Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
mBio ; 15(5): e0063224, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38534159

ABSTRACT

Bordetella species that cause respiratory infections in mammals include B. pertussis, which causes human whooping cough, and B. bronchiseptica, which infects nearly all mammals. Both bacterial species produce filamentous hemagglutinin (FhaB) and adenylate cyclase toxin (ACT), prominent surface-associated and secreted virulence factors that contribute to persistence in the lower respiratory tract by inhibiting clearance by phagocytic cells. FhaB and ACT proteins interact with themselves, each other, and host cells. Using immunoblot analyses, we showed that ACT binds to FhaB on the bacterial surface before it can be detected in culture supernatants. We determined that SphB1, a surface protease identified based on its requirement for FhaB cleavage, is also required for ACT cleavage, and we determined that the presence of ACT blocks SphB1-dependent and -independent cleavage of FhaB, but the presence of FhaB does not affect SphB1-dependent cleavage of ACT. The primary SphB1-dependent cleavage site on ACT is proximal to ACT's active site, in a region that is critical for ACT activity. We also determined that FhaB-bound ACT on the bacterial surface can intoxicate host cells producing CR3, the receptor for ACT. In addition to increasing our understanding of FhaB, ACT, and FhaB-ACT interactions on the Bordetella surface, our data are consistent with a model in which FhaB functions as a novel toxin delivery system by binding to ACT and allowing its release upon binding of ACT to its receptor, CR3, on phagocytic cells.IMPORTANCEBacteria need to control the variety, abundance, and conformation of proteins on their surface to survive. Members of the Gram-negative bacterial genus Bordetella include B. pertussis, which causes whooping cough in humans, and B. bronchiseptica, which causes respiratory infections in a broad range of mammals. These species produce two prominent virulence factors, the two-partner secretion (TPS) effector FhaB and adenylate cyclase toxin (ACT), that interact with themselves, each other, and host cells. Here, we determined that ACT binds FhaB on the bacterial surface before being detected in culture supernatants and that ACT bound to FhaB can be delivered to eukaryotic cells. Our data are consistent with a model in which FhaB delivers ACT specifically to phagocytic cells. This is the first report of a TPS system facilitating the delivery of a separate polypeptide toxin to target cells and expands our understanding of how TPS systems contribute to bacterial pathogenesis.


Subject(s)
Adenylate Cyclase Toxin , Phagocytes , Virulence Factors, Bordetella , Adenylate Cyclase Toxin/metabolism , Adenylate Cyclase Toxin/genetics , Phagocytes/metabolism , Phagocytes/microbiology , Virulence Factors, Bordetella/metabolism , Virulence Factors, Bordetella/genetics , Humans , Bordetella pertussis/metabolism , Bordetella pertussis/genetics , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/genetics , Bordetella bronchiseptica/metabolism , Bordetella bronchiseptica/genetics , Protein Binding , Animals
2.
Sci Rep ; 13(1): 7157, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37130958

ABSTRACT

Bordetella bronchiseptica is a gram-negative bacterium that causes respiratory diseases in different animals, including mice, making B. bronchiseptica the gold-standard model to investigate host-pathogen interaction at the molecular level. B. bronchiseptica utilizes many different mechanisms to precisely regulate the expression of virulence factors. Cyclic di-GMP is a second messenger synthesized by diguanylate cyclases and degraded by phosphodiesterases that regulates the expression of multiple virulence factors including biofilm formation. As in other bacteria, we have previously shown that c-di-GMP regulates motility and biofilm formation in B. bronchiseptica. This work describes the diguanylate cyclase BdcB (Bordetella diguanylate cyclase B) as an active diguanylate cyclase that promotes biofilm formation and inhibits motility in B. bronchiseptica. The absence of BdcB increased macrophage cytotoxicity in vitro and induced a greater production of TNF-α, IL-6, and IL-10 by macrophages. Our study reveals that BdcB regulates the expression of components of T3SS, an important virulence factor of B. bronchiseptica. The Bb∆bdcB mutant presented increased expression of T3SS-mediated toxins such as bteA, responsible for cytotoxicity. Our in vivo results revealed that albeit the absence of bdcB did not affect the ability of B. bronchiseptica to infect and colonize the respiratory tract of mice, mice infected with Bb∆bdcB presented a significantly higher pro-inflammatory response than those infected with wild type B. bronchiseptica.


Subject(s)
Bordetella bronchiseptica , Type III Secretion Systems , Mice , Animals , Type III Secretion Systems/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella bronchiseptica/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Cyclic GMP/metabolism , Immunity , Gene Expression Regulation, Bacterial
3.
Viruses ; 14(1)2022 01 14.
Article in English | MEDLINE | ID: mdl-35062352

ABSTRACT

Respiratory disease in horses is caused by a multifactorial complex of infectious agents and environmental factors. An important pathogen in horses is equine herpesvirus type 1 (EHV-1). During co-evolution with this ancient alphaherpesvirus, the horse's respiratory tract has developed multiple antiviral barriers. However, these barriers can become compromised by environmental threats. Pollens and mycotoxins enhance mucosal susceptibility to EHV-1 by interrupting cell junctions, allowing the virus to reach its basolateral receptor. Whether bacterial toxins also play a role in this impairment has not been studied yet. Here, we evaluated the role of α-hemolysin (Hla) and adenylate cyclase (ACT), toxins derived from the facultative pathogenic bacterium Staphylococcus aureus (S. aureus) and the primary pathogen Bordetella bronchiseptica (B. bronchiseptica), respectively. Equine respiratory mucosal explants were cultured at an air-liquid interface and pretreated with these toxins, prior to EHV-1 inoculation. Morphological analysis of hematoxylin-eosin (HE)-stained sections of the explants revealed a decreased epithelial thickness upon treatment with both toxins. Additionally, the Hla toxin induced detachment of epithelial cells and a partial loss of cilia. These morphological changes were correlated with increased EHV-1 replication in the epithelium, as assessed by immunofluorescent stainings and confocal microscopy. In view of these results, we argue that the ACT and Hla toxins increase the susceptibility of the epithelium to EHV-1 by disrupting the epithelial barrier function. In conclusion, this study is the first to report that bacterial exotoxins increase the horse's sensitivity to EHV-1 infection. Therefore, we propose that horses suffering from infection by S. aureus or B. bronchiseptica may be more susceptible to EHV-1 infection.


Subject(s)
Bacterial Toxins/pharmacology , Bordetella bronchiseptica/metabolism , Herpesviridae Infections/drug therapy , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/drug effects , Horse Diseases/virology , Respiratory Tract Diseases/virology , Staphylococcus aureus/metabolism , Animals , Epithelial Cells/virology , Hemolysin Proteins , Horses , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , Virus Replication/drug effects
4.
Sci Rep ; 11(1): 19814, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615931

ABSTRACT

Pore-forming repeats in toxins (RTX) are key virulence factors of many Gram-negative pathogens. We have recently shown that the aromatic side chain of the conserved tyrosine residue 940 within the acylated segment of the RTX adenylate cyclase toxin-hemolysin (CyaA, ACT or AC-Hly) plays a key role in target cell membrane interaction of the toxin. Therefore, we used a truncated CyaA-derived RTX719 construct to analyze the impact of Y940 substitutions on functional folding of the acylated segment of CyaA. Size exclusion chromatography combined with CD spectroscopy revealed that replacement of the aromatic side chain of Y940 by the side chains of alanine or proline residues disrupted the calcium-dependent folding of RTX719 and led to self-aggregation of the otherwise soluble and monomeric protein. Intriguingly, corresponding alanine substitutions of the conserved Y642, Y643 and Y639 residues in the homologous RtxA, HlyA and ApxIA hemolysins from Kingella kingae, Escherichia coli and Actinobacillus pleuropneumoniae, affected the membrane insertion, pore-forming (hemolytic) and cytotoxic capacities of these toxins only marginally. Activities of these toxins were impaired only upon replacement of the conserved tyrosines  by proline residues. It appears, hence, that the critical role of the aromatic side chain of the Y940 residue is highly specific for the functional folding of the acylated domain of CyaA and determines its capacity to penetrate target cell membrane.


Subject(s)
Adenylate Cyclase Toxin/genetics , Bordetella Infections/microbiology , Bordetella bronchiseptica , Bordetella pertussis , Animals , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/metabolism , Bordetella pertussis/genetics , Bordetella pertussis/metabolism , Cell Membrane/metabolism , Female , Hemolysis , Humans , Mice , Mice, Inbred BALB C , THP-1 Cells
5.
Biochemistry ; 60(36): 2727-2738, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34455776

ABSTRACT

Zinc homeostasis in mammals is constantly and precisely maintained by sophisticated regulatory proteins. Among them, the Zrt/Irt-like protein (ZIP) regulates the influx of zinc into the cytoplasm. In this work, we have employed all-atom molecular dynamics simulations to investigate the Zn2+ transport mechanism in prokaryotic ZIP obtained from Bordetella bronchiseptica (BbZIP) in a membrane bilayer. Additionally, the structural and dynamical transformations of BbZIP during this process have been analyzed. This study allowed us to develop a hypothesis for the zinc influx mechanism and formation of the metal-binding site. We have created a model for the outward-facing form of BbZIP (experimentally only the inward-facing form has been characterized) that has allowed us, for the first time, to observe the Zn2+ ion entering the channel and binding to the negatively charged M2 site. It is thought that the M2 site is less favored than the M1 site, which then leads to metal ion egress; however, we have not observed the M1 site being occupied in our simulations. Furthermore, removing both Zn2+ ions from this complex resulted in the collapse of the metal-binding site, illustrating the "structural role" of metal ions in maintaining the binding site and holding the proteins together. Finally, due to the long Cd2+-residue bond distances observed in the X-ray structures, we have proposed the existence of an H3O+ ion at the M2 site that plays an important role in protein stability in the absence of the metal ion.


Subject(s)
Bordetella bronchiseptica/metabolism , Carrier Proteins/chemistry , Cation Transport Proteins/metabolism , Computer Simulation/standards , Zinc/metabolism , Carrier Proteins/metabolism , Molecular Dynamics Simulation , Protein Structural Elements
6.
J Biol Chem ; 296: 100607, 2021.
Article in English | MEDLINE | ID: mdl-33789161

ABSTRACT

The respiratory pathogens Bordetella pertussis and Bordetella bronchiseptica employ a type III secretion system (T3SS) to inject a 69-kDa BteA effector protein into host cells. This effector is known to contain two functional domains, including an N-terminal lipid raft targeting (LRT) domain and a cytotoxic C-terminal domain that induces nonapoptotic and caspase-1-independent host cell death. However, the exact molecular mechanisms underlying the interaction of BteA with plasma membrane (PM) as well as its cytotoxic activity in the course of Bordetella infections remain poorly understood. Using a protein-lipid overlay assay and surface plasmon resonance, we show here that the recombinant LRT domain binds negatively charged membrane phospholipids. Specifically, we determined that the dissociation constants of the LRT domain-binding liposomes containing phosphatidylinositol 4,5-bisphosphate, phosphatidic acid, and phosphatidylserine were ∼450 nM, ∼490 nM, and ∼1.2 µM, respectively. Both phosphatidylserine and phosphatidylinositol 4,5-bisphosphate were required to target the LRT domain and/or full-length BteA to the PM of yeast cells. The membrane association further involved electrostatic and hydrophobic interactions of LRT and depended on a leucine residue in the L1 loop between the first two helices of the four-helix bundle. Importantly, charge-reversal substitutions within the L1 region disrupted PM localization of the BteA effector without hampering its cytotoxic activity during B. bronchiseptica infection of HeLa cells. The LRT-mediated targeting of BteA to the cytosolic leaflet of the PM of host cells is, therefore, dispensable for effector cytotoxicity.


Subject(s)
Bacterial Proteins/metabolism , Bordetella bronchiseptica/metabolism , Cell Membrane/metabolism , Lipid Bilayers/metabolism , Membrane Microdomains/metabolism , Phospholipids/metabolism , Bacterial Proteins/genetics , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/growth & development , HeLa Cells , Humans , Protein Binding , Protein Domains
7.
Commun Biol ; 4(1): 46, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420409

ABSTRACT

Copper is both essential and toxic to living beings, which tightly controls its intracellular concentration. At the host-pathogen interface, copper is used by phagocytic cells to kill invading microorganisms. We investigated copper homeostasis in Bordetella pertussis, which lives in the human respiratory mucosa and has no environmental reservoir. B. pertussis has considerably streamlined copper homeostasis mechanisms relative to other Gram-negative bacteria. Its single remaining defense line consists of a metallochaperone diverted for copper passivation, CopZ, and two peroxide detoxification enzymes, PrxGrx and GorB, which together fight stresses encountered in phagocytic cells. Those proteins are encoded by an original, composite operon assembled in an environmental ancestor, which is under sensitive control by copper. This system appears to contribute to persistent infection in the nasal cavity of B. pertussis-infected mice. Combining responses to co-occurring stresses in a tailored operon reveals a strategy adopted by a host-restricted pathogen to optimize survival at minimal energy expenditure.


Subject(s)
Bordetella pertussis/metabolism , Copper/metabolism , Operon , Bordetella bronchiseptica/metabolism , Bordetella pertussis/genetics , Homeostasis , Peroxides/metabolism
8.
Virulence ; 12(1): 84-95, 2021 12.
Article in English | MEDLINE | ID: mdl-33372837

ABSTRACT

Bordetella (B.) bronchiseptica and Streptococcus (S.) suis are major pathogens in pigs, which are frequently isolated from co-infections in the respiratory tract and contribute to the porcine respiratory disease complex (PRDC). Despite the high impact of co-infections on respiratory diseases of swine (and other hosts), very little is known about pathogen-pathogen-host interactions and the mechanisms of pathogenesis. In the present study, we established a porcine precision-cut lung slice (PCLS) model to analyze the effects of B. bronchiseptica infection on adherence, colonization, and cytotoxic effects of S. suis. We hypothesized that induction of ciliostasis by a clinical isolate of B. bronchiseptica may promote subsequent infection with a virulent S. suis serotype 2 strain. To investigate this theory, we monitored the ciliary activity by light microscopy, measured the release of lactate dehydrogenase, and calculated the number of PCLS-associated bacteria. To study the role of the pore-forming toxin suilysin (SLY) in S. suis-induced cytotoxicity, we included a SLY-negative isogenic mutant and the complemented mutant strain. Furthermore, we analyzed infected PCLS by histopathology, immunofluorescence microscopy, and field emission scanning electron microscopy. Our results showed that pre-infection with B. bronchiseptica promoted adherence, colonization, and, as a consequence of the increased colonization, the cytotoxic effects of S. suis, probably by reduction of the ciliary activity. Moreover, cytotoxicity induced by S. suis is strictly dependent on the presence of SLY. Though the underlying molecular mechanisms remain to be fully clarified, our results clearly support the hypothesis that B. bronchiseptica paves the way for S. suis infection.


Subject(s)
Bacterial Adhesion , Bordetella bronchiseptica/metabolism , Bordetella bronchiseptica/pathogenicity , Host-Pathogen Interactions , Lung/microbiology , Streptococcus suis/pathogenicity , Animals , Bordetella bronchiseptica/genetics , Cilia/metabolism , Coinfection , Hemolysin Proteins/genetics , In Vitro Techniques , Streptococcus suis/metabolism , Swine , Swine Diseases/microbiology
9.
J Biol Chem ; 294(36): 13327-13335, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31320477

ABSTRACT

Regulated ion diffusion across biological membranes is vital for cell function. In a nanoscale ion channel, the active role of discrete water molecules in modulating hydrodynamic behaviors of individual ions is poorly understood because of the technical challenge of tracking water molecules through the channel. Here we report the results of a hydroxyl radical footprinting analysis of the zinc-selective channel ZIPB from the Gram-negative bacterium, Bordetella bronchiseptica Irradiating ZIPB by microsecond X-ray pulses activated water molecules to form covalent hydroxyl radical adducts at nearby residues, which were identified by bottom-up proteomics to detect residues that interact either with zinc or water in response to zinc binding. We found a series of residues exhibiting reciprocal changes in water accessibility attributed to alternating zinc and water binding. Mapping these residues to the previously reported crystal structure of ZIPB, we identified a water-reactive pathway that superimposed on a zinc translocation pathway consisting of two binuclear metal centers and an interim zinc-binding site. The cotranslocation of zinc and water suggested that pore-lining residues undergo a mode switch between zinc coordination and water binding to confer zinc mobility. The unprecedented details of water-mediated zinc transport identified here highlight an essential role of solvated waters in driving zinc coordination dynamics and transmembrane crossing.


Subject(s)
Bordetella bronchiseptica/metabolism , Cation Transport Proteins/metabolism , Water/metabolism , Zinc/metabolism , Biological Transport , Bordetella bronchiseptica/chemistry , Cation Transport Proteins/chemistry , Diffusion , Water/chemistry , Zinc/chemistry
10.
J Bacteriol ; 201(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31235515

ABSTRACT

To detect and respond to the diverse environments they encounter, bacteria often use two-component regulatory systems (TCS) to coordinate essential cellular processes required for survival. In pathogenic Bordetella species, the BvgAS TCS regulates expression of hundreds of genes, including those encoding all known protein virulence factors, and its kinase activity is essential for respiratory infection. Maintenance of BvgS kinase activity in the lower respiratory tract (LRT) depends on the function of another TCS, PlrSR. While the periplasmic Venus flytrap domains of BvgS have been implicated in responding to so-called modulating signals in vitro (nicotinic acid and MgSO4), a role for the cytoplasmic Per-Arnt-Sim (PAS) domain in signal perception has not previously been demonstrated. By comparing B. bronchiseptica strains with mutations in the PAS domain-encoding region of bvgS with wild-type bacteria in vitro and in vivo, we found that although the PAS domain is not required to sense modulating signals in vitro, it is required for the inactivation of BvgS that occurs in the absence of PlrS in the LRTs of mice, suggesting that the BvgS PAS domain functions as an independent signal perception domain. Our data also indicate that the BvgS PAS domain is important for controlling absolute levels of BvgS kinase activity and the efficiency of the response to modulating signals in vitro Our results provide evidence that BvgS integrates sensory inputs from both the periplasm and the cytoplasm to control precise gene expression patterns under diverse environmental conditions.IMPORTANCE Despite high rates of vaccination, pertussis, a severe, highly contagious respiratory disease caused by the bacterium Bordetella pertussis, has reemerged as a significant health threat. In Bordetella pertussis and the closely related species Bordetella bronchiseptica, activity of the BvgAS two-component regulatory system is critical for colonization of the mammalian respiratory tract. We show here that the cytoplasmic PAS domain of BvgS can function as an independent signal perception domain that influences BvgS activity in response to environmental conditions. Our work is significant because it reveals a critical, yet previously unrecognized, role for the PAS domain in the BvgAS phosphorelay and provides a greater understanding of virulence regulation in Bordetella.


Subject(s)
Bacterial Proteins/metabolism , Bordetella Infections/microbiology , Bordetella bronchiseptica/metabolism , Respiratory Tract Infections/microbiology , Animals , Bacterial Proteins/genetics , Female , Gene Expression Regulation, Bacterial/physiology , Mice , Mice, Inbred BALB C , Mutation , Protein Conformation , Protein Domains , Transcription Factors/metabolism , Transcription, Genetic
11.
Mol Microbiol ; 112(3): 820-836, 2019 09.
Article in English | MEDLINE | ID: mdl-31152610

ABSTRACT

Filamentous hemagglutinin (FHA) is a critically important virulence factor produced by Bordetella species that cause respiratory infections in humans and other animals. It is also a prototypical member of the widespread two partner secretion (TPS) pathway family of proteins. First synthesized as a ~370 kDa protein called FhaB, its C-terminal ~1,200 amino acid 'prodomain' is removed during translocation to the cell surface via the outer membrane channel FhaC. Here, we identify CtpA as a periplasmic protease that is responsible for the regulated degradation of the prodomain and for creation of an intermediate polypeptide that is cleaved by the autotransporter protease SphB1 to generate FHA. We show that the central prodomain region is required to initiate degradation of the prodomain and that CtpA degrades the prodomain after a third, unidentified protease (P3) first removes the extreme C-terminus of the prodomain. Stepwise proteolysis by P3, CtpA and SphB1 is required for maturation of FhaB, release of FHA into the extracellular milieu, and full function in vivo. These data support a substantially updated model for the mechanism of secretion, maturation and function of this model TPS protein.


Subject(s)
Adhesins, Bacterial/metabolism , Algal Proteins/metabolism , Bacterial Proteins/metabolism , Bordetella bronchiseptica/metabolism , Bordetella pertussis/enzymology , Carboxypeptidases/metabolism , Hemagglutinins/metabolism , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Algal Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bordetella bronchiseptica/chemistry , Bordetella bronchiseptica/genetics , Bordetella pertussis/genetics , Bordetella pertussis/metabolism , Carboxypeptidases/genetics , Hemagglutinins/chemistry , Hemagglutinins/genetics , Proprotein Convertases/genetics , Protein Domains , Protein Processing, Post-Translational , Serine Endopeptidases/genetics
12.
J Am Soc Mass Spectrom ; 30(9): 1679-1689, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31190311

ABSTRACT

Bordetella bronchiseptica, known to infect animals and rarely humans, expresses a lipopolysaccharide that plays an essential role in host interactions, being critical for early clearance of the bacteria. On a B. bronchiseptica 9.73 isolate, mutants defective in the expression of genes involved in the biosynthesis of the core region were previously constructed. Herein, a comparative detailed structural analysis of the expressed lipids A by MALDI-TOF mass spectrometry was performed. The Bb3394 LPS defective in a 2-amino-2-deoxy-D-galacturonic acid lateral residue of the core presented a penta-acylated diglucosamine backbone modified with two glucosamine phosphates, similar to the wild-type lipid A. In contrast, BbLP39, resulting in the interruption of the LPS core oligosaccharide synthesis, presented lipid A species consisting in a diglucosamine backbone N-substituted with C14:0(3-O-C12:0) in C-2 and C14:0(3-O-C14:0) in C-2', O-acylated with C14:0(3-O-C10:0(3-OH) in C-3' and with a pyrophosphate in C-1. Regarding Bb3398 also presenting a rough LPS, the lipid A is formed by a hexa-acylated diglucosamine backbone carrying one pyrophosphate group in C-1 and one phosphate in C-4', both substituted with ethanolamine groups. As far as we know, this is the first description of a phosphoethanolamine modification in B. bronchiseptica lipid A. Our results demonstrate that although gene deletions were not directed to the lipid A moiety, each mutant presented different modifications. MALDI-TOF mass spectrometry was an excellent tool to highlight the structural diversity of the lipid A structures biosynthesized during its transit through the periplasm to the final localization in the outer surface of the outer membrane. Graphical Abstract.


Subject(s)
Bordetella bronchiseptica/genetics , Glycosyltransferases/genetics , Lipid A/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bordetella bronchiseptica/chemistry , Bordetella bronchiseptica/metabolism , Diphosphates/chemistry , Glucosamine/chemistry , Glycosyltransferases/chemistry , Lipid A/analysis , Lipid A/genetics , Mutation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
13.
Biochemistry ; 58(13): 1751-1763, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30810301

ABSTRACT

6-Hydroxynicotinate 3-monooxygenase (NicC) is a Group A FAD-dependent monooxygenase that catalyzes the decarboxylative hydroxylation of 6-hydroxynicotinic acid (6-HNA) to 2,5-dihydroxypyridine (2,5-DHP) with concomitant oxidation of NADH in nicotinic acid degradation by aerobic bacteria. Two mechanisms for the decarboxylative hydroxylation half-reaction have been proposed [Hicks, K., et al. (2016) Biochemistry 55, 3432-3446]. Results with Bordetella bronchiseptica RB50 NicC here show that a homocyclic analogue of 6-HNA, 4-hydroxybenzoic acid (4-HBA), is decarboxylated and hydroxylated by NicC with a 420-fold lower catalytic efficiency than is 6-HNA. The 13( V/ K), measured with wild-type NicC by isotope ratio mass spectrometry following the natural abundance of 13C in the CO2 product, is inverse for both 6-HNA (0.9989 ± 0.0002) and 4-HBA (0.9942 ± 0.0004) and becomes negligible (0.9999 ± 0.0004) for 5-chloro-6-HNA, an analogue that is 10-fold more catalytically efficient than 6-HNA. Covalently bound 6-HNA complexes of NicC are not observed by mass spectrometry. Comparative steady-state kinetic and Kd6HNA analyses of active site NicC variants (C202A, H211A, H302A, H47E, Y215F, and Y225F) identify Tyr215 and His47 as critical determinants both of 6-HNA binding ( KdY215F/ KdWT > 240; KdH47E/ KdWT > 350) and in coupling rates of 2,5-DHP and NAD+ product formation ([2,5-DHP]/[NAD+] = 1.00 (WT), 0.005 (Y215F), and 0.07 (H47E)]. Results of these functional analyses are in accord with an electrophilic aromatic substitution reaction mechanism in which His47-Tyr215 may serve as the general base to catalyze substrate hydroxylation and refine the structural model for substrate binding by NicC.


Subject(s)
Bacterial Proteins/metabolism , Bordetella bronchiseptica/metabolism , Mixed Function Oxygenases/metabolism , Niacin/metabolism , Bordetella Infections/microbiology , Bordetella bronchiseptica/enzymology , Flavin-Adenine Dinucleotide/metabolism , Humans , Hydroxylation , Kinetics , Nicotinic Acids/metabolism , Parabens/metabolism , Pyridines/metabolism , Substrate Specificity
14.
Microbiol Immunol ; 62(12): 743-754, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30407657

ABSTRACT

Bordetella species, including B. pertussis, have a type III secretion system that is highly conserved among gram-negative pathogenic bacteria. Genes encoding the component proteins of the type III secretion system are localized at the bsc locus in the Bordetella genome. Here, the function of a hypothetical protein Bcr4 encoded at the bsc locus in the B. bronchiseptica genome was investigated. A Bcr4-deficient mutant was created and the amounts of type III secreted proteins (e.g., BopB, BopN and Bsp22) in both the supernatant fraction and whole-cell lysates of the Bcr4-deficient mutant were determined. It was found that the amounts of these proteins were significantly lower than in the wild-type strain. The amounts of type III secreted proteins in the supernatant fraction and whole-cell lysates were much greater in a Bcr4-overproducing strain than in the wild-type strain. The type III secreted protein BspR reportedly negatively regulates the type III secretion system. Here, it was observed that a Bcr4 + BspR double-knockout mutant did not secrete type III secreted proteins, whereas the amounts of these proteins in whole-cell lysates of this mutant were nearly equal to those in whole-cell lysates of the BspR-deficient mutant. Bcr4 thus appears to play an essential role in the extracellular secretion of type III secreted proteins. Our data also suggest that Bcr4 antagonizes the negative regulatory function of BspR.


Subject(s)
Bacterial Proteins/genetics , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/metabolism , Genes, Bacterial/genetics , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Animals , Bordetella pertussis/genetics , Carrier Proteins/genetics , Cell Line , Gene Expression Regulation, Bacterial , Gene Knockout Techniques , Molecular Weight , Mutation , Protein Transport , Rats , Transcriptome
15.
J Bacteriol ; 200(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29581411

ABSTRACT

Many of the pathogenic species of the genus Bordetella have an absolute requirement for nicotinic acid (NA) for laboratory growth. These Gram-negative bacteria also harbor a gene cluster homologous to the nic cluster of Pseudomonas putida which is involved in the aerobic degradation of NA and its transcriptional control. We report here that BpsR, a negative regulator of biofilm formation and Bps polysaccharide production, controls the growth of Bordetella bronchiseptica by repressing the expression of nic genes. The severe growth defect of the ΔbpsR strain in Stainer-Scholte medium was restored by supplementation with NA, which also functioned as an inducer of nic genes at low micromolar concentrations that are usually present in animals and humans. Purified BpsR protein bound to the nic promoter region, and its DNA binding activity was inhibited by 6-hydroxynicotinic acid (6-HNA), the first metabolite of the NA degradative pathway. Reporter assays with the isogenic mutant derivative of the wild-type (WT) strain harboring deletion in nicA, which encodes a putative nicotinic acid hydroxylase responsible for conversion of NA to 6-HNA, showed that 6-HNA is the actual inducer of the nic genes in the bacterial cell. Gene expression profiling further showed that BpsR dually activated and repressed the expression of genes associated with pathogenesis, transcriptional regulation, metabolism, and other cellular processes. We discuss the implications of these findings with respect to the selection of pyridines such as NA and quinolinic acid for optimum bacterial growth depending on the ecological niche.IMPORTANCE BpsR, the previously described regulator of biofilm formation and Bps polysaccharide production, controls Bordetella bronchiseptica growth by regulating the expression of genes involved in the degradation of nicotinic acid (NA). 6-Hydroxynicotinic acid (6-HNA), the first metabolite of the NA degradation pathway prevented BpsR from binding to DNA and was the actual in vivo inducer. We hypothesize that BpsR enables Bordetella bacteria to efficiently and selectively utilize NA for their survival depending on the environment in which they reside. The results reported herein lay the foundation for future investigations of how BpsR and the alteration of its activity by NA orchestrate the control of Bordetella growth, metabolism, biofilm formation, and pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bordetella bronchiseptica/growth & development , Bordetella bronchiseptica/metabolism , Gene Expression Regulation, Bacterial , Niacin/metabolism , Bacterial Proteins/genetics , Base Sequence , Bordetella bronchiseptica/genetics , Gene Deletion , Genes, Regulator , Transcription, Genetic
16.
J Infect Dis ; 216(7): 899-906, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28973366

ABSTRACT

Background: The lack of animal models to experimentally study how infectious agents transmit between hosts limits our understanding of what makes some pathogens so contagious. Methods: We recently developed a Bordetella bronchiseptica mouse model to study transmission and have used it to assess, for the first time, which of several well-studied "virulence factors" common to classical Bordetella species contribute to transmission. Results: Among 13 mutants screened, a mutant lacking an extracellular polysaccharide (EPS) locus consistently failed to transmit. The loss of EPS had no obvious effect on in vitro characteristics of growth, adherence, cytotoxicity, or serum resistance, though it profoundly reduced the ability of the mutant to colonize the lower respiratory tract of mice. While wild-type B. bronchiseptica was shed from colonized mice and efficiently transmitted to cage-mates, the mutant colonized less efficiently, shed at lower numbers, and consequently did not transmit to naive animals. Conclusions: These results have important implications for potential roles of polysaccharides in the pathogenesis and transmission of Bordetella species as well as other respiratory pathogens. Cases of pertussis (whooping cough) caused by Bordetella pertussis are on the rise, and understanding factors that contribute to their spread is critical to its control.


Subject(s)
Bordetella Infections/microbiology , Bordetella Infections/transmission , Bordetella bronchiseptica/metabolism , Polysaccharides, Bacterial/metabolism , Animals , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation , Polysaccharides, Bacterial/genetics
17.
Microbiol Immunol ; 61(6): 206-214, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28500733

ABSTRACT

Bordetella bronchiseptica infects a wide variety of mammals, the type III secretion system (T3SS) being involved in long-term colonization by Bordetella of the trachea and lung. T3SS translocates virulence factors (commonly referred to as effectors) into host cells, leading to alterations in the host's physiological function. The Bordetella effectors BopN and BteA are known to have roles in up-regulation of IL-10 and cytotoxicity, respectively. Nevertheless, the mechanism by which BopN is translocated into host cells has not been examined in sufficient detail. Therefore, to determine the precise mechanisms of translocation of BopN into host cells, truncated derivatives of BopN were built and the derivatives' ability to translocate into host cells evaluated by adenylate cyclase-mediated translocation assay. It was found that N-terminal amino acid (aa) residues 1-200 of BopN are sufficient for its translocation into host cells. Interestingly, BopN translocation was completely blocked by deletion of the N-terminal aa residues 6-50, indicating that the N-terminal region is critical for BopN translocation. Furthermore, BopN appears to play an auxiliary role in BteA-mediated cytotoxicity. Thus, BopN can apparently translocate into host cells and may facilitate activity of BteA.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella Infections/microbiology , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/metabolism , Protein Transport , Adenylyl Cyclases , Amino Acid Sequence , Animals , Antibodies, Bacterial , Bacterial Proteins/immunology , Cell Line , Cytotoxicity, Immunologic , DNA, Bacterial , Escherichia coli/genetics , Gene Expression Regulation , Genes, Bacterial , Genetic Vectors , Host-Pathogen Interactions , Interleukin-10 , Protein Transport/physiology , Rats , Sequence Deletion , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Up-Regulation , Virulence Factors/metabolism
18.
Bioorg Med Chem Lett ; 27(3): 432-436, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28040392

ABSTRACT

Bordetella bronchiseptica produces respiratory disease primarily in mammals including humans. Although a considerably amount of research has been generated regarding lipopolysaccharide (LPS) role during infection and stimulating innate and adaptive immune response, mechanisms involved in LPS synthesis are still unknown. In this context we searched in B. bronchiseptica genome for putative glycosyltransferases. We found possible genes codifying for enzymes involved in sugar substitution of the LPS structure. We decided to analyse BB3394 to BB3400 genes, closed to a previously described LPS biosynthetic locus in B. pertussis. Particularly, conservation of BB3394 in sequenced B. bronchiseptica genomes suggests the importance of this gene for bacteria normal physiology. Deletion of BB3394 abolished resistance to naive serum as described for other LPS mutants. When purified LPS was analyzed, differences in the LPS core structure were found. Particularly, a GalNA branched sugar substitution in the core was absent in the LPS obtained from BB3394 deletion mutant. Absence of GalNA in core LPS alters immune response in vivo but is able to induce protective response against B. bronchiseptica infection.


Subject(s)
Bordetella bronchiseptica/metabolism , Lipopolysaccharides/biosynthesis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/immunology , Genes, Bacterial , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Immune System/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Mice , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Wei Sheng Wu Xue Bao ; 57(2): 264-9, 2017 Feb 04.
Article in Chinese | MEDLINE | ID: mdl-29750489

ABSTRACT

Objective: We constructed Bordetella bronchiseptica QH0814 hcp mutant to characterize its pathogenicity.[Methods] Through the homologous recombination mediated by a suicide plasmid pRE112, we acquired the mutant QH0814Δhcp successfully. Then, we evaluated the growth condition, the ability of adhesion and invasion, the median lethal dose (LD50) and the infection capacity. Methods: Through the homologous recombination mediated by a suicide plasmid pRE112, we acquired the mutant QH0814Δhcp successfully. Then, we evaluated the growth condition, the ability of adhesion and invasion, the median lethal dose (LD50) and the infection capacity. Results: There was no significant variation of the growth rate between the mutant and the parental strain. Compared with the parental strain, the adherence ability of the mutant did not change remarkably. However, the invasion ability decreased significantly. Mice lethal test showed that the LD50 of the mutant was higher than that of the parental strain. Correspondingly, the bacterial colonization of the mutant in mice blood, lung and liver was much less than that of the parental strain. Conclusion: The knocking-out of the hcp gene had no influence on bacterial growth, but it could attenuate significantly the invasion and colonization of the bacterium. Therefore, the gene may play a role in the pathogenesis of Bordetella bronchiseptica.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bordetella Infections/veterinary , Bordetella bronchiseptica/metabolism , Swine Diseases/microbiology , Animals , Bordetella Infections/microbiology , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/growth & development , Bordetella bronchiseptica/pathogenicity , Mice , Mutation , Plasmids/genetics , Plasmids/metabolism , Swine , Virulence
20.
Mol Microbiol ; 103(2): 214-228, 2017 01.
Article in English | MEDLINE | ID: mdl-27731909

ABSTRACT

Bordetella pertussis, the causative agent of whooping cough, secretes and releases adenylate cyclase toxin (ACT), which is a protein bacterial toxin that targets host cells and disarms immune defenses. ACT binds filamentous haemagglutinin (FHA), a surface-displayed adhesin, and until now, the consequences of this interaction were unknown. A B. bronchiseptica mutant lacking ACT produced more biofilm than the parental strain; leading Irie et al. to propose the ACT-FHA interaction could be responsible for biofilm inhibition. Here we characterize the physical interaction of ACT with FHA and provide evidence linking that interaction to inhibition of biofilm in vitro. Exogenous ACT inhibits biofilm formation in a concentration-dependent manner and the N-terminal catalytic domain of ACT (AC domain) is necessary and sufficient for this inhibitory effect. AC Domain interacts with the C-terminal segment of FHA with ∼650 nM affinity. ACT does not inhibit biofilm formation by Bordetella lacking the mature C-terminal domain (MCD), suggesting the direct interaction between AC domain and the MCD is required for the inhibitory effect. Additionally, AC domain disrupts preformed biofilm on abiotic surfaces. The demonstrated inhibition of biofilm formation by a host-directed protein bacterial toxin represents a novel regulatory mechanism and identifies an unprecedented role for ACT.


Subject(s)
Adenylate Cyclase Toxin/metabolism , Adhesins, Bacterial/metabolism , Biofilms/growth & development , Bordetella bronchiseptica/metabolism , Bordetella pertussis/physiology , Virulence Factors, Bordetella/metabolism , Adenylate Cyclase Toxin/genetics , Adhesins, Bacterial/genetics , Bordetella bronchiseptica/genetics , Bordetella pertussis/genetics , Bordetella pertussis/metabolism , Hemagglutinins/metabolism , Virulence Factors, Bordetella/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...