Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Antimicrob Agents Chemother ; 66(11): e0070822, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36226972

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis and presents with diverse clinical manifestations. Naturally occurring infection occurs following contamination of cuts or skin abrasions, or ingestion of contaminated water, and occasionally through inhalational of infected soil or water particles. The influence of the route of disease acquisition on the efficacy of medical countermeasures has not been explored in humans or in appropriate animal models. The efficacy of co-trimoxazole against melioidosis acquired by different routes of exposure was assessed in postexposure prophylaxis (PEP) and treatment studies in marmoset models of melioidosis. Following challenge with B. pseudomallei by the inhalational, subcutaneous, or ingestion routes of administration, animals were given co-trimoxazole at 12 hourly intervals for 14 days, starting either 6 h postchallenge or at the onset of fever. Animals were then observed for 28 days. All animals that received antibiotic 6 h postchallenge survived the duration of dosing. All animals that received antibiotics at the onset of fever completed the treatment, but 10%, 57%, and 60% of those with ingestion, subcutaneous, and inhalation challenge relapsed, respectively. Bacteriological and histological differences were observed between placebo-control animals and those that relapsed. Immunological profiles indicate difference between animals given placebo and those that relapsed or survived the duration of the study. A broad T-cell activation was observed in animals that survived. Overall, these data suggest the efficacy of co-trimoxazole, as measured in the incidence of relapse, differs depending on the disease-acquisition route. Therefore, there are implications in treating this disease in regions of endemicity.


Subject(s)
Burkholderia pseudomallei , Melioidosis , Humans , Animals , Melioidosis/drug therapy , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Anti-Bacterial Agents/therapeutic use , Water , Disease Models, Animal
2.
BMC Microbiol ; 17(1): 163, 2017 Jul 21.
Article in English | MEDLINE | ID: mdl-28732479

ABSTRACT

BACKGROUND: The World Health Organization has categorized plague as a re-emerging disease and the potential for Yersinia pestis to also be used as a bioweapon makes the identification of new drug targets against this pathogen a priority. Environmental temperature is a key signal which regulates virulence of the bacterium. The bacterium normally grows outside the human host at 28 °C. Therefore, understanding the mechanisms that the bacterium used to adapt to a mammalian host at 37 °C is central to the development of vaccines or drugs for the prevention or treatment of human disease. RESULTS: Using a library of over 1 million Y. pestis CO92 random mutants and transposon-directed insertion site sequencing, we identified 530 essential genes when the bacteria were cultured at 28 °C. When the library of mutants was subsequently cultured at 37 °C we identified 19 genes that were essential at 37 °C but not at 28 °C, including genes which encode proteins that play a role in enabling functioning of the type III secretion and in DNA replication and maintenance. Using genome-scale metabolic network reconstruction we showed that growth conditions profoundly influence the physiology of the bacterium, and by combining computational and experimental approaches we were able to identify 54 genes that are essential under a broad range of conditions. CONCLUSIONS: Using an integrated computational-experimental approach we identify genes which are required for growth at 37 °C and under a broad range of environments may be the best targets for the development of new interventions to prevent or treat plague in humans.


Subject(s)
Bacterial Proteins/genetics , Computational Biology/methods , Genes, Essential , Plague/microbiology , Yersinia pestis/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Mutation , Yersinia pestis/growth & development , Yersinia pestis/metabolism
3.
Bioconjug Chem ; 27(6): 1435-46, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27124182

ABSTRACT

Melioidosis is an emerging infectious disease caused by Burkholderia pseudomallei and is associated with high morbidity and mortality rates in endemic areas. Antibiotic treatment is protracted and not always successful; even with appropriate therapy, up to 40% of individuals presenting with melioidosis in Thailand succumb to infection. In these circumstances, an effective vaccine has the potential to have a dramatic impact on both the scale and the severity of disease. Currently, no vaccines are licensed for human use. A leading vaccine candidate is the capsular polysaccharide consisting of a homopolymer of unbranched 1→3 linked 2-O-acetyl-6-deoxy-ß-d-manno-heptopyranose. Here, we present the chemical synthesis of this challenging antigen using a novel modular disaccharide assembly approach. The resulting hexasaccharide was coupled to the nontoxic Hc domain of tetanus toxin as a carrier protein to promote recruitment of T-cell help and provide a scaffold for antigen display. Mice immunized with the glycoconjugate developed IgM and IgG responses capable of recognizing native capsule, and were protected against infection with over 120 × LD50 of B. pseudomallei strain K96243. This is the first report of the chemical synthesis of an immunologically relevant and protective hexasaccharide fragment of the capsular polysaccharide of B. pseudomallei and serves as the rational starting point for the development of an effective licensed vaccine for this emerging infectious disease.


Subject(s)
Glycoconjugates/chemistry , Glycoconjugates/immunology , Mannose/chemistry , Melioidosis/prevention & control , Oligosaccharides/chemistry , Animals , Bacterial Vaccines/chemistry , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Burkholderia pseudomallei/physiology , Female , Mice , Mice, Inbred BALB C , Oligosaccharides/chemical synthesis
4.
Int J Exp Pathol ; 96(6): 414-26, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26852689

ABSTRACT

The marmoset model of melioidosis was used to explore whether there was any difference in the disease presentation and/or the lesion formation following inhalational challenge with one of four strains of Burkholderia pseudomallei (K96243, 1026b, HBPUB10303a and HBPUB10134a). Marmosets were challenged with a range of bacterial doses and bacterial load, histological and physiological features were determined temporally following lethal disease. Melioidosis presented as an acute, febrile disease with bacteraemia, bacterial dissemination, necrotizing hepatitis, splenitis and pneumonia which was independent of the challenge strain. Generally, there were no major differences in the manifestation of melioidosis following challenge by the different strains of B. pseudomallei; however, there were some differences in the time to death and the severity of the pathological features. The pathological features observed in the liver and spleen of animals challenged with B. pseudomallei strain 1026b were statistically less severe (P < 0.05) and less frequent. However, more severe foci of disease were evident in the lungs of animals challenged with strain 1026b. In all cases, the lesions developed from small areas of bacteria-infected macrophages surrounded by non-infected neutrophils into large lesions with both immune cell types infected. The marmoset model was a useful tool enabling the distinction of subtle difference in the pathological response to B. pseudomallei.


Subject(s)
Burkholderia pseudomallei/pathogenicity , Inhalation Exposure , Liver/pathology , Lung/pathology , Melioidosis/pathology , Spleen/pathology , Aerosols , Animals , Bacterial Load , Burkholderia pseudomallei/classification , Callithrix , Disease Models, Animal , Disease Progression , Female , Host-Pathogen Interactions , Liver/microbiology , Lung/microbiology , Macrophages/microbiology , Macrophages/pathology , Male , Melioidosis/microbiology , Neutrophils/pathology , Severity of Illness Index , Spleen/microbiology , Time Factors
5.
Infect Immun ; 82(8): 3206-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24866807

ABSTRACT

Burkholderia pseudomallei, the etiologic agent of melioidosis, is a CDC tier 1 select agent that causes severe disease in both humans and animals. Diagnosis and treatment of melioidosis can be challenging, and in the absence of optimal chemotherapeutic intervention, acute disease is frequently fatal. Melioidosis is an emerging infectious disease for which there are currently no licensed vaccines. Due to the potential malicious use of B. pseudomallei as well as its impact on public health in regions where the disease is endemic, there is significant interest in developing vaccines for immunization against this disease. In the present study, type A O-polysaccharide (OPS) and manno-heptose capsular polysaccharide (CPS) antigens were isolated from nonpathogenic, select-agent-excluded strains of B. pseudomallei and covalently linked to carrier proteins. By using these conjugates (OPS2B1 and CPS2B1, respectively), it was shown that although high-titer IgG responses against the OPS or CPS component of the glycoconjugates could be raised in BALB/c mice, only those animals immunized with CPS2B1 were protected against intraperitoneal challenge with B. pseudomallei. Extending upon these studies, it was also demonstrated that when the mice were immunized with a combination of CPS2B1 and recombinant B. pseudomallei LolC, rather than with CPS2B1 or LolC individually, they exhibited higher survival rates when challenged with a lethal dose of B. pseudomallei. Collectively, these results suggest that CPS-based glycoconjugates are promising candidates for the development of subunit vaccines for immunization against melioidosis.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Capsules/immunology , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Melioidosis/prevention & control , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Female , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Survival Analysis , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
6.
Int J Exp Pathol ; 95(6): 378-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25477002

ABSTRACT

Glanders and melioidosis are caused by two distinct Burkholderia species and have generally been considered to have similar disease progression. While both of these pathogens are HHS/CDC Tier 1 agents, natural infection with both these pathogens is primarily through skin inoculation. The common marmoset (Callithrix jacchus) was used to compare disease following experimental subcutaneous challenge. Acute, lethal disease was observed in marmosets following challenge with between 26 and 1.2 × 10(8) cfu Burkholderia pseudomallei within 22-85 h. The reproducibility and progression of the disease were assessed following a challenge of 1 × 10(2) cfu of B. pseudomallei. Melioidosis was characterised by high levels of bacteraemia, focal microgranuloma progressing to non-necrotic multifocal solid lesions in the livers and spleens and multi-organ failure. Lethal disease was observed in 93% of animals challenged with Burkholderia mallei, occurring between 5 and 10.6 days. Following challenge with 1 × 10(2) cfu of B. mallei, glanders was characterised with lymphatic spread of the bacteria and non-necrotic, multifocal solid lesions progressing to a multifocal lesion with severe necrosis and pneumonia. The experimental results confirmed that the disease pathology and presentation is strikingly different between the two pathogens. The marmoset provides a model of the human syndrome for both diseases facilitating the development of medical countermeasures.


Subject(s)
Burkholderia mallei , Burkholderia pseudomallei , Glanders/microbiology , Glanders/pathology , Melioidosis/microbiology , Melioidosis/pathology , Animals , Antigens, Bacterial , Bacterial Load , Callithrix , Disease Models, Animal , Female , Glanders/mortality , Injections, Subcutaneous , Male , Melioidosis/mortality , Severity of Illness Index
7.
J Bacteriol ; 193(14): 3577-87, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602339

ABSTRACT

Glycosylation of proteins is known to impart novel physical properties and biological roles to proteins from both eukaryotes and prokaryotes. In this study, gel-based glycoproteomics were used to identify glycoproteins of the potential biothreat agent Burkholderia pseudomallei and the closely related but nonpathogenic B. thailandensis. Top-down and bottom-up mass spectrometry (MS) analyses identified that the flagellin proteins of both species were posttranslationally modified by novel glycans. Analysis of proteins from two strains of each species demonstrated that B. pseudomallei flagellin proteins were modified with a glycan with a mass of 291 Da, while B. thailandensis flagellin protein was modified with related glycans with a mass of 300 or 342 Da. Structural characterization of the B. thailandensis carbohydrate moiety suggests that it is an acetylated hexuronic acid. In addition, we have identified through mutagenesis a gene from the lipopolysaccharide (LPS) O-antigen biosynthetic cluster which is involved in flagellar glycosylation, and inactivation of this gene eliminates flagellar glycosylation and motility in B. pseudomallei. This is the first report to conclusively demonstrate the presence of a carbohydrate covalently linked to a protein in B. pseudomallei and B. thailandensis, and it suggests new avenues to explore in order to examine the marked differences in virulence between these two species.


Subject(s)
Burkholderia pseudomallei/metabolism , Burkholderia/metabolism , Flagellin/metabolism , Amino Acid Sequence , Burkholderia/chemistry , Burkholderia/genetics , Burkholderia pseudomallei/chemistry , Burkholderia pseudomallei/genetics , Flagellin/chemistry , Flagellin/genetics , Glycosylation , Mass Spectrometry , Molecular Sequence Data , Molecular Weight , Peptide Mapping
8.
Infect Immun ; 79(4): 1512-25, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300775

ABSTRACT

The Burkholderia pseudomallei K96243 genome encodes six type VI secretion systems (T6SSs), but little is known about the role of these systems in the biology of B. pseudomallei. In this study, we purified recombinant Hcp proteins from each T6SS and tested them as vaccine candidates in the BALB/c mouse model of melioidosis. Recombinant Hcp2 protected 80% of mice against a lethal challenge with K96243, while recombinant Hcp1, Hcp3, and Hcp6 protected 50% of mice against challenge. Hcp6 was the only Hcp constitutively produced by B. pseudomallei in vitro; however, it was not exported to the extracellular milieu. Hcp1, on the other hand, was produced and exported in vitro when the VirAG two-component regulatory system was overexpressed in trans. We also constructed six hcp deletion mutants (Δhcp1 through Δhcp6) and tested them for virulence in the Syrian hamster model of infection. The 50% lethal doses (LD(50)s) for the Δhcp2 through Δhcp6 mutants were indistinguishable from K96243 (<10 bacteria), but the LD(50) for the Δhcp1 mutant was >10(3) bacteria. The hcp1 deletion mutant also exhibited a growth defect in RAW 264.7 macrophages and was unable to form multinucleated giant cells in this cell line. Unlike K96243, the Δhcp1 mutant was only weakly cytotoxic to RAW 264.7 macrophages 18 h after infection. The results suggest that the cluster 1 T6SS is essential for virulence and plays an important role in the intracellular lifestyle of B. pseudomallei.


Subject(s)
Gene Expression Profiling , Melioidosis/microbiology , Virulence Factors/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/metabolism , Cricetinae , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique , Gene Expression , Genes, Bacterial , Humans , Immunoblotting , Liver/microbiology , Liver/pathology , Macrophages/microbiology , Macrophages/pathology , Melioidosis/genetics , Melioidosis/metabolism , Mesocricetus , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Virulence/genetics , Virulence Factors/genetics
9.
PLoS Negl Trop Dis ; 15(2): e0009016, 2021 02.
Article in English | MEDLINE | ID: mdl-33617546

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, which is a Gram negative, facultative intracellular bacterium. Disease is prevalent in SE Asia and in northern Australia, as well as in other tropical and subtropical regions. Recently, there is an increasing awareness of the importance of bacterial ingestion as a potential route of infection, particularly in cases of unexplained origin of the disease. The marmoset is a New World Monkey (NWM) species that is being developed as an alternative NHP model to complement the more traditionally used Old World Monkeys (OWM). Models have been developed for the traditional routes of disease acquisition, subcutaneous and inhalational. This manuscript details the development and characterisation of an ingestion model of melioidosis. Dose-ranging study assessed the lethality of B. pseudomallei and disease progression was assessed by euthanizing animals at predetermined time points, 12, 36, 48 and 54 hours post-challenge. Challenge doses of greater than 6.2 x 106 cfu resulted in an acute, lethal, febrile disease. Following challenge the lung was the first organ, outside of the gastrointestinal tract, to become colonised. Enteritis (duodenitis, ileitis and/or jejunitis) was observed in sections of the small intestine from animals that succumbed to disease. However, the most severe pathological features were observed in the mesenteric lymph nodes from these animals. These findings are consistent with lymphatic draining as route of dissemination.


Subject(s)
Burkholderia pseudomallei/physiology , Lymphatic System/microbiology , Melioidosis/pathology , Animals , Burkholderia pseudomallei/pathogenicity , Callithrix , Disease Models, Animal , Enteritis/microbiology , Female , Lung/microbiology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Male , Melioidosis/microbiology
10.
Front Cell Infect Microbiol ; 10: 621635, 2020.
Article in English | MEDLINE | ID: mdl-33585288

ABSTRACT

Common marmosets (Callithrix jacchus) were shown to be susceptible to inhalational infection with Coxiella burnetii, in a dose-dependent manner, producing a disease similar to human Q fever, characterized by a resolving febrile response. Illness was also associated with weight loss, liver enzyme dysfunction, characteristic cellular activation, circulating INF-γ and bacteraemia. Viable C. burnetii was recovered from various tissues during disease and from 75% of the animal's lungs on 28 days post challenge, when there were no overt clinical features of disease but there was histological evidence of macrophage and lymphocyte infiltration into the lung resulting in granulomatous alveolitis. Taken together, these features of disease progression, physiology and bacterial spread appear to be consistent with human disease and therefore the common marmoset can be considered as a suitable model for studies on the pathogenesis or the development of medical counter measures of inhalational Q fever.


Subject(s)
Callithrix , Q Fever , Administration, Inhalation , Animals , Disease Models, Animal , Humans , Q Fever/diagnosis , Spleen
11.
Front Microbiol ; 10: 1552, 2019.
Article in English | MEDLINE | ID: mdl-31379760

ABSTRACT

Historically, disease progression in animal models of Q fever has been carried out using PCR to monitor the presence of Coxiella burnetii DNA in the host. However, the colonization and dissemination of other bacterial infections in animal models are tracked using viable counts, enabling an accurate assessment of viable bacterial load within tissues. Following recent advances in the culture methods, it has become possible to do the same with C. burnetii. Here we compare and contrast the different information gained by using PCR or viable counts to study this disease. Viable bacteria were cleared from organs much faster than previously reported when assessed by bacterial DNA, but weight loss and clinical signs improved while animals were still heavily infected.

12.
Article in English | MEDLINE | ID: mdl-30834386

ABSTRACT

We have resequenced the genomes of four Burkholderia pseudomallei K96243 laboratory cultures and compared them to the reported genome sequence that was published in 2004. Compared with the reference genome, these laboratory cultures harbored up to 42 single-nucleotide variants and up to 11 indels, including a 31.7-kb deletion in one culture.

13.
J Immunol Res ; 2018: 8087916, 2018.
Article in English | MEDLINE | ID: mdl-30622981

ABSTRACT

There is a requirement for an efficacious vaccine to protect people against infection from Francisella tularensis, the etiological agent of tularemia. The lipopolysaccharide (LPS) of F. tularensis is suboptimally protective against a parenteral lethal challenge in mice. To develop a more efficacious subunit vaccine, we have used a novel biosynthetic technique of protein glycan coupling technology (PGCT) that exploits bacterial N-linked glycosylation to recombinantly conjugate F. tularensis O-antigen glycans to the immunogenic carrier protein Pseudomonas aeruginosa exoprotein A (ExoA). Previously, we demonstrated that an ExoA glycoconjugate with two glycosylation sequons was capable of providing significant protection to mice against a challenge with a low-virulence strain of F. tularensis. Here, we have generated a more heavily glycosylated conjugate vaccine and evaluated its efficacy in a Fischer 344 rat model of tularemia. We demonstrate that this glycoconjugate vaccine protected rats against disease and the lethality of an inhalational challenge with F. tularensis Schu S4. Our data highlights the potential of this biosynthetic approach for the creation of next-generation tularemia subunit vaccines.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/physiology , Glycoconjugates/immunology , Hexosyltransferases/immunology , Tularemia/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Inhalation , Mice , Mice, Inbred BALB C , Protein Binding , Pseudomonas aeruginosa/metabolism , Rats , Rats, Inbred F344 , Vaccination
14.
Sci Rep ; 7: 41923, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165493

ABSTRACT

Massively parallel sequencing technology coupled with saturation mutagenesis has provided new and global insights into gene functions and roles. At a simplistic level, the frequency of mutations within genes can indicate the degree of essentiality. However, this approach neglects to take account of the positional significance of mutations - the function of a gene is less likely to be disrupted by a mutation close to the distal ends. Therefore, a systematic bioinformatics approach to improve the reliability of essential gene identification is desirable. We report here a parametric model which introduces a novel mutation feature together with a noise trimming approach to predict the biological significance of Tn5 mutations. We show improved performance of essential gene prediction in the bacterium Yersinia pestis, the causative agent of plague. This method would have broad applicability to other organisms and to the identification of genes which are essential for competitiveness or survival under a broad range of stresses.


Subject(s)
Bacterial Proteins/genetics , Genes, Essential , High-Throughput Nucleotide Sequencing/methods , Mutation , Plague/microbiology , Yersinia pestis/growth & development , Yersinia pestis/genetics , Computational Biology , Gene Expression Regulation, Bacterial , Genome, Bacterial , Phenotype , Virulence
15.
FEMS Microbiol Lett ; 250(1): 77-83, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16040202

ABSTRACT

Caenorhabditis elegans has been used as a host for the study of bacteria that cause disease in mammals. However, a significant limitation of the model is that C. elegans is not viable at 37 degrees C. We report that the gonochoristic nematode Panagrellus redivivus survives at 37 degrees C and maintains its life cycle at temperatures up to and including 31.5 degrees C. The C. elegans pathogens Pseudomonas aeruginosa, Salmonella enterica, Staphylococcus aureus, but not Yersinia pseudotuberculosis, reduced P. redivivus lifespan. Of four strains of Burkholderia multivorans tested, one reduced P. redivivus lifespan at both temperatures, one was avirulent at both temperatures and two strains reduced P. redivivus lifespan only at 37 degrees C. The mechanism by which one of these strains killed P. redivivus at 37 degrees C, but not at 25 degrees C, was investigated further. Killing required viable bacteria, did not involve bacterial invasion of tissues, is unlikely to be due to a diffusible, bacterial toxin and was not associated with increased numbers of live bacteria within the intestine of the worm. We believe B. multivorans may kill P. redivivus by a temperature-regulated mechanism similar to B. pseudomallei killing of C. elegans.


Subject(s)
Bacteria/pathogenicity , Rhabditida/microbiology , Animals , Bacteria/isolation & purification , Bacteria/ultrastructure , Burkholderia/pathogenicity , Caenorhabditis elegans/microbiology , Female , Humans , Longevity , Male , Microscopy, Electron , Pseudomonas aeruginosa/pathogenicity , Rhabditida/ultrastructure , Salmonella enterica/pathogenicity , Species Specificity , Staphylococcus aureus/pathogenicity , Temperature , Virulence , Yersinia pseudotuberculosis/pathogenicity
16.
PLoS One ; 10(4): e0121271, 2015.
Article in English | MEDLINE | ID: mdl-25830295

ABSTRACT

Burkholderia pseudomallei is the causative agent of the severe tropical disease melioidosis, which commonly presents as sepsis. The B. pseudomallei K96243 genome encodes eleven predicted autotransporters, a diverse family of secreted and outer membrane proteins often associated with virulence. In a systematic study of these autotransporters, we constructed insertion mutants in each gene predicted to encode an autotransporter and assessed them for three pathogenesis-associated phenotypes: virulence in the BALB/c intra-peritoneal mouse melioidosis model, net intracellular replication in J774.2 murine macrophage-like cells and survival in 45% (v/v) normal human serum. From the complete repertoire of eleven autotransporter mutants, we identified eight mutants which exhibited an increase in median lethal dose of 1 to 2-log10 compared to the isogenic parent strain (bcaA, boaA, boaB, bpaA, bpaC, bpaE, bpaF and bimA). Four mutants, all demonstrating attenuation for virulence, exhibited reduced net intracellular replication in J774.2 macrophage-like cells (bimA, boaB, bpaC and bpaE). A single mutant (bpaC) was identified that exhibited significantly reduced serum survival compared to wild-type. The bpaC mutant, which demonstrated attenuation for virulence and net intracellular replication, was sensitive to complement-mediated killing via the classical and/or lectin pathway. Serum resistance was rescued by in trans complementation. Subsequently, we expressed recombinant proteins of the passenger domain of four predicted autotransporters representing each of the phenotypic groups identified: those attenuated for virulence (BcaA), those attenuated for virulence and net intracellular replication (BpaE), the BpaC mutant with defects in virulence, net intracellular replication and serum resistance and those displaying wild-type phenotypes (BatA). Only BcaA and BpaE elicited a strong IFN-γ response in a restimulation assay using whole blood from seropositive donors and were recognised by seropositive human sera from the endemic area. To conclude, several predicted autotransporters contribute to B. pseudomallei virulence and BpaC may do so by conferring resistance against complement-mediated killing.


Subject(s)
Bacterial Proteins/genetics , Burkholderia pseudomallei/pathogenicity , Type V Secretion Systems/genetics , Virulence/genetics , Adaptive Immunity , Animals , Bacterial Proteins/metabolism , Burkholderia pseudomallei/genetics , Cell Line , Disease Models, Animal , Humans , Immunity, Cellular , Lectins/metabolism , Melioidosis/metabolism , Melioidosis/pathology , Mice , Mice, Inbred BALB C , Microbial Viability , Mutagenesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Serum/chemistry , Type V Secretion Systems/metabolism
17.
FEMS Microbiol Lett ; 234(2): 281-7, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15135534

ABSTRACT

Caenorhabditis elegans has previously been proposed as an alternative host for models of infectious disease caused by human pathogens. When exposed to some human pathogenic bacteria, the life span of nematodes is significantly reduced. We have shown that mutations in the age-1, and/or age-2 genes of C. elegans, that normally enhance life expectancy, can also increase resistance to killing by the bacterial pathogens Pseudomonas aeruginosa, Salmonella enterica var. Typhimurium, Burkholderia cepacia or Yersinia pseudotuberculosis. We also found that the rate at which wild-type C. elegans was killed by the bacterial pathogens tested increased as nematodes aged. In the case of P. aeruginosa infection, the difference in life span of wild type and age-1 mutants of C. elegans was not due to differences in the level of bacterial colonisation of the gut.


Subject(s)
Bacteria/pathogenicity , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Immunity, Innate/genetics , Animals , Burkholderia cepacia/pathogenicity , Disease Models, Animal , Infections , Mutation , Salmonella/pathogenicity , Yersinia/pathogenicity , Yersinia pseudotuberculosis/pathogenicity
18.
FEMS Microbiol Lett ; 210(2): 181-5, 2002 May 07.
Article in English | MEDLINE | ID: mdl-12044672

ABSTRACT

Caenorhabditis elegans has previously been used as an alternative to mammalian models of infection with bacterial pathogens. We have developed a liquid-based assay to measure the effect of bacteria on the feeding ability of C. elegans. Using this assay we have shown that Pseudomonas aeruginosa strain PA14, Burkholderia pseudomallei and Yersinia pestis were able to inhibit feeding of C. elegans strain N2. An increase in sensitivity of the assay was achieved by using C. elegans mutant phm-2, in place of the wild-type strain. Using this assay,P. aeruginosa PA01 inhibited the feeding of C. elegans mutant phm-2. Such liquid-based feeding assays are ideally suited to the high-throughput screening of mutants of bacterial pathogens.


Subject(s)
Biological Assay/methods , Burkholderia pseudomallei/pathogenicity , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Pseudomonas aeruginosa/pathogenicity , Yersinia pestis/pathogenicity , Animals , Bacterial Infections/genetics , Bacterial Infections/microbiology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Feeding Behavior , Models, Biological , Mutation , Sensitivity and Specificity
19.
J Med Microbiol ; 53(Pt 12): 1177-1182, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15585494

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, which is a major cause of morbidity and mortality in endemic regions. Currently there is no human vaccine against melioidosis. In this study, LPS or capsular polysaccharide was used to immunize BALB/c mice. The different polysaccharide antigens induced antibody responses. Mice vaccinated with LPS developed predominantly IgM and IgG3 responses. Contrastingly, mice vaccinated with capsular polysaccharide developed a predominantly IgG2b response. After immunization, mice were challenged by the intra-peritoneal route and an increased mean time to death was observed compared with unvaccinated controls. Immunization with LPS provided an optimal protective response. Mice challenged by the aerosol route showed a small increase in the mean time to death compared with the unvaccinated controls. The passive transfer of antigen from immunized into naive mice provided protection against a subsequent challenge. This study is the first time antigens protective by active immunization have been identified and suggests that polysaccharides have potential as vaccine candidates against melioidosis.


Subject(s)
Bacterial Capsules/immunology , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Lipid A/analogs & derivatives , Lipopolysaccharides/immunology , Melioidosis/prevention & control , Aerosols , Animals , Antibodies, Bacterial/immunology , Bacterial Capsules/administration & dosage , Cell Wall Skeleton/administration & dosage , Cell Wall Skeleton/immunology , Cord Factors/administration & dosage , Cord Factors/immunology , Female , Immunization, Passive , Immunoglobulin G/biosynthesis , Injections, Intraperitoneal , Lipid A/administration & dosage , Lipid A/immunology , Lipopolysaccharides/administration & dosage , Melioidosis/immunology , Mice , Mice, Inbred BALB C , Specific Pathogen-Free Organisms , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL