Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(7): e0130964, 2015.
Article in English | MEDLINE | ID: mdl-26158540

ABSTRACT

The classical bordetellae are comprised of three subspecies that differ from broad to very limited host specificity. Although several lineages appear to have specialized to particular host species, most retain the ability to colonize and grow in mice, providing a powerful common experimental model to study their differences. One of the subspecies, Bordetella parapertussis, is composed of two distinct clades that have specialized to different hosts: one to humans (Bpphu), and the other to sheep (Bppov). While Bpphu and the other classical bordetellae can efficiently colonize mice, Bppov strains are severely defective in their ability to colonize the murine respiratory tract. Bppov genomic analysis did not reveal the loss of adherence genes, but substantial mutations and deletions of multiple genes involved in the production of O-antigen, which is required to prevent complement deposition on B. bronchiseptica and Bpphu strains. Bppov lacks O-antigen and, like O-antigen mutants of other bordetellae, is highly sensitive to murine complement-mediated killing in vitro. Based on these results, we hypothesized that Bppov failed to colonize mice because of its sensitivity to murine complement. Consistent with this, the Bppov defect in the colonization of wild type mice was not observed in mice lacking the central complement component C3. Furthermore, Bppov strains were highly susceptible to killing by murine complement, but not by sheep complement. These data demonstrate that the failure of Bppov to colonize mice is due to sensitivity to murine, but not sheep, complement, providing a mechanistic example of how specialization that accompanies expansion in one host can limit host range.


Subject(s)
Bordetella Infections/immunology , Bordetella parapertussis/immunology , Complement System Proteins/immunology , Sheep Diseases/immunology , Animals , Bordetella Infections/genetics , Bordetella Infections/microbiology , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/immunology , Bordetella bronchiseptica/pathogenicity , Bordetella parapertussis/genetics , Bordetella parapertussis/pathogenicity , Complement C3/genetics , Complement C3/immunology , Complement System Proteins/genetics , Host Specificity/genetics , Host Specificity/immunology , Humans , Lung/immunology , Lung/microbiology , Mice, Inbred C57BL , Mice, Knockout , Nasal Cavity/immunology , Nasal Cavity/microbiology , O Antigens/genetics , O Antigens/immunology , Sheep , Sheep Diseases/genetics , Sheep Diseases/microbiology , Species Specificity , Trachea/immunology , Trachea/microbiology , Virulence/genetics , Virulence/immunology
2.
BMC Evol Biol ; 13: 209, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-24067113

ABSTRACT

BACKGROUND: Horizontal gene transfer (HGT) allows for rapid spread of genetic material between species, increasing genetic and phenotypic diversity. Although HGT contributes to adaptation and is widespread in many bacteria, others show little HGT. This study builds on previous work to analyze the evolutionary mechanisms contributing to variation within the locus encoding a prominent antigen of the classical bordetellae. RESULTS: We observed amongst classical bordetellae discrete regions of the lipopolysaccharide O-antigen locus with higher sequence diversity than the genome average. Regions of this locus had less than 50% sequence similarity, low dN/dS ratios and lower GC content compared to the genome average. Additionally, phylogenetic tree topologies based on genome-wide SNPs were incongruent with those based on genes within these variable regions, suggesting portions of the O-antigen locus may have been horizontally transferred. Furthermore, several predicted recombination breakpoints correspond with the ends of these variable regions. To examine the evolutionary forces that might have selected for this rare example of HGT in bordetellae, we compared in vitro and in vivo phenotypes associated with different O-antigen types. Antibodies against O1- and O2-serotypes were poorly cross-reactive, and did not efficiently kill or mediate clearance of alternative O-type bacteria, while a distinct and poorly immunogenic O-antigen offered no protection against colonization. CONCLUSIONS: This study suggests that O-antigen variation was introduced to the classical bordetellae via HGT through recombination. Additionally, genetic variation may be maintained within the O-antigen locus because it can provide escape from immunity to different O-antigen types, potentially allowing for the circulation of different Bordetella strains within the same host population.


Subject(s)
Bordetella/genetics , Gene Transfer, Horizontal , O Antigens/genetics , Animals , Bordetella/immunology , Bordetella Infections/immunology , Bordetella Infections/microbiology , Evolution, Molecular , Genetic Variation , Mice , Mice, Inbred C57BL , O Antigens/immunology , Phylogeny , Recombination, Genetic
3.
PLoS One ; 7(10): e47635, 2012.
Article in English | MEDLINE | ID: mdl-23112828

ABSTRACT

Sensing the environment allows pathogenic bacteria to coordinately regulate gene expression to maximize survival within or outside of a host. Here we show that Bordetella species regulate virulence factor expression in response to carbon dioxide levels that mimic in vivo conditions within the respiratory tract. We found strains of Bordetella bronchiseptica that did not produce adenylate cyclase toxin (ACT) when grown in liquid or solid media with ambient air aeration, but produced ACT and additional antigens when grown in air supplemented to 5% CO(2). Transcriptome analysis and quantitative real time-PCR analysis revealed that strain 761, as well as strain RB50, increased transcription of genes encoding ACT, filamentous hemagglutinin (FHA), pertactin, fimbriae and the type III secretion system in 5% CO(2) conditions, relative to ambient air. Furthermore, transcription of cyaA and fhaB in response to 5% CO(2) was increased even in the absence of BvgS. In vitro analysis also revealed increases in cytotoxicity and adherence when strains were grown in 5% CO(2). The human pathogens B. pertussis and B. parapertussis also increased transcription of several virulence factors when grown in 5% CO(2), indicating that this response is conserved among the classical bordetellae. Together, our data indicate that Bordetella species can sense and respond to physiologically relevant changes in CO(2) concentrations by regulating virulence factors important for colonization, persistence and evasion of the host immune response.


Subject(s)
Bordetella Infections/microbiology , Bordetella/genetics , Bordetella/metabolism , Carbon Dioxide/metabolism , Gene Expression Regulation, Bacterial , Virulence Factors, Bordetella/metabolism , Adenylate Cyclase Toxin/genetics , Adenylate Cyclase Toxin/metabolism , Animals , Bordetella/pathogenicity , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/metabolism , Bordetella bronchiseptica/pathogenicity , Bordetella parapertussis/genetics , Bordetella parapertussis/metabolism , Bordetella parapertussis/pathogenicity , Bordetella pertussis/genetics , Bordetella pertussis/metabolism , Bordetella pertussis/pathogenicity , Cell Line , Gene Expression Profiling , Humans , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Virulence Factors, Bordetella/genetics
4.
PLoS One ; 7(10): e45892, 2012.
Article in English | MEDLINE | ID: mdl-23071529

ABSTRACT

Type VI Secretion Systems (T6SSs) have been identified in numerous gram-negative pathogens, but the lack of a natural host infection model has limited analysis of T6SS contributions to infection and pathogenesis. Here, we describe disruption of a gene within locus encoding a putative T6SS in Bordetella bronchiseptica strain RB50, a respiratory pathogen that circulates in a broad range of mammals, including humans, domestic animals, and mice. The 26 gene locus encoding the B. bronchiseptica T6SS contains apparent orthologs to all known core genes and possesses thirteen novel genes. By generating an in frame deletion of clpV, which encodes a putative ATPase required for some T6SS-dependent protein secretion, we observe that ClpV contributes to in vitro macrophage cytotoxicity while inducing several eukaryotic proteins associated with apoptosis. Additionally, ClpV is required for induction of IL-1ß, IL-6, IL-17, and IL-10 production in J774 macrophages infected with RB50. During infections in wild type mice, we determined that ClpV contributes to altered cytokine production, increased pathology, delayed lower respiratory tract clearance, and long term nasal cavity persistence. Together, these results reveal a natural host infection system in which to interrogate T6SS contributions to immunomodulation and pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Bordetella Infections/immunology , Bordetella bronchiseptica/genetics , Multigene Family/physiology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Animals , Bacterial Proteins/genetics , Bordetella bronchiseptica/pathogenicity , Bordetella bronchiseptica/physiology , Immunomodulation , Interleukin-17/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Macrophages/immunology , Mice , Up-Regulation , Virulence , Virulence Factors/genetics
5.
BMC Microbiol ; 12: 179, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22897969

ABSTRACT

BACKGROUND: The cell envelope of a bacterial pathogen can be damaged by harsh conditions in the environment outside a host and by immune factors during infection. Cell envelope stress responses preserve the integrity of this essential compartment and are often required for virulence. Bordetella species are important respiratory pathogens that possess a large number of putative transcription factors. However, no cell envelope stress responses have been described in these species. Among the putative Bordetella transcription factors are a number of genes belonging to the extracytoplasmic function (ECF) group of alternative sigma factors, some of which are known to mediate cell envelope stress responses in other bacteria. Here we investigate the role of one such gene, sigE, in stress survival and pathogenesis of Bordetella bronchiseptica. RESULTS: We demonstrate that sigE encodes a functional sigma factor that mediates a cell envelope stress response. Mutants of B. bronchiseptica strain RB50 lacking sigE are more sensitive to high temperature, ethanol, and perturbation of the envelope by SDS-EDTA and certain ß-lactam antibiotics. Using a series of immunocompromised mice deficient in different components of the innate and adaptive immune responses, we show that SigE plays an important role in evading the innate immune response during lethal infections of mice lacking B cells and T cells. SigE is not required, however, for colonization of the respiratory tract of immunocompetent mice. The sigE mutant is more efficiently phagocytosed and killed by peripheral blood polymorphonuclear leukocytes (PMNs) than RB50, and exhibits decreased cytotoxicity toward macrophages. These altered interactions with phagocytes could contribute to the defects observed during lethal infection. CONCLUSIONS: Much of the work on transcriptional regulation during infection in B. bronchiseptica has focused on the BvgAS two-component system. This study reveals that the SigE regulon also mediates a discrete subset of functions associated with virulence. SigE is the first cell envelope stress-sensing system to be described in the bordetellae. In addition to its role during lethal infection of mice deficient in adaptive immunity, our results indicate that SigE is likely to be important for survival in the face of stresses encountered in the environment between hosts.


Subject(s)
Bacterial Proteins/metabolism , Bordetella Infections/microbiology , Bordetella bronchiseptica/pathogenicity , Sigma Factor/metabolism , Stress, Physiological , Virulence Factors/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/physiology , Disease Models, Animal , Gene Deletion , Immune Evasion , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Microbial Viability , Molecular Sequence Data , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis , Regulon , Sequence Alignment , Sigma Factor/genetics , Survival Analysis , Virulence , Virulence Factors/genetics
6.
Infect Immun ; 79(1): 527-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974829

ABSTRACT

Interleukin-1 receptor-deficient (IL-1R(-/-)) mice are healthy despite being colonized by commensal microbes but are defective in defenses against specific pathogens, suggesting that IL-1R-mediated effects contribute to immune responses against specific pathogenic mechanisms. To better define the role of IL-1R in immunity to respiratory infections, we challenged IL-1R(-/-) mice with Bordetella pertussis and Bordetella parapertussis, the causative agents of whooping cough. Following inoculation with B. pertussis, but not B. parapertussis, IL-1R(-/-) mice showed elevated bacterial numbers and more extensive inflammatory pathology than wild-type mice. Acellular B. pertussis vaccines were not efficiently protective against B. pertussis in IL-1R(-/-) mice. B. pertussis-stimulated dendritic cells from IL-1R(-/-) mice produced higher levels of tumor necrosis factor alpha (TNF-α) and IL-6 than wild-type cells. Moreover, elevated levels of gamma interferon (IFN-γ) and TNF-α but lower levels of IL-10 were detected during B. pertussis infection in IL-1R(-/-) mice. Since B. parapertussis did not cause severe disease in IL-1R(-/-) mice, we hypothesized that the extreme requirement for IL-1R involves pertussis toxin (Ptx), which is expressed only by B. pertussis. An isogenic Ptx-deficient B. pertussis strain had only a modest phenotype in wild-type mice but was completely defective in causing lethal disease in IL-1R(-/-) mice, indicating that the particular virulence of B. pertussis in these mice requires Ptx. Ptx contributes to IL-1ß induction by B. pertussis, which is involved in IL-10 induction through IL-1R signaling. IL-10 treatment reduced B. pertussis numbers in IL-1R(-/-) mice, suggesting that the lower IL-10 responses partially account for the uncontrolled inflammation and bacterial growth in these mice.


Subject(s)
Bordetella pertussis/immunology , Pertussis Toxin/toxicity , Receptors, Interleukin-1 Type I/metabolism , Animals , Bordetella parapertussis/immunology , Cells, Cultured , Gene Expression Regulation , Interleukin-1/genetics , Interleukin-1/metabolism , Macrophages/physiology , Mice , Mice, Knockout , Receptors, Interleukin-1 Type I/genetics , Signal Transduction , Time
7.
J Immunol ; 184(3): 1392-400, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042578

ABSTRACT

Bordetella parapertussis causes the prolonged coughing illness known as pertussis or whooping cough, persisting for weeks within the respiratory tracts of infected hosts but inducing a very poor T cell response relative to that induced by Bordetella pertussis, the more common cause of pertussis. In this study, we examine the contributions of cytokines involved in the clearance of B. parapertussis and immunomodulation that delays effective clearance. The slow elimination of this pathogen from the respiratory tracts of mice coincides with the gradual accumulation of CD4(+) T cells in the lungs and B. parapertussis-responsive IFN-gamma-producing cells in the spleen. IFN-gamma-deficient mice were defective in the accumulation of leukocytes in lungs and in clearance of B. parapertussis from the lungs. In vitro B. parapertussis-stimulated macrophages produced IL-10, which inhibited the generation of the IFN-gamma response that is required for protection in vivo. As compared with wild-type mice, IL-10-deficient mice produced significantly higher levels of IFN-gamma, had higher numbers of leukocytes accumulated in the lungs, and cleared B. parapertussis more rapidly. Together, these data indicate that B. parapertussis induces the production of IL-10, which facilitates its persistence within infected hosts by limiting a protective IFN-gamma response.


Subject(s)
Bordetella parapertussis/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Animals , Bordetella Infections/immunology , Bordetella Infections/microbiology , Bordetella Infections/pathology , Bordetella parapertussis/growth & development , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , Cell Line, Transformed , Cell Migration Inhibition/genetics , Cell Migration Inhibition/immunology , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interferon-gamma/deficiency , Interferon-gamma/physiology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/physiology , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
8.
J Bacteriol ; 190(15): 5502-11, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18556799

ABSTRACT

Bordetella bronchiseptica is a gram-negative respiratory pathogen that infects a wide range of hosts and causes a diverse spectrum of disease. This diversity is likely affected by multiple factors, such as host immune status, polymicrobial infection, and strain diversity. In a murine model of infection, we found that the virulence of B. bronchiseptica strains, as measured by the mean lethal dose, varied widely. Strain 253 was less virulent than the typically studied strain, RB50. Transcriptome analysis showed that cyaA, the gene encoding adenylate cyclase toxin (CyaA), was the most downregulated transcript identified in strain 253 compared to that in strain RB50. Comparative genomic hybridization and genome sequencing of strain 253 revealed that the cya locus, which encodes, activates, and secretes CyaA, was replaced by an operon (ptp) predicted to encode peptide transport proteins. Other B. bronchiseptica strains from the same phylogenetic lineage as that of strain 253 also lacked the cya locus, contained the ptp genes, and were less virulent than strain RB50. Although the loss of CyaA would be expected to be counterselected since it is conserved among the classical bordetellae and believed to be important to their success, our data indicate that the loss of this toxin and the gain of the ptp genes occurred in an ancestral strain that then expanded into a lineage. This suggests that there may be ecological niches in which CyaA is not critical for the success of B. bronchiseptica.


Subject(s)
Adenylate Cyclase Toxin/genetics , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/pathogenicity , Animals , Bordetella Infections/microbiology , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Evolution, Molecular , Gene Expression Profiling , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Operon , Sequence Analysis, DNA , Survival Analysis , Synteny , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...