Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Microbiologyopen ; 11(1): e1264, 2022 02.
Article in English | MEDLINE | ID: mdl-35212475

ABSTRACT

Adaptation of opportunistic pathogens to their host environment requires reprogramming of a vast array of genes to facilitate survival in the host. Burkholderia cenocepacia, a Gram-negative bacterium with a large genome of ∼8 Mb that colonizes environmental niches, is exquisitely adaptable to the hypoxic environment of the cystic fibrosis lung and survives in macrophages. We previously identified an immunoreactive acidic protein encoded on replicon 3, BCAS0292. Deletion of the BCAS0292 gene significantly altered the abundance of 979 proteins by 1.5-fold or more; 19 proteins became undetectable while 545 proteins showed ≥1.5-fold reduced abundance, suggesting the BCAS0292 protein is a global regulator. Moreover, the ∆BCAS0292 mutant showed a range of pleiotropic effects: virulence and host-cell attachment were reduced, antibiotic susceptibility was altered, and biofilm formation enhanced. Its growth and survival were impaired in 6% oxygen. In silico prediction of its three-dimensional structure revealed BCAS0292 presents a dimeric ß-structure with a negative surface charge. The ΔBCAS0292 mutant displayed altered DNA supercoiling, implicated in global regulation of gene expression. Three proteins were identified in pull-downs with FLAG-tagged BCAS0292, including the Histone H1-like protein, HctB, which is recognized as a global transcriptional regulator. We propose that BCAS0292 protein, which we have named Burkholderia negatively surface-charged regulatory protein 1 (Bnr1), acts as a DNA-mimic and binds to DNA-binding proteins, altering DNA topology and regulating the expression of multiple genes, thereby enabling the adaptation of B. cenocepacia to highly diverse environments.


Subject(s)
Adaptation, Physiological/physiology , Bacterial Proteins/physiology , Burkholderia cenocepacia/physiology , DNA, Bacterial/physiology , Molecular Mimicry/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Burkholderia cenocepacia/genetics , Burkholderia cenocepacia/pathogenicity , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Multigene Family/genetics , Virulence
2.
Cell Microbiol ; 19(5)2017 05.
Article in English | MEDLINE | ID: mdl-27886433

ABSTRACT

The Burkholderia cepacia complex (Bcc) is a group of Gram-negative opportunistic pathogens causing infections in people with cystic fibrosis (CF). Bcc is highly antibiotic resistant, making conventional antibiotic treatment problematic. The identification of novel targets for anti-virulence therapies should improve therapeutic options for infected CF patients. We previously identified that the peptidoglycan-associated lipoprotein (Pal) was immunogenic in Bcc infected CF patients; however, its role in Bcc pathogenesis is unknown. The virulence of a pal deletion mutant (Δpal) in Galleria mellonella was 88-fold reduced (p < .001) compared to wild type. The lipopolysaccharide profiles of wild type and Δpal were identical, indicating no involvement of Pal in O-antigen transport. However, Δpal was more susceptible to polymyxin B. Structural elucidation by X-ray crystallography and calorimetry demonstrated that Pal binds peptidoglycan fragments. Δpal showed a 1.5-fold reduced stimulation of IL-8 in CF epithelial cells relative to wild type (p < .001), demonstrating that Pal is a significant driver of inflammation. The Δpal mutant had reduced binding to CFBE41o- cells, but adhesion of Pal-expressing recombinant E. coli to CFBE41o- cells was enhanced compared to wild-type E. coli (p < .0001), confirming that Pal plays a direct role in host cell attachment. Overall, Bcc Pal mediates host cell attachment and stimulation of cytokine secretion, contributing to Bcc pathogenesis.


Subject(s)
Bacterial Proteins/chemistry , Burkholderia Infections/immunology , Burkholderia cenocepacia/immunology , Epithelial Cells/physiology , Lipoproteins/chemistry , Animals , Bacterial Adhesion , Bacterial Proteins/physiology , Binding Sites , Burkholderia Infections/microbiology , Burkholderia cenocepacia/pathogenicity , Cell Adhesion , Cells, Cultured , Crystallography, X-Ray , Cystic Fibrosis/microbiology , Cytokines/metabolism , Drug Resistance, Bacterial , Epithelial Cells/microbiology , Escherichia coli , Humans , Larva/microbiology , Lipopolysaccharides/physiology , Lipoproteins/physiology , Models, Molecular , Moths , Peptidoglycan/chemistry , Polymyxins/pharmacology , Protein Binding , Protein Domains
3.
Infect Immun ; 84(5): 1424-1437, 2016 05.
Article in English | MEDLINE | ID: mdl-26902727

ABSTRACT

Members of the Burkholderia cepacia complex (Bcc) cause chronic opportunistic lung infections in people with cystic fibrosis (CF), resulting in a gradual lung function decline and, ultimately, patient death. The Bcc is a complex of 20 species and is rarely eradicated once a patient is colonized; therefore, vaccination may represent a better therapeutic option. We developed a new proteomics approach to identify bacterial proteins that are involved in the attachment of Bcc bacteria to lung epithelial cells. Fourteen proteins were reproducibly identified by two-dimensional gel electrophoresis from four Bcc strains representative of two Bcc species: Burkholderia cenocepacia, the most virulent, and B. multivorans, the most frequently acquired. Seven proteins were identified in both species, but only two were common to all four strains, linocin and OmpW. Both proteins were selected based on previously reported data on these proteins in other species. Escherichia coli strains expressing recombinant linocin and OmpW showed enhanced attachment (4.2- and 3.9-fold) to lung cells compared to the control, confirming that both proteins are involved in host cell attachment. Immunoproteomic analysis using serum from Bcc-colonized CF patients confirmed that both proteins elicit potent humoral responses in vivo Mice immunized with either recombinant linocin or OmpW were protected from B. cenocepacia and B. multivorans challenge. Both antigens induced potent antigen-specific antibody responses and stimulated strong cytokine responses. In conclusion, our approach identified adhesins that induced excellent protection against two Bcc species and are promising vaccine candidates for a multisubunit vaccine. Furthermore, this study highlights the potential of our proteomics approach to identify potent antigens against other difficult pathogens.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , Bacteriocins/metabolism , Burkholderia Infections/prevention & control , Burkholderia cepacia complex/physiology , Epithelial Cells/microbiology , Adhesins, Bacterial/immunology , Animals , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Bacteriocins/immunology , Burkholderia Infections/immunology , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/physiology , Female , Gene Expression , Humans , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Treatment Outcome
4.
PLoS One ; 8(11): e80796, 2013.
Article in English | MEDLINE | ID: mdl-24260482

ABSTRACT

Burkholderia cepacia complex (Bcc) is an opportunistic bacterial pathogen that causes chronic infections in people with cystic fibrosis (CF). It is a highly antibiotic resistant organism and Bcc infections are rarely cleared from patients, once they are colonized. The two most clinically relevant species within Bcc are Burkholderia cenocepacia and Burkholderia multivorans. The virulence of these pathogens has not been fully elucidated and the virulence proteins expressed during human infection have not been identified to date. Furthermore, given its antibiotic resistance, prevention of infection with a prophylactic vaccine may represent a better alternative than eradication of an existing infection. We have compared the immunoproteome of two strains each from these two species of Bcc, with the aim of identifying immunogenic proteins which are common to both species. Fourteen immunoreactive proteins were exclusive to both B. cenocepacia strains, while 15 were exclusive to B. multivorans. A total of 15 proteins were immunogenic across both species. DNA-directed RNA polymerase, GroEL, 38kDa porin and elongation factor-Tu were immunoreactive proteins expressed by all four strains examined. Many proteins which were immunoreactive in both species, warrant further investigations in order to aid in the elucidation of the mechanisms of pathogenesis of this difficult organism. In addition, identification of some of these could also allow the development of protective vaccines which may prevent colonisation.


Subject(s)
Bacterial Proteins/metabolism , Burkholderia cepacia complex/metabolism , Proteomics , Bacterial Proteins/immunology , Burkholderia Infections/microbiology , Burkholderia cepacia complex/immunology , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Humans , Membrane Proteins/immunology , Membrane Proteins/metabolism , Proteomics/methods
5.
Curr Protein Pept Sci ; 13(8): 807-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23305366

ABSTRACT

The increase in antibiotic resistance and the shortage of new antimicrobials to prevent difficult bacterial infections underlines the importance of prophylactic therapies to prevent infection by bacterial pathogens. Vaccination has reduced the incidence of many serious diseases, including respiratory bacterial infections. However, there are many pathogens for which no vaccine is available and some vaccines are not effective among all age groups or among immunocompromised individuals. Immunoproteomics is a powerful technique which has been used to identify potential vaccine candidates to protect against pathogenic bacteria. The combination of proteomics with the detection of immunoreactive antigens using serum highlights immunogenic proteins that are expressed during infection. This is particularly useful when patient serum is used as the antigens that promote a humoral response during human infection are identified. This review outlines examples of vaccine candidates that have been identified using immunoproteomics and have successfully protected animals against challenge when tested in immunisation studies. Many immunoreactive proteins are common to several unrelated pathogens, however some of these are not always protective in animal immunisation and challenge studies. Furthermore, examples of well-established immunogens, including Bordetella pertussis antigen FHA were not detected in immunoproteomics studies, indicating that this technology may underrepresent the immunoreactive proteins in a pathogen. Although only one step in the pathway towards an efficacious approved vaccine, immunoproteomics is an important technology in the identification of novel vaccine antigens.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Infections/immunology , Bacterial Infections/prevention & control , Bacterial Vaccines/immunology , Proteomics/methods , Respiratory Tract Infections/immunology , Respiratory Tract Infections/prevention & control , Animals , Bacterial Infections/microbiology , Host-Pathogen Interactions/immunology , Humans , Respiratory Tract Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...