG-1 played a major role in regulating bacterial protease production, while VgrG-2 and VgrG-3 were critical in regulating bacterial motility and biofilm formation. In an intraperitoneal murine model of infection, all Hcp and VgrG paralogues were required for optimal bacterial virulence and dissemination to mouse peripheral organs. Importantly, the observed phenotypic alterations of the T6SS mutants could be fully complemented. Taking these results together, we have further established the roles played by the two known T6SS effectors of A. hydrophila by defining their contributions to T6SS function and virulence in both in vitro and in vivo models of infection.


Subject(s)
Aeromonas hydrophila/pathogenicity , Bacterial Proteins/metabolism , Virulence Factors/metabolism , Aeromonas hydrophila/genetics , Aeromonas hydrophila/physiology , Animal Structures/microbiology , Animals , Bacterial Proteins/genetics , Biofilms/growth & development , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Disease Models, Animal , Female , Gene Deletion , Genetic Complementation Test , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Locomotion , Mice , Molecular Sequence Data , Peptide Hydrolases/metabolism , Sequence Analysis, DNA , Virulence , Virulence Factors/genetics
10.
Infect Immun ; 81(3): 815-28, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23275092

ABSTRACT

Braun (murein) lipoprotein (Lpp) and lipopolysaccharide (LPS) are major components of the outer membranes of Enterobacteriaceae family members that are capable of triggering inflammatory immune responses by activating Toll-like receptors 2 and 4, respectively. Expanding on earlier studies that demonstrated a role played by Lpp in Yersinia pestis virulence in mouse models of bubonic and pneumonic plague, we characterized an msbB in-frame deletion mutant incapable of producing an acyltransferase that is responsible for the addition of lauric acid to the lipid A moiety of LPS, as well as a Δlpp ΔmsbB double mutant of the highly virulent Y. pestis CO92 strain. Although the ΔmsbB single mutant was minimally attenuated, the Δlpp single mutant and the Δlpp ΔmsbB double mutant were significantly more attenuated than the isogenic wild-type (WT) bacterium in bubonic and pneumonic animal models (mouse and rat) of plague. These data correlated with greatly reduced survivability of the aforementioned mutants in murine macrophages. Furthermore, the Δlpp ΔmsbB double mutant was grossly compromised in its ability to disseminate to distal organs in mice and in evoking cytokines/chemokines in infected animal tissues. Importantly, mice that survived challenge with the Δlpp ΔmsbB double mutant, but not the Δlpp or ΔmsbB single mutant, in a pneumonic plague model were significantly protected against a subsequent lethal WT CO92 rechallenge. These data were substantiated by the fact that the Δlpp ΔmsbB double mutant maintained an immunogenicity comparable to that of the WT strain and induced long-lasting T-cell responses against heat-killed WT CO92 antigens. Taken together, the data indicate that deletion of the msbB gene augmented the attenuation of the Δlpp mutant by crippling the spread of the double mutant to the peripheral organs of animals and by inducing cytokine/chemokine responses. Thus, the Δlpp ΔmsbB double mutant could provide a new live-attenuated background vaccine candidate strain, and this should be explored in the future.


Subject(s)
Lipopolysaccharides/metabolism , Lipoproteins/metabolism , Plague/microbiology , Yersinia pestis/pathogenicity , Animals , Anti-Bacterial Agents/pharmacology , Female , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Gentamicins/pharmacology , Lipoproteins/genetics , Mice , Microbial Sensitivity Tests , Rats , Virulence , Yersinia pestis/drug effects , Yersinia pestis/genetics
11.
Microb Pathog ; 55: 39-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23063826

ABSTRACT

The gold standard in microbiology for monitoring bacterial dissemination in infected animals has always been viable plate counts. This method, despite being quantitative, requires sacrificing the infected animals. Recently, however, an alternative method of in vivo imaging of bioluminescent bacteria (IVIBB) for monitoring microbial dissemination within the host has been employed. Yersinia pestis is a Gram-negative bacterium capable of causing bubonic, septicemic, and pneumonic plague. In this study, we compared the conventional counting of bacterial colony forming units (cfu) in the various infected tissues to IVIBB in monitoring Y. pestis dissemination in a mouse model of pneumonic plague. By using a transposon mutagenesis system harboring the luciferase (luc) gene, we screened approximately 4000 clones and obtained a fully virulent, luc-positive Y. pestis CO92 (Y. pestis-luc2) reporter strain in which transposition occurred within the largest pMT1 plasmid which possesses murine toxin and capsular antigen encoding genes. The aforementioned reporter strain and the wild-type CO92 exhibited similar growth curves, formed capsule based on immunofluorescence microscopy and flow cytometry, and had a similar LD(50). Intranasal infection of mice with 15 LD(50) of CO92-luc2 resulted in animal mortality by 72 h, and an increasing number of bioluminescent bacteria were observed in various mouse organs over a 24-72 h period when whole animals were imaged. However, following levofloxacin treatment (10 mg/kg/day) for 6 days 24 h post infection, no luminescence was observed after 72 h of infection, indicating that the tested antimicrobial killed bacteria preventing their detection in host peripheral tissues. Overall, we demonstrated that IVIBB is an effective and non-invasive way of monitoring bacterial dissemination in animals following pneumonic plague having strong correlation with cfu, and our reporter CO92-luc2 strain can be employed as a useful tool to monitor the efficacy of antimicrobial countermeasures in real time.


Subject(s)
Microscopy, Fluorescence/methods , Molecular Imaging/methods , Plague/microbiology , Yersinia pestis/chemistry , Animals , Animals, Outbred Strains , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Female , Flow Cytometry , Genes, Reporter , Humans , Levofloxacin , Luciferases/genetics , Luciferases/metabolism , Mice , Ofloxacin/pharmacology , Virulence/drug effects , Yersinia pestis/drug effects , Yersinia pestis/genetics , Yersinia pestis/pathogenicity
12.
Clin Vaccine Immunol ; 20(2): 227-38, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23239803

ABSTRACT

Plague caused by Yersinia pestis manifests itself in bubonic, septicemic, and pneumonic forms. Although the U.S. Food and Drug Administration recently approved levofloxacin, there is no approved human vaccine against plague. The capsular antigen F1 and the low-calcium-response V antigen (LcrV) of Y. pestis represent excellent vaccine candidates; however, the inability of the immune responses to F1 and LcrV to provide protection against Y. pestis F1(-) strains or those which harbor variants of LcrV is a significant concern. Here, we show that the passive transfer of hyperimmune sera from rats infected with the plague bacterium and rescued by levofloxacin protected naive animals against pneumonic plague. Furthermore, 10 to 12 protein bands from wild-type (WT) Y. pestis CO92 reacted with the aforementioned hyperimmune sera upon Western blot analysis. Based on mass spectrometric analysis, four of these proteins were identified as attachment invasion locus (Ail/OmpX), plasminogen-activating protease (Pla), outer membrane protein A (OmpA), and F1. The genes encoding these proteins were cloned, and the recombinant proteins purified from Escherichia coli for immunization purposes before challenging mice and rats with either the F1(-) mutant or WT CO92 in bubonic and pneumonic plague models. Although antibodies to Ail and OmpA protected mice against bubonic plague when challenged with the F1(-) CO92 strain, Pla antibodies were protective against pneumonic plague. In the rat model, antibodies to Ail provided protection only against pneumonic plague after WT CO92 challenge. Together, the addition of Y. pestis outer membrane proteins to a new-generation recombinant vaccine could provide protection against a wide variety of Y. pestis strains.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Immunization, Passive , Plague Vaccine/immunology , Recombinant Proteins/immunology , Yersinia pestis/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Immune Sera , Immunization , Levofloxacin , Mice , Ofloxacin/therapeutic use , Plague/immunology , Plague/microbiology , Plague/prevention & control , Plague Vaccine/therapeutic use , Plasminogen Activators/genetics , Plasminogen Activators/immunology , Rats , Recombinant Proteins/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , Virulence Factors/genetics , Virulence Factors/immunology , Yersinia pestis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...