Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 17535, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34475490

ABSTRACT

Cystic fibrosis (CF) is an autosomal recessive disease caused by dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) protein, and is marked by an accumulation of mucus in affected airways resulting in persistent infection and chronic inflammation. Quantitative differences in inflammatory markers have been observed in CF patient serum, tracheal cells, and bronchoalveolar lavage fluid, in the absence of detectable infection, implying that absent CFTR function alone may result in dysregulated immune responses. To examine the relationship between absent CFTR and systemic inflammation, 22 analytes were measured in CF mice (F508del/F508del) sera using the MSD multiplex platform. Pro-inflammatory cytokines IL-2, TNF-α, IL-17α, IFN-γ, IL-1ß, and MIP-3α are significantly elevated in infection-naïve CF mice (p < 0.050). Anti-inflammatory cytokines IL-10 and IL-4 are also significantly increased (p = 0.00003, p = 0.004). Additionally, six general markers of inflammation are significantly different from non-CF controls (p < 0.050). To elucidate the effects of chronic infection on the CF inflammatory profile, we examined CF mice exposed to spontaneous Bordetella pseudohinzii infections. There are no statistical differences in nearly all inflammatory markers when compared to their infection-naïve CF counterparts, except in the Th2-derived IL-4 and IL-5 which demonstrate significant decreases following exposure (p = 0.046, p = 0.045). Lastly, following acute infection, CF mice demonstrate elevations in nearly all inflammatory markers, but exhibit a shortened return to uninfected levels over time, and suppression of Th1-derived IL-2 and IL-5 (p = 0.043, p = 0.011). These results imply that CF mice have a persistent inflammatory profile often indistinguishable from chronic infection, and a dysregulated humoral response during and following active infection.


Subject(s)
Bordetella Infections/complications , Bordetella/isolation & purification , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/microbiology , Cytokines/blood , Inflammation/diagnosis , Mutation , Animals , Bordetella Infections/metabolism , Bordetella Infections/microbiology , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Disease Models, Animal , Female , Inflammation/blood , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL
2.
PLoS Pathog ; 17(8): e1009735, 2021 08.
Article in English | MEDLINE | ID: mdl-34347835

ABSTRACT

Whooping cough is resurging in the United States despite high vaccine coverage. The rapid rise of Bordetella pertussis isolates lacking pertactin (PRN), a key vaccine antigen, has led to concerns about vaccine-driven evolution. Previous studies showed that pertactin can mediate binding to mammalian cells in vitro and act as an immunomodulatory factor in resisting neutrophil-mediated clearance. To further investigate the role of PRN in vivo, we examined the functions of pertactin in the context of a more naturally low dose inoculation experimental system using C3H/HeJ mice that is more sensitive to effects on colonization, growth and spread within the respiratory tract, as well as an experimental approach to measure shedding and transmission between hosts. A B. bronchiseptica pertactin deletion mutant was found to behave similarly to its wild-type (WT) parental strain in colonization of the nasal cavity, trachea, and lungs of mice. However, the pertactin-deficient strain was shed from the nares of mice in much lower numbers, resulting in a significantly lower rate of transmission between hosts. Histological examination of respiratory epithelia revealed that pertactin-deficient bacteria induced substantially less inflammation and mucus accumulation than the WT strain and in vitro assays verified the effect of PRN on the induction of TNF-α by murine macrophages. Interestingly, only WT B. bronchiseptica could be recovered from the spleen of infected mice and were further observed to be intracellular among isolated splenocytes, indicating that pertactin contributes to systemic dissemination involving intracellular survival. These results suggest that pertactin can mediate interactions with immune cells and augments inflammation that contributes to bacterial shedding and transmission between hosts. Understanding the relative contributions of various factors to inflammation, mucus production, shedding and transmission will guide novel strategies to interfere with the reemergence of pertussis.


Subject(s)
Alveolar Epithelial Cells/microbiology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Shedding , Bordetella Infections/transmission , Bordetella bronchiseptica/pathogenicity , Inflammation/pathology , Virulence Factors, Bordetella/metabolism , Animals , Bacterial Adhesion , Bacterial Outer Membrane Proteins/genetics , Bordetella Infections/metabolism , Bordetella Infections/microbiology , Female , Humans , Inflammation/metabolism , Inflammation/microbiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Virulence Factors, Bordetella/genetics
3.
Sci Rep ; 11(1): 9373, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931696

ABSTRACT

Secretion of pertussis toxin (PT) is the preeminent virulence trait of the human pathogen Bordetella pertussis, causing whooping cough. Bordetella bronchiseptica, although it harbors an intact 12-kb ptx-ptl operon, does not express PT due to an inactive ptx promoter (Pptx), which contains 18 SNPs (single nucleotide polymorphisms) relative to B. pertussis Pptx. A systematic analysis of these SNPs was undertaken to define the degree of mutational divergence necessary to activate B. bronchiseptica Pptx. A single change (C-13T), which created a better - 10 element, was capable of activating B. bronchiseptica Pptx sufficiently to allow secretion of low but measureable levels of active PT. Three additional changes in the BvgA-binding region, only in the context of C-13T mutant, raised the expression of PT to B. pertussis levels. These results illuminate a logical evolutionary pathway for acquisition of this key virulence trait in the evolution of B. pertussis from a B. bronchiseptica-like common ancestor.


Subject(s)
Bacterial Proteins/genetics , Bordetella Infections/metabolism , Bordetella bronchiseptica/physiology , Gene Expression Regulation, Bacterial , Mutation , Pertussis Toxin/metabolism , Promoter Regions, Genetic , Amino Acid Sequence , Bordetella Infections/microbiology , Bordetella Infections/pathology , Evolution, Molecular , Pertussis Toxin/genetics , Sequence Homology
4.
J Bacteriol ; 201(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31209073

ABSTRACT

Bacteria can be motile and planktonic or, alternatively, sessile and participating in the biofilm mode of growth. The transition between these lifestyles can be regulated by a second messenger, cyclic dimeric GMP (c-di-GMP). High intracellular c-di-GMP concentration correlates with biofilm formation and motility inhibition in most bacteria, including Bordetella bronchiseptica, which causes respiratory tract infections in mammals and forms biofilms in infected mice. We previously described the diguanylate cyclase BdcA as involved in c-di-GMP synthesis and motility regulation in B. bronchiseptica; here, we further describe the mechanism whereby BdcA is able to regulate motility and biofilm formation. Amino acid replacement of GGDEF with GGAAF in BdcA is consistent with the conclusion that diguanylate cyclase activity is necessary for biofilm formation and motility regulation, although we were unable to confirm the stability of the mutant protein. In the absence of the bdcA gene, B. bronchiseptica showed enhanced motility, strengthening the hypothesis that BdcA regulates motility in B. bronchiseptica We showed that c-di-GMP-mediated motility inhibition involved regulation of flagellin expression, as high c-di-GMP levels achieved by expressing BdcA significantly reduced the level of flagellin protein. We also demonstrated that protein BB2109 is necessary for BdcA activity, motility inhibition, and biofilm formation. Finally, absence of the bdcA gene affected bacterial infection, implicating BdcA-regulated functions as important for bacterium-host interactions. This work supports the role of c-di-GMP in biofilm formation and motility regulation in B. bronchiseptica, as well as its impact on pathogenesis.IMPORTANCE Pathogenesis of Bordetella spp., like that of a number of other pathogens, involves biofilm formation. Biofilms increase tolerance to biotic and abiotic factors and are proposed as reservoirs of microbes for transmission to other organs (trachea, lungs) or other hosts. Bis-(3'-5')-cyclic dimeric GMP (c-di-GMP) is a second messenger that regulates transition between biofilm and planktonic lifestyles. In Bordetella bronchiseptica, high c-di-GMP levels inhibit motility and favor biofilm formation. In the present work, we characterized a B. bronchiseptica diguanylate cyclase, BdcA, which regulates motility and biofilm formation and affects the ability of B. bronchiseptica to colonize the murine respiratory tract. These results provide us with a better understanding of how B. bronchiseptica can infect a host.


Subject(s)
Bacterial Proteins/metabolism , Bordetella Infections/metabolism , Bordetella Infections/microbiology , Bordetella bronchiseptica/enzymology , Escherichia coli Proteins/metabolism , Phosphorus-Oxygen Lyases/metabolism , Respiratory Tract Infections/microbiology , Animals , Bacterial Proteins/genetics , Bordetella Infections/genetics , Bordetella bronchiseptica/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Mice , Mice, Inbred C57BL , Movement , Phosphorus-Oxygen Lyases/genetics
5.
J R Soc Interface ; 10(80): 20120588, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23256186

ABSTRACT

Co-infection by multiple parasites is common within individuals. Interactions between co-infecting parasites include resource competition, direct competition and immune-mediated interactions and each are likely to alter the dynamics of single parasites. We posit that co-infection is a driver of variation in parasite establishment and growth, ultimately altering the production of parasite transmission stages. To test this hypothesis, three different treatment groups of laboratory mice were infected with the gastrointestinal helminth Heligmosomoides polygyrus, the respiratory bacterial pathogen Bordetella bronchiseptica lux(+) or co-infected with both parasites. To follow co-infection simultaneously, self-bioluminescent bacteria were used to quantify infection in vivo and in real-time, while helminth egg production was monitored in real-time using faecal samples. Co-infection resulted in high bacterial loads early in the infection (within the first 5 days) that could cause host mortality. Co-infection also produced helminth 'super-shedders'; individuals that chronically shed the helminth eggs in larger than average numbers. Our study shows that co-infection may be one of the underlying mechanisms for the often-observed high variance in parasite load and shedding rates, and should thus be taken into consideration for disease management and control. Further, using self-bioluminescent bacterial reporters allowed quantification of the progression of infection within the whole animal of the same individuals at a fine temporal scale (daily) and significantly reduced the number of animals used (by 85%) compared with experiments that do not use in vivo techniques. Thus, we present bioluminescent imaging as a novel, non-invasive tool offering great potential to be taken forward into other applications of infectious disease ecology.


Subject(s)
Bordetella Infections/metabolism , Bordetella bronchiseptica/metabolism , Coinfection/metabolism , Nematospiroides dubius/metabolism , Strongylida Infections/metabolism , Animals , Female , Mice , Mice, Inbred BALB C , Nematospiroides dubius/microbiology , Ovum/metabolism
6.
Infect Immun ; 80(12): 4317-32, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23027529

ABSTRACT

Throughout infection, pathogenic bacteria induce dramatic changes in host transcriptional repertoires. An understanding of how bacterial factors influence host reprogramming will provide insight into disease pathogenesis. In the human respiratory pathogen Bordetella pertussis, the causative agent of whooping cough, pertussis toxin (PT) is a key virulence factor that promotes colonization, suppresses innate immune responses during early infection, and causes systemic disease symptoms. To determine the full extent of PT-associated gene regulation in the airways through the peak of infection, we measured global transcriptional profiles in the lungs of BALB/c mice infected with wild-type (WT) or PT-deficient (ΔPT) B. pertussis. ΔPT bacteria were inoculated at a dose equivalent to the WT dose and at a high dose (ΔPT(high)) to distinguish effects caused by higher bacterial loads achieved in WT infection from effects associated with PT. The results demonstrated that PT was associated with a significant upregulation of immune and inflammatory response genes as well as several other genes implicated in airway pathology. In contrast to the early, transient responses observed for ΔPT(high) infection, WT infection induced a prolonged expression of inflammatory genes and increased the extent and duration of lung histopathology. In addition, the administration of purified PT to ΔPT(high)-infected mice 1 day after bacterial inoculation exacerbated and prolonged inflammatory responses and airway pathology. These data indicate that PT not only is associated with exacerbated host airway responses during peak B. pertussis infection but also may inhibit host mechanisms of attenuating and resolving inflammation in the airways, suggesting possible links between PT and pertussis disease symptoms.


Subject(s)
Bordetella Infections/physiopathology , Bordetella pertussis/pathogenicity , Gene Expression Regulation/drug effects , Inflammation/immunology , Lung/immunology , Pertussis Toxin/immunology , Whooping Cough/physiopathology , Animals , Bordetella Infections/immunology , Bordetella Infections/metabolism , Bordetella Infections/pathology , Bordetella pertussis/immunology , Female , Gene Expression Profiling , Humans , Immunity, Innate , Inflammation/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pertussis Toxin/pharmacology , Real-Time Polymerase Chain Reaction , Whooping Cough/immunology , Whooping Cough/microbiology , Whooping Cough/pathology
7.
Curr Top Microbiol Immunol ; 361: 113-29, 2012.
Article in English | MEDLINE | ID: mdl-22411430

ABSTRACT

Atrophic rhinitis is a widespread and economically important swine disease caused by Pasteurella multocida and Bordetella bronchiseptica. The disease is characterized by atrophy of the nasal turbinate bones, which results in a shortened and deformed snout in severe cases. P. multocida toxin and B. bronchiseptica dermonecrotic toxin have been considered to independently or cooperatively disturb the osteogenesis of the turbinate bone by inhibiting osteoblastic differentiation and/or stimulating bone resorption by osteoclasts. Recently, the intracellular targets and molecular actions of both toxins have been clarified, enabling speculation on the intracellular signals leading to the inhibition of osteogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Bordetella Infections/metabolism , Bordetella bronchiseptica/metabolism , Pasteurella multocida/metabolism , Rhinitis, Atrophic/metabolism , Swine Diseases/metabolism , Transglutaminases/metabolism , Virulence Factors, Bordetella/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bone Resorption/microbiology , Bone Resorption/pathology , Bordetella Infections/genetics , Bordetella Infections/microbiology , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/pathogenicity , Coinfection , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Pasteurella multocida/genetics , Pasteurella multocida/pathogenicity , Rhinitis, Atrophic/genetics , Rhinitis, Atrophic/microbiology , Signal Transduction , Swine , Swine Diseases/microbiology , Swine Diseases/pathology , Transglutaminases/chemistry , Transglutaminases/genetics , Turbinates/microbiology , Turbinates/pathology , Virulence Factors, Bordetella/chemistry , Virulence Factors, Bordetella/genetics
8.
Mol Microbiol ; 84(3): 446-62, 2012 May.
Article in English | MEDLINE | ID: mdl-22458330

ABSTRACT

Bordetella bronchiseptica is a pathogen that can acquire iron using its native alcaligin siderophore system, but can also use the catechol xenosiderophore enterobactin via the BfeA outer membrane receptor. Transcription of bfeA is positively controlled by a regulator that requires induction by enterobactin. Catecholamine hormones also induce bfeA transcription and B. bronchiseptica can use the catecholamine noradrenaline for growth on transferrin. In this study, B. bronchiseptica was shown to use catecholamines to obtain iron from both transferrin and lactoferrin in the absence of siderophore. In the presence of siderophore, noradrenaline augmented transferrin utilization by B. bronchiseptica, as well as siderophore function in vitro. Genetic analysis identified BfrA, BfrD and BfrE as TonB-dependent outer membrane catecholamine receptors. The BfeA enterobactin receptor was found to not be involved directly in catecholamine utilization; however, the BfrA, BfrD and BfrE catecholamine receptors could serve as receptors for enterobactin and its degradation product 2,3-dihydroxybenzoic acid. Thus, there is a functional link between enterobactin-dependent and catecholamine-dependent transferrin utilization. This investigation characterizes a new B. bronchiseptica mechanism for iron uptake from transferrin that uses host stress hormones that not only deliver iron directly to catecholamine receptors, but also potentiate siderophore activity by acting as iron shuttles.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bordetella Infections/metabolism , Bordetella bronchiseptica/metabolism , Catecholamines/metabolism , Iron/metabolism , Receptors, Catecholamine/metabolism , Transferrin/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella Infections/microbiology , Bordetella bronchiseptica/genetics , Hormones/metabolism , Host-Pathogen Interactions , Humans , Lactoferrin/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Catecholamine/genetics , Siderophores/metabolism
9.
PLoS One ; 7(12): e52903, 2012.
Article in English | MEDLINE | ID: mdl-23300813

ABSTRACT

Bordetella pertussis (B. pertussis) is the causative agent of whooping cough, a respiratory disease that is reemerging worldwide. Mechanisms of selective lymphocyte trafficking to the airways are likely to be critical in the immune response to this pathogen. We compared murine infection by B. pertussis, B. parapertussis, and a pertussis toxin-deleted B. pertussis mutant (BpΔPTX) to test the hypothesis that effector memory T-helper cells (emTh) display an altered pattern of trafficking receptor expression in B. pertussis infection due to a defect in imprinting. Increased cell recruitment to the lungs at 5 days post infection (p.i.) with B. parapertussis, and to a lesser extent with BpΔPTX, coincided with an increased frequency of circulating emTh cells expressing the mucosal-associated trafficking receptors α4ß7 and α4ß1 while a reduced population of these cells was observed in B. pertussis infection. These cells were highly evident in the blood and lungs in B. pertussis infection only at 25 days p.i. when B. parapertussis and BpΔPTX infections were resolved. Although at 5 days p.i., an equally high percentage of lung dendritic cells (DCs) from all infections expressed maturation markers, this expression persisted only in B. pertussis infection at 25 days p.i. Furthermore, at 5 days p.i with B. pertussis, lung DCs migration to draining lymph nodes may be compromised as evidenced by decreased frequency of CCR7(+) DCs, inhibited CCR7-mediated in vitro migration, and fewer DCs in lung draining lymph nodes. Lastly, a reduced frequency of allogeneic CD4(+) cells expressing α4ß1 was detected following co-culture with lung DCs from B. pertussis-infected mice, suggesting a defect in DC imprinting in comparison to the other infection groups. The findings in this study suggest that B. pertussis may interfere with imprinting of lung-associated trafficking receptors on T lymphocytes leading to extended survival in the host and a prolonged course of disease.


Subject(s)
Bordetella Infections/metabolism , Bordetella pertussis , Integrin alpha4/metabolism , Lung/metabolism , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Bordetella Infections/immunology , Bordetella Infections/pathology , Bordetella parapertussis , Cell Adhesion/immunology , Cell Movement/immunology , Female , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Helper-Inducer/pathology
10.
BMC Microbiol ; 10: 247, 2010 Sep 25.
Article in English | MEDLINE | ID: mdl-20868510

ABSTRACT

BACKGROUND: Bordetella dermonecrotic toxin (DNT) causes the turbinate atrophy in swine atrophic rhinitis, caused by a Bordetella bronchiseptica infection of pigs, by inhibiting osteoblastic differentiation. The toxin is not actively secreted from the bacteria, and is presumed to be present in only small amounts in infected areas. How such small amounts can affect target tissues is unknown. RESULTS: Fluorescence microscopy revealed that DNT associated with a fibrillar structure developed on cultured cells. A cellular component cross-linked with DNT conjugated with a cross-linker was identified as fibronectin by mass spectrometry. Colocalization of the fibronectin network on the cells with DNT was also observed by fluorescence microscope. Several lines of evidence suggested that DNT interacts with fibronectin not directly, but through another cellular component that remains to be identified. The colocalization was observed in not only DNT-sensitive cells but also insensitive cells, indicating that the fibronectin network neither serves as a receptor for the toxin nor is involved in the intoxicating procedures. The fibronectin network-associated toxin was easily liberated when the concentration of toxin in the local environment decreased, and was still active. CONCLUSIONS: Components in the extracellular matrix are known to regulate activities of various growth factors by binding and liberating them in response to alterations in the extracellular environment. Similarly, the fibronectin-based extracellular matrix may function as a temporary storage system for DNT, enabling small amounts of the toxin to efficiently affect target tissues or cells.


Subject(s)
Bordetella/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Transglutaminases/metabolism , Virulence Factors, Bordetella/metabolism , Animals , Bordetella Infections/metabolism , Bordetella Infections/microbiology , Bordetella Infections/pathology , Cell Line , Fibronectins/metabolism , Humans , Mice , Rhinitis, Atrophic/metabolism , Rhinitis, Atrophic/microbiology , Rhinitis, Atrophic/pathology
11.
Cell Microbiol ; 11(12): 1735-49, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19650828

ABSTRACT

The Bordetella type III secretion system (T3SS) effector protein BteA is necessary and sufficient for rapid cytotoxicity in a wide range of mammalian cells. We show that BteA is highly conserved and functionally interchangeable between Bordetella bronchiseptica, Bordetella pertussis and Bordetella parapertussis. The identification of BteA sequences required for cytotoxicity allowed the construction of non-cytotoxic mutants for localization studies. BteA derivatives were targeted to lipid rafts and showed clear colocalization with cortical actin, ezrin and the lipid raft marker GM1. We hypothesized that BteA associates with the cytoplasmic face of lipid rafts to locally modulate host cell responses to Bordetella attachment. B. bronchiseptica adhered to host cells almost exclusively to GM1-enriched lipid raft microdomains and BteA colocalized to these same sites following T3SS-mediated translocation. Disruption of lipid rafts with methyl-beta-cyclodextrin protected cells from T3SS-induced cytotoxicity. Localization to lipid rafts was mediated by a 130-amino-acid lipid raft targeting domain at the N-terminus of BteA, and homologous domains were identified in virulence factors from other bacterial species. Lipid raft targeting sequences from a T3SS effector (Plu4750) and an RTX-type toxin (Plu3217) from Photorhabdus luminescens directed fusion proteins to lipid rafts in a manner identical to the N-terminus of BteA.


Subject(s)
Amino Acid Motifs , Bacterial Proteins/chemistry , Bordetella Infections/metabolism , Bordetella/metabolism , Membrane Microdomains/metabolism , Secretory Pathway , Virulence Factors, Bordetella/metabolism , Amino Acid Sequence , Animals , Bacterial Adhesion/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella/drug effects , Bordetella/genetics , Bordetella Infections/microbiology , Cell Line , Cytoskeletal Proteins/metabolism , Host-Pathogen Interactions , Humans , Membrane Microdomains/drug effects , Mice , Molecular Sequence Data , Rats , beta-Cyclodextrins/pharmacology
12.
Biometals ; 22(1): 33-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19130264

ABSTRACT

The bacterial respiratory pathogens Bordetella pertussis and Bordetella bronchiseptica employ multiple alternative iron acquisition pathways to adapt to changes in the mammalian host environment during infection. The alcaligin, enterobactin, and heme utilization pathways are differentially expressed in response to the cognate iron source availability by a mechanism involving substrate-inducible positive regulators. As inducers, the iron sources function as chemical signals termed ferrimones. Ferrimone-sensing allows the pathogen to adapt and exploit early and late events in the infection process.


Subject(s)
Bacterial Proteins/metabolism , Bordetella/metabolism , Iron/metabolism , Signal Transduction/physiology , Animals , Bacterial Proteins/genetics , Bordetella/genetics , Bordetella/pathogenicity , Bordetella Infections/metabolism , Enterobactin/chemistry , Enterobactin/metabolism , Gene Expression Regulation, Bacterial , Heme/genetics , Heme/metabolism , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/metabolism , Siderophores/chemistry , Siderophores/metabolism
13.
PLoS One ; 4(1): e4280, 2009.
Article in English | MEDLINE | ID: mdl-19169359

ABSTRACT

The recognition of bacterial lipopolysaccharide (LPS) by host Toll-like receptor (TLR)4 is a crucial step in developing protective immunity against several gram negative bacterial pathogens. Bordetella bronchiseptica and B. pertussis stimulate robust TLR4 responses that are required to control the infection, but a close relative, B. parapertussis, poorly stimulates this receptor, and TLR4 deficiency does not affect its course of infection. This led us to hypothesize that inefficient TLR4 stimulation enables B. parapertussis to evade host immunity. In a mouse model of infection, B. parapertussis grew rapidly in the lungs, but no measurable increase in TLR4-mediated cytokine, chemokine, or leukocyte responses were observed over the first few days of infection. Delivery of a TLR4 stimulant in the inoculum resulted in a robust inflammatory response and a 10- to 100-fold reduction of B. parapertussis numbers. As we have previously shown, B. parapertussis grows efficiently during the first week of infection even in animals passively immunized with antibodies. We show that this evasion of antibody-mediated clearance is dependent on the lack of TLR4 stimulation by B. parapertussis as co-inoculation with a TLR4 agonist resulted in 10,000-fold lower B. parapertussis numbers on day 3 in antibody-treated wild type, but not TLR4-deficient, mice. Together, these results indicate that inefficient TLR4 stimulation by B. parapertussis enables it to avoid host immunity and grow to high numbers in the respiratory tract of naïve and immunized hosts.


Subject(s)
Bordetella Infections/pathology , Bordetella parapertussis/metabolism , Toll-Like Receptor 4/metabolism , Animals , Bordetella Infections/metabolism , Chemokines/metabolism , Cytokines/metabolism , Immune System , Inflammation , Leukocytes/metabolism , Lipopolysaccharides/metabolism , Lung/microbiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Models, Biological
14.
Mol Microbiol ; 71(2): 492-504, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19040642

ABSTRACT

Type III secretion system (T3SS) tip complexes serve as adaptors that bridge the T3SS needle and the pore-forming translocation apparatus. In this report we demonstrate that Bsp22, the most abundantly secreted substrate of the Bordetella T3SS, self-polymerizes to form the Bordetella bronchiseptica tip complex. Bsp22 is required for both T3SS-mediated cytotoxicity against eukaryotic cells and haemoglobin release from erythrocytes. Bacterial two-hybrid analysis and protein pull-down assays demonstrated the ability of Bsp22 to associate with itself and to bind BopD, a component of the Bordetella translocation pore. Immunoblot and cross-linking analysis of secreted proteins or purified Bsp22 showed extensive multimerization which was shown by transmission electron microscopy to lead to the formation of variable length flexible filaments. Immunoelectron microscopy revealed Bsp22 filaments on the surface of bacterial cells. Given its required role in secretion and cell-surface exposure, we tested the protective effects of antibodies against Bsp22 in vitro and in vivo. Polyclonal antisera against Bsp22 fully protected epithelial cells from T3SS-dependent killing and immunization with Bsp22 protected mice against Bordetella infection. Of the approximately 30 genes which encode the Bordetella T3SS apparatus, bsp22 is the only one without characterized orthologues in other well-characterized T3SS loci. A maximum likelihood phylogenetic analysis indicated that Bsp22 defines a new subfamily of T3SS tip complex proteins. Given its immunogenic and immunoprotective properties and high degree of conservation among Bordetella species, Bsp22 and its homologues may prove useful for diagnostics and next-generation subunit vaccines.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bordetella bronchiseptica/metabolism , Animals , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Bordetella Infections/immunology , Bordetella Infections/metabolism , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/immunology , Cells, Cultured , Epithelial Cells/microbiology , Erythrocytes/microbiology , Gene Expression Regulation, Bacterial , Hemoglobins/metabolism , Mice , Mice, Inbred C57BL , Phylogeny , Protein Multimerization , Sequence Alignment
15.
J Biol Chem ; 281(10): 6589-600, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16407269

ABSTRACT

In Bordetella bronchiseptica, the functional type III secretion system (TTSS) is required for the induction of necrotic cell death in infected mammalian cells. To identify the factor(s) involved in necrotic cell death, type III-secreted proteins from B. bronchiseptica were analyzed using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and electrospray ionization tandem mass spectrometry. We identified a 69-kDa secreted protein designated BopC. The gene encoding BopC is located outside of the TTSS locus and is also highly conserved in both Bordetella parapertussis and Bordetella pertussis. The results of a lactate dehydrogenase release assay and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling assay demonstrated that BopC is required for necrotic cell death. It has been reported that tyrosine-phosphorylated proteins (PY) of host cells are dephosphorylated during B. bronchiseptica infection in a TTSS-dependent manner. We found that BopC is also involved in PY dephosphorylation in infected host cells. It appears that the necrotic cell death triggered by BopC occurs prior to the PY reduction in host cells, because Bordetella-induced cell death was not affected even in the presence of a dephosphorylation inhibitor. Furthermore, a translocation assay showed that the signal sequence for both secretion into culture supernatant and translocation into the host cell is located in 48 amino acid residues of the BopC N terminus. This report reveals for the first time that a novel type III effector, BopC, is required for the induction of necrotic cell death during Bordetella infection.


Subject(s)
Bacterial Proteins/physiology , Bordetella Infections/metabolism , Bordetella bronchiseptica/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella Infections/pathology , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , HeLa Cells , Humans , Necrosis , Phosphorylation , Tyrosine/metabolism
16.
J Clin Invest ; 115(12): 3594-601, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16294220

ABSTRACT

Whooping cough is considered a childhood disease, although there is growing evidence that children are infected by adult carriers. Additionally, increasing numbers of vaccinated adults are being diagnosed with Bordetella pertussis disease. Thus it is critical to understand how B. pertussis remains endemic even in highly vaccinated or immune populations. Here we used the mouse model to examine the nature of sterilizing immunity to B. pertussis. Antibodies were necessary to control infection but did not rapidly clear B. pertussis from the lungs. However, antibodies affected B. pertussis after a delay of at least a week by a mechanism that involved neutrophils and Fc receptors, suggesting that neutrophils phagocytose and clear antibody-opsonized bacteria via Fc receptors. B. pertussis blocked migration of neutrophils and inhibited their recruitment to the lungs during the first week of infection by a pertussis toxin-dependent (PTx-dependent) mechanism; a PTx mutant of B. pertussis induced rapid neutrophil recruitment and was rapidly cleared from the lungs by adoptively transferred antibodies. Depletion of neutrophils abrogated the defects of the PTx mutant. Together these results indicate that PTx inhibits neutrophil recruitment, which consequently allows B. pertussis to avoid rapid antibody-mediated clearance and therefore successfully infect immune hosts.


Subject(s)
Bordetella pertussis/metabolism , Neutrophils/metabolism , Neutrophils/microbiology , Pertussis Toxin/pharmacology , Animals , Antigens, Bacterial/chemistry , Aorta/cytology , Bordetella Infections/metabolism , Cell Movement , Chemokines/metabolism , Cytokines/metabolism , Endothelium, Vascular/cytology , Humans , Leukocytes/cytology , Leukocytes/microbiology , Lung/microbiology , Mice , Mice, Inbred C57BL , Mutation , Neutrophils/cytology , Neutrophils/drug effects , Pertussis Toxin/metabolism , Phagocytosis , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Time Factors , Virulence Factors, Bordetella/metabolism , Whooping Cough/microbiology , Whooping Cough/therapy
17.
Infect Immun ; 73(12): 8144-52, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16299309

ABSTRACT

Bordetella pertussis, B. parapertussis, and B. bronchiseptica are closely related species associated with respiratory disease in humans and other mammals. While B. bronchiseptica has a wide host range, B. pertussis and B. parapertussis evolved separately from a B. bronchiseptica-like progenitor to naturally infect only humans. Despite very different doubling times in vitro, all three establish similar levels of infection in the mouse lung within 72 h. Recent work has revealed separate roles for Toll-like receptor 4 (TLR4) in immunity to B. pertussis and B. bronchiseptica, while no role for TLR4 during B. parapertussis infection has been described. Here we compared the requirement for TLR4 in innate host defense to these organisms using the same mouse infection model. While B. bronchiseptica causes lethal disease in TLR4-deficient mice, B. pertussis and B. parapertussis do not. Correspondingly, TLR4 is critical in limiting B. bronchiseptica but not B. pertussis or B. parapertussis bacterial numbers during the first 72 h. Interestingly, B. bronchiseptica induces a TLR4-dependent cytokine response that is considerably larger than that induced by B. pertussis or B. parapertussis. Analysis of their endotoxins using RAW cells suggests that B. bronchiseptica lipopolysaccharide (LPS) is 10- and 100-fold more stimulatory than B. pertussis or B. parapertussis LPS, respectively. The difference in LPS stimulus is more pronounced when using HEK293 cells expressing human TLR4. Thus, it appears that in adapting to infect humans, B. pertussis and B. parapertussis independently modified their LPS to reduce TLR4-mediated responses, which may compensate for slower growth rates and facilitate host colonization.


Subject(s)
Bordetella Infections/immunology , Pneumonia, Bacterial/immunology , Toll-Like Receptor 4/physiology , Animals , Bordetella Infections/metabolism , Bordetella Infections/microbiology , Bordetella bronchiseptica , Bordetella parapertussis , Bordetella pertussis , Cells, Cultured , Cytokines/metabolism , Humans , Immunity, Innate/immunology , Lipopolysaccharides/pharmacology , Lung/immunology , Lung/microbiology , Macrophages/drug effects , Mice , Mice, Inbred Strains , Neutrophils/immunology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
J Immunol ; 175(7): 4647-52, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16177111

ABSTRACT

Chronic bacterial infection reflects a balance between the host immune response and bacterial factors that promote colonization and immune evasion. Bordetella bronchiseptica uses a type III secretion system (TTSS) to persist in the lower respiratory tract of mice. We hypothesize that colonization is facilitated by bacteria-driven modulation of dendritic cells (DCs), which leads to an immunosuppressive adaptive host response. Migration of DCs to the draining lymph nodes of the respiratory tract was significantly increased in mice infected with wild-type B. bronchiseptica compared with mice infected with TTSS mutant bacteria. Reduced colonization by TTSS-deficient bacteria was evident by 7 days after infection, whereas colonization by wild-type bacteria remained high. This decrease in colonization correlated with peak IFN-gamma production by restimulated splenocytes from infected animals. Wild-type bacteria also elicited peak IFN-gamma production on day 7, but the quantity was significantly lower than that elicited by TTSS mutant bacteria. Additionally, wild-type bacteria elicited higher levels of the immunosuppressive cytokine IL-10 compared with the TTSS mutant bacteria. B. bronchiseptica colonization in IL-10(-/-) mice was significantly reduced compared with infections in wild-type mice. These findings suggest that B. bronchiseptica use the TTSS to rapidly drive respiratory DCs to secondary lymphoid tissues where these APCs stimulate an immunosuppressive response characterized by increased IL-10 and decreased IFN-gamma production that favors bacterial persistence.


Subject(s)
Bordetella Infections/immunology , Bordetella Infections/microbiology , Bordetella bronchiseptica/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Immunosuppression Therapy , Animals , Bacterial Proteins/physiology , Bordetella Infections/metabolism , Bordetella bronchiseptica/growth & development , Bordetella bronchiseptica/pathogenicity , Dendritic Cells/cytology , Down-Regulation/immunology , Interferon Type I/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/microbiology , Up-Regulation/immunology
19.
Infect Immun ; 73(8): 4960-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16041011

ABSTRACT

Filamentous hemagglutinin (FHA) is a large (>200 kDa), rod-shaped protein expressed by bordetellae that is both surface-associated and secreted. FHA mediates bacterial adherence to epithelial cells and macrophages in vitro and is absolutely required for tracheal colonization in vivo. The recently sequenced Bordetella bronchiseptica genome revealed the presence of a gene, fhaS, that is nearly identical to fhaB, the FHA structural gene. We show that although fhaS expression requires the BvgAS virulence control system, it is maximal only under a subset of conditions in which BvgAS is active, suggesting an additional level of regulation. We also show that, like FHA, FhaS undergoes a C-terminal proteolytic processing event and is both surface-associated and secreted and that export across the outer membrane requires the channel-forming protein FhaC. Unlike FHA, however, FhaS was unable to mediate adherence of B. bronchiseptica to epithelial cell lines in vitro and was not required for respiratory tract colonization in vivo. In a coinfection experiment, a DeltafhaS strain was out-competed by wild-type B. bronchiseptica, indicating that fhaS is expressed in vivo and that FhaS contributes to bacterial fitness in a manner revealed when the mutant must compete with wild-type bacteria. These data suggest that FHA and FhaS perform distinct functions during the Bordetella infectious cycle. A survey of various Bordetella strains revealed two distinct fhaS alleles that segregate according to pathogen host range and that B. parapertussis(hu) most likely acquired its fhaS allele from B. pertussis horizontally, suggesting fhaS may contribute to host-species specificity.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Bordetella bronchiseptica/genetics , Adhesins, Bacterial/genetics , Alleles , Animals , Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/metabolism , Bordetella Infections/metabolism , Bordetella bronchiseptica/metabolism , Bordetella bronchiseptica/pathogenicity , Bordetella parapertussis/genetics , Bordetella pertussis/genetics , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Hemagglutinins/genetics , Protein Transport/physiology , Rats , Rats, Wistar , Virulence/genetics , Virulence Factors, Bordetella/genetics
20.
Mol Microbiol ; 48(3): 725-36, 2003 May.
Article in English | MEDLINE | ID: mdl-12694617

ABSTRACT

Bordetella bronchiseptica lipopolysaccharide (LPS) expression varies depending on growth conditions, regulated by the Bvg system. A B. bronchiseptica pagP homologue was identified that is required for Bvg-mediated modification of the lipid A core region of LPS that occurs on switching from the Bvg- to the Bvg+ phase. Structural analysis demonstrated that the lipid A of a B. bronchiseptica pagP mutant differed from wild-type lipid A by the absence of a palmitate group in secondary acylation at the C3' position. The putative pagP promoter drove the expression of a green fluorescent protein (GFP) reporter gene in a Bvg-regulated fashion. These data suggest that B. bronchiseptica pagP encodes a Bvg-regulated lipid A palmitoyl transferase that mediates modification of the lipid A as part of the overall Bvg-mediated adaptation of this organism to changing environmental conditions. We also show that pagP is not required for the initial colonization of the mouse respiratory tract by B. bronchiseptica, but is required for persistence of the organism within this organ.


Subject(s)
Acyltransferases/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bordetella bronchiseptica/metabolism , Respiratory System/microbiology , Acyltransferases/genetics , Animals , Bacterial Outer Membrane Proteins/genetics , Bordetella Infections/metabolism , Bordetella bronchiseptica/chemistry , Bordetella bronchiseptica/genetics , Carbohydrate Sequence , Female , Genes, Reporter , Humans , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Molecular Structure , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Respiratory System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...