Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38328073

RESUMEN

Despite global vaccination, pertussis caused by Bordetella pertussis (Bp) is resurging. Pertussis resurgence is correlated with the switch from whole cell vaccines (wPV) that elicit TH1/TH17 polarized immune responses to acellular pertussis vaccines (aPV) that elicit primarily TH2 polarized immune responses. One explanation for the increased incidence in aPV-immunized individuals is the lack of bacterial clearance from the nose. To understand the host and bacterial mechanisms that contribute to Bp persistence, we evaluated bacterial localization and the immune response in the nasal associated tissues (NT) of naïve and immunized mice following Bp challenge. Bp resided in the NT of unimmunized and aPV-immunized mice as biofilms. In contrast, Bp biofilms were not observed in wPV-immunized mice. Following infection, Siglec-F+ neutrophils, critical for eliminating Bp from the nose, were recruited to the nose at higher levels in wPV immunized mice compared to aPV immunized mice. Consistent with this observation, the neutrophil chemokine CXCL1 was only detected in the NT of wPV immunized mice. Importantly, the bacteria and immune cells were primarily localized within the NT and were not recovered by nasal lavage (NL). Together, our data suggest that the TH2 polarized immune response generated by aPV vaccination facilitates persistence in the NT by impeding the infiltration of immune effectors and the eradication of biofilms In contrast, the TH1/TH17 immune phenotype generated by wPV, recruits Siglec-F+ neutrophils that rapidly eliminate the bacterial burden and prevent biofilm establishment. Thus, our work shows that aPV and wPV have opposing effects on Bp biofilm formation in the respiratory tract and provides a mechanistic explanation for the inability of aPV vaccination to control bacterial numbers in the nose and prevent transmission.

2.
Front Immunol ; 14: 1181876, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37275891

RESUMEN

Introduction: Resurgence of pertussis, caused by Bordetella pertussis, necessitates novel vaccines and vaccination strategies to combat this disease. Alum-adjuvanted acellular pertussis vaccines (aPV) delivered intramuscularly reduce bacterial numbers in the lungs of immunized animals and humans, but do not reduce nasal colonization. Thus, aPV-immunized individuals are sources of community transmission. We showed previously that modification of a commercial aPV (Boostrix) by addition of the Th1/17 polarizing adjuvant Bordetella Colonization Factor A (BcfA) attenuated Th2 responses elicited by alum and accelerated clearance of B. pertussis from mouse lungs. Here we tested whether a heterologous immunization strategy with systemic priming and mucosal booster (prime-pull) would reduce nasal colonization. Methods: Adult male and female mice were immunized intramuscularly (i.m.) with aPV or aPV/BcfA and boosted either i.m. or intranasally (i.n.) with the same formulation. Tissue-resident memory (TRM) responses in the respiratory tract were quantified by flow cytometry, and mucosal and systemic antibodies were quantified by ELISA. Immunized and naïve mice were challenged i.n. with Bordetella pertussis and bacterial load in the nose and lungs enumerated at days 1-14 post-challenge. Results: We show that prime-pull immunization with Boostrix plus BcfA (aPV/BcfA) generated IFNγ+ and IL-17+ CD4+ lung resident memory T cells (TRM), and CD4+IL-17+ TRM in the nose. In contrast, aPV alone delivered by the same route generated IL-5+ CD4+ resident memory T cells in the lungs and nose. Importantly, nasal colonization was only reduced in mice immunized with aPV/BcfA by the prime-pull regimen. Conclusions: These results suggest that TH17 polarized TRM generated by aPV/BcfA may reduce nasal colonization thereby preventing pertussis transmission and subsequent resurgence.


Asunto(s)
Bordetella pertussis , Tos Ferina , Animales , Femenino , Masculino , Ratones , Adyuvantes Inmunológicos , Adyuvantes Farmacéuticos , Linfocitos T CD4-Positivos , Interleucina-17 , Vacuna contra la Tos Ferina , Tos Ferina/prevención & control
3.
J Immunol ; 210(9): 1257-1271, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36881867

RESUMEN

Vaccines against SARS-CoV-2 that induce mucosal immunity capable of preventing infection and disease remain urgently needed. In this study, we demonstrate the efficacy of Bordetella colonization factor A (BcfA), a novel bacteria-derived protein adjuvant, in SARS-CoV-2 spike-based prime-pull immunizations. We show that i.m. priming of mice with an aluminum hydroxide- and BcfA-adjuvanted spike subunit vaccine, followed by a BcfA-adjuvanted mucosal booster, generated Th17-polarized CD4+ tissue-resident memory T cells and neutralizing Abs. Immunization with this heterologous vaccine prevented weight loss following challenge with mouse-adapted SARS-CoV-2 (MA10) and reduced viral replication in the respiratory tract. Histopathology showed a strong leukocyte and polymorphonuclear cell infiltrate without epithelial damage in mice immunized with BcfA-containing vaccines. Importantly, neutralizing Abs and tissue-resident memory T cells were maintained until 3 mo postbooster. Viral load in the nose of mice challenged with the MA10 virus at this time point was significantly reduced compared with naive challenged mice and mice immunized with an aluminum hydroxide-adjuvanted vaccine. We show that vaccines adjuvanted with alum and BcfA, delivered through a heterologous prime-pull regimen, provide sustained protection against SARS-CoV-2 infection.


Asunto(s)
Hidróxido de Aluminio , COVID-19 , Humanos , Animales , Ratones , Inmunidad Mucosa , Vacunas contra la COVID-19 , COVID-19/prevención & control , SARS-CoV-2 , Inmunización , Adyuvantes Inmunológicos , Anticuerpos Antivirales , Anticuerpos Neutralizantes
4.
PLoS Pathog ; 19(2): e1011193, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36821596

RESUMEN

Traditionally, whooping cough or pertussis caused by the obligate human pathogen Bordetella pertussis (Bp) is described as an acute disease with severe symptoms. However, many individuals who contract pertussis are either asymptomatic or show very mild symptoms and yet can serve as carriers and sources of bacterial transmission. Biofilms are an important survival mechanism for bacteria in human infections and disease. However, bacterial determinants that drive biofilm formation in humans are ill-defined. In the current study, we show that Bp infection of well-differentiated primary human bronchial epithelial cells leads to formation of bacterial aggregates, clusters, and highly structured biofilms which are colocalized with cilia. These findings mimic observations from pathological analyses of tissues from pertussis patients. Distinct arrangements (mono-, bi-, and tri-partite) of the polysaccharide Bps, extracellular DNA, and bacterial cells were visualized, suggesting complex heterogeneity in bacteria-matrix interactions. Analyses of mutant biofilms revealed positive roles in matrix production, cell cluster formation, and biofilm maturity for three critical Bp virulence factors: Bps, filamentous hemagglutinin, and adenylate cyclase toxin. Adherence assays identified Bps as a new Bp adhesin for primary human airway cells. Taken together, our results demonstrate the multi-factorial nature of the biofilm extracellular matrix and biofilm development process under conditions mimicking the human respiratory tract and highlight the importance of model systems resembling the natural host environment to investigate pathogenesis and potential therapeutic strategies.


Asunto(s)
Bordetella pertussis , Tos Ferina , Humanos , Bordetella pertussis/genética , Tos Ferina/microbiología , Biopelículas , Epitelio , Sistema Respiratorio
5.
Bioorg Med Chem ; 74: 117066, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36283250

RESUMEN

Pertussis is a highly contagious respiratory disease caused by the Gram-negative bacterial pathogen, Bordetella pertussis. Despite high global vaccination rates, pertussis is resurging worldwide. Here we discuss the development of current pertussis vaccines and their limitations, which highlight the need for new vaccines that can protect against the disease and prevent development of the carrier state, thereby reducing transmission. The lipo-oligosaccharide of Bp is an attractive antigen for vaccine development as the anti-glycan antibodies could have bactericidal activities. The structure of the lipo-oligosaccharide has been determined and its immunological properties analyzed. Strategies enabling the expression, isolation, and bioconjugation have been presented. However, obtaining the saccharide on a large scale with high purity remains one of the main obstacles. Chemical synthesis provides a complementary approach to accessing the carbohydrate epitopes in a pure and structurally well-defined form. The first total synthesis of the non-reducing end pertussis pentasaccharide is discussed. The conjugate of the synthetic glycan with a powerful immunogenic carrier, bacteriophage Qß, results in high levels and long-lasting anti-glycan IgG antibodies, paving the way for the development of a new generation of anti-pertussis vaccines with high bactericidal activities and biocompatibilities.


Asunto(s)
Anticuerpos Antibacterianos , Tos Ferina , Humanos , Vacuna contra la Tos Ferina , Bordetella pertussis , Tos Ferina/prevención & control , Oligosacáridos/química
6.
PLoS Pathog ; 18(8): e1010764, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35969621

RESUMEN

Infections and disease caused by the obligate human pathogen Bordetella pertussis (Bp) are increasing, despite widespread vaccinations. The current acellular pertussis vaccines remain ineffective against nasopharyngeal colonization, carriage, and transmission. In this work, we tested the hypothesis that Bordetella polysaccharide (Bps), a member of the poly-ß-1,6-N-acetyl-D-glucosamine (PNAG/PGA) family of polysaccharides promotes respiratory tract colonization of Bp by resisting killing by antimicrobial peptides (AMPs). Genetic deletion of the bpsA-D locus, as well as treatment with the specific glycoside hydrolase Dispersin B, increased susceptibility to AMP-mediated killing. Bps was found to be both cell surface-associated and released during laboratory growth and mouse infections. Addition of bacterial supernatants containing Bps and purified Bps increased B. pertussis resistance to AMPs. By utilizing ELISA, immunoblot and flow cytometry assays, we show that Bps functions as a dual surface shield and decoy. Co-inoculation of C57BL/6J mice with a Bps-proficient strain enhanced respiratory tract survival of the Bps-deficient strain. In combination, the presented results highlight the critical role of Bps as a central driver of B. pertussis pathogenesis. Heterologous production of Bps in a non-pathogenic E. coli K12 strain increased AMP resistance in vitro, and augmented bacterial survival and pathology in the mouse respiratory tract. These studies can serve as a foundation for other PNAG/PGA polysaccharides and for the development of an effective Bp vaccine that includes Bps.


Asunto(s)
Infecciones por Escherichia coli , Tos Ferina , Animales , Humanos , Ratones , Péptidos Antimicrobianos , Biopelículas , Bordetella pertussis/genética , Escherichia coli , Ratones Endogámicos C57BL , Vacuna contra la Tos Ferina , Polisacáridos
7.
Sci Rep ; 12(1): 8439, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589928

RESUMEN

Contamination of soil by antibiotics and heavy metals originating from hospital facilities has emerged as a major cause for the development of resistant microbes. We collected soil samples surrounding a hospital effluent and measured the resistance of bacterial isolates against multiple antibiotics and heavy metals. One strain BMCSI 3 was found to be sensitive to all tested antibiotics. However, it was resistant to many heavy metals and metalloids like cadmium, chromium, copper, mercury, arsenic, and others. This strain was motile and potentially spore-forming. Whole-genome shotgun assembly of BMCSI 3 produced 4.95 Mb genome with 4,638 protein-coding genes. The taxonomic and phylogenetic analysis revealed it, to be a Bordetella petrii strain. Multiple genomic islands carrying mobile genetic elements; coding for heavy metal resistant genes, response regulators or transcription factors, transporters, and multi-drug efflux pumps were identified from the genome. A comparative genomic analysis of BMCSI 3 with annotated genomes of other free-living B. petrii revealed the presence of multiple transposable elements and several genes involved in stress response and metabolism. This study provides insights into how genomic reorganization and plasticity results in evolution of heavy metals resistance by acquiring genes from its natural environment.


Asunto(s)
Metales Pesados , Suelo , Antibacterianos , Bordetella , Genómica , Hospitales , Metales Pesados/toxicidad , Filogenia
9.
Angew Chem Int Ed Engl ; 59(16): 6451-6458, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-31953912

RESUMEN

With the infection rate of Bordetella pertussis at a 60-year high, there is an urgent need for new anti-pertussis vaccines. The lipopolysaccharide (LPS) of B. pertussis is an attractive antigen for vaccine development. With the presence of multiple rare sugars and unusual glycosyl linkages, the B. pertussis LPS is a highly challenging synthetic target. In this work, aided by molecular dynamics simulation and modeling, a pertussis-LPS-like pentasaccharide was chemically synthesized for the first time. The pentasaccharide was conjugated with a powerful carrier, bacteriophage Qß, as a vaccine candidate. Immunization of mice with the conjugate induced robust anti-glycan IgG responses with IgG titers reaching several million enzyme-linked immunosorbent assay (ELISA) units. The antibodies generated were long lasting and boostable and could recognize multiple clinical strains of B. pertussis, highlighting the potential of Qß-glycan as a new anti-pertussis vaccine.


Asunto(s)
Oligosacáridos/inmunología , Vacuna contra la Tos Ferina/síntesis química , Animales , Bovinos , Ensayo de Inmunoadsorción Enzimática , Fucosa/química , Hemocianinas/química , Inmunoglobulina G/sangre , Lipopolisacáridos/síntesis química , Lipopolisacáridos/química , Lipopolisacáridos/inmunología , Ratones , Oligosacáridos/síntesis química , Oligosacáridos/química , Vacuna contra la Tos Ferina/química , Vacuna contra la Tos Ferina/inmunología , Albúmina Sérica Bovina/química
10.
PLoS One ; 14(11): e0223387, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31697703

RESUMEN

Bordetella bacteria are respiratory pathogens of humans, birds, and livestock. Bordetella pertussis the causative agent of whopping cough remains a significant health issue. The transcriptional regulator, BpsR, represses a number of Bordetella genes relating to virulence, cell adhesion, cell motility, and nicotinic acid metabolism. DNA binding of BpsR is allosterically regulated by interaction with 6-hydroxynicotinic acid (6HNA), the first product in the nicotinic acid degradation pathway. To understand the mechanism of this regulation, we have determined the crystal structures of BpsR and BpsR in complex with 6HNA. The structures reveal that BpsR binding of 6HNA induces a conformational change in the protein to prevent DNA binding. We have also identified homologs of BpsR in other Gram negative bacteria in which the amino acids involved in recognition of 6HNA are conserved, suggesting a similar mechanism for regulating nicotinic acid degradation.


Asunto(s)
Biopelículas/crecimiento & desarrollo , ADN/metabolismo , Ácidos Nicotínicos/metabolismo , Factores de Transcripción/genética , Aminoácidos/metabolismo , Proteínas Bacterianas , Bordetella pertussis/metabolismo , Niacina/metabolismo , Factores de Transcripción/metabolismo , Virulencia/fisiología
11.
Infect Immun ; 87(10)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31308083

RESUMEN

Bordetella bronchiseptica is an etiologic agent of respiratory diseases in animals and humans. Despite the widespread use of veterinary B. bronchiseptica vaccines, there is limited information on their composition and relative efficacy and on the immune responses that they elicit. Furthermore, human B. bronchiseptica vaccines are not available. We leveraged the dual antigenic and adjuvant functions of Bordetella colonization factor A (BcfA) to develop acellular B. bronchiseptica vaccines in the absence of an additional adjuvant. BALB/c mice immunized with BcfA alone or a trivalent vaccine containing BcfA and the Bordetella antigens FHA and Prn were equally protected against challenge with a prototype B. bronchiseptica strain. The trivalent vaccine protected mice significantly better than the canine vaccine Bronchicine and provided protection against a B. bronchiseptica strain isolated from a dog with kennel cough. Th1/17-polarized immune responses correlate with long-lasting protection against bordetellae and other respiratory pathogens. Notably, BcfA strongly attenuated the Th2 responses elicited by FHA and Prn, resulting in Th1/17-skewed responses in inherently Th2-skewed BALB/c mice. Thus, BcfA functions as both an antigen and an adjuvant, providing protection as a single-component vaccine. BcfA-adjuvanted vaccines may improve the efficacy and durability of vaccines against bordetellae and other pathogens.


Asunto(s)
Adhesinas Bacterianas/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Antígenos Bacterianos/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Vacunas Bacterianas/administración & dosificación , Infecciones por Bordetella/prevención & control , Bordetella bronchiseptica/efectos de los fármacos , Factores de Virulencia de Bordetella/administración & dosificación , Animales , Infecciones por Bordetella/inmunología , Infecciones por Bordetella/microbiología , Bordetella bronchiseptica/inmunología , Bordetella bronchiseptica/patogenicidad , Perros , Femenino , Humanos , Inmunización , Inmunogenicidad Vacunal , Masculino , Ratones , Ratones Endogámicos BALB C , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/microbiología , Balance Th1 - Th2/efectos de los fármacos , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/microbiología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/microbiología
12.
Xenotransplantation ; 26(5): e12534, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31342586

RESUMEN

BACKGROUND: Xenografts are an attractive alternative to traditional bone grafts because of the large supply from donors with predictable morphology and biology as well as minimal risk of human disease transmission. Clinical series involving xenograft bone transplantation, most commonly from bovine sources, have reported poor results with frequent graft rejection and failure to integrate with host tissue. Failures have been attributed to residual alpha-Gal epitope in the xenograft which humans produce natural antibody against. To the authors' knowledge, there is currently no xenograft-derived bone graft substitute that has been adopted by orthopedic surgeons for routine clinical use. METHODS: In the current study, a bone scaffold intended to serve as a bone graft substitute was derived from porcine cancellous bone using a tissue decellularization and chemical oxidation protocol. In vitro cytocompatibility, pathogen clearance, and alpha-Gal quantification tests were used to assess the safety of the bone scaffold intended for human use. RESULTS: In vitro studies showed the scaffold was free of processing chemicals and biocompatible with mouse and human cell lines. When bacterial and viral pathogens were purposefully added to porcine donor tissue, processing successfully removed these pathogens to comply with sterility assurance levels established by allograft tissue providers. Critically, 98.5% of the alpha-Gal epitope was removed from donor tissue after decellularization as shown by ELISA inhibition assay and immunohistochemical staining. CONCLUSIONS: The current investigation supports the biologic safety of bone scaffolds derived from porcine donors using a decellularization protocol that meets current sterility assurance standards. The majority of the highly immunogenic xenograft carbohydrate was removed from donor tissue, and these findings support further in vivo investigation of xenograft-derived bone tissue for orthopedic clinical application.


Asunto(s)
Sustitutos de Huesos/metabolismo , Xenoinjertos/inmunología , Andamios del Tejido , Trasplante Heterólogo , alfa-Galactosidasa/metabolismo , Animales , Biomarcadores/metabolismo , Ensayo de Inmunoadsorción Enzimática , Xenoinjertos/metabolismo , Xenoinjertos/microbiología , Humanos , Inmunohistoquímica , Porcinos , Andamios del Tejido/microbiología , alfa-Galactosidasa/inmunología
13.
J Vis Exp ; (144)2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30855568

RESUMEN

Vaccines are a 20th century medical marvel. They have dramatically reduced the morbidity and mortality caused by infectious diseases and contributed to a striking increase in life expectancy around the globe. Nonetheless, determining vaccine efficacy remains a challenge. Emerging evidence suggests that the current acellular vaccine (aPV) for Bordetella pertussis (B. pertussis) induces suboptimal immunity. Therefore, a major challenge is designing a next-generation vaccine that induces protective immunity without the adverse side effects of a whole-cell vaccine (wPV). Here we describe a protocol that we used to test the efficacy of a promising, novel adjuvant that skews immune responses to a protective Th1/Th17 phenotype and promotes a better clearance of a B. pertussis challenge from the murine respiratory tract. This article describes the protocol for mouse immunization, bacterial inoculation, tissue harvesting, and analysis of immune responses. Using this method, within our model, we have successfully elucidated crucial mechanisms elicited by a promising, next-generation acellular pertussis vaccine. This method can be applied to any infectious disease model in order to determine vaccine efficacy.


Asunto(s)
Interacciones Huésped-Patógeno , Vacuna contra la Tos Ferina/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Bordetella pertussis/inmunología , Bordetella pertussis/fisiología , Femenino , Masculino , Ratones , Células Th17/inmunología , Vacunación , Tos Ferina/prevención & control
15.
PLoS Pathog ; 14(4): e1006998, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29684093

RESUMEN

Poly-ß(1,6)-N-acetyl-D-glucosamine (PNAG) is a major biofilm component of many pathogenic bacteria. The production, modification, and export of PNAG in Escherichia coli and Bordetella species require the protein products encoded by the pgaABCD operon. PgaB is a two-domain periplasmic protein that contains an N-terminal deacetylase domain and a C-terminal PNAG binding domain that is critical for export. However, the exact function of the PgaB C-terminal domain remains unclear. Herein, we show that the C-terminal domains of Bordetella bronchiseptica PgaB (PgaBBb) and E. coli PgaB (PgaBEc) function as glycoside hydrolases. These enzymes hydrolyze purified deacetylated PNAG (dPNAG) from Staphylococcus aureus, disrupt PNAG-dependent biofilms formed by Bordetella pertussis, Staphylococcus carnosus, Staphylococcus epidermidis, and E. coli, and potentiate bacterial killing by gentamicin. Furthermore, we found that PgaBBb was only able to hydrolyze PNAG produced in situ by the E. coli PgaCD synthase complex when an active deacetylase domain was present. Mass spectrometry analysis of the PgaB-hydrolyzed dPNAG substrate showed a GlcN-GlcNAc-GlcNAc motif at the new reducing end of detected fragments. Our 1.76 Å structure of the C-terminal domain of PgaBBb reveals a central cavity within an elongated surface groove that appears ideally suited to recognize the GlcN-GlcNAc-GlcNAc motif. The structure, in conjunction with molecular modeling and site directed mutagenesis led to the identification of the dPNAG binding subsites and D474 as the probable catalytic acid. This work expands the role of PgaB within the PNAG biosynthesis machinery, defines a new glycoside hydrolase family GH153, and identifies PgaB as a possible therapeutic agent for treating PNAG-dependent biofilm infections.


Asunto(s)
Amidohidrolasas/metabolismo , Biopelículas/crecimiento & desarrollo , Bordetella/enzimología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Glicósido Hidrolasas/metabolismo , beta-Glucanos/química , Acetilación , Amidohidrolasas/química , Bordetella/crecimiento & desarrollo , Cristalografía por Rayos X , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/química , Glicósido Hidrolasas/química , Operón , Conformación Proteica , beta-Glucanos/metabolismo
16.
Infect Immun ; 86(6)2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29531137

RESUMEN

The reemergence of pertussis or whooping cough in several countries highlights the need for better vaccines. Acellular pertussis vaccines (aPV) contain alum as the adjuvant and elicit Th2-biased immune responses that are less effective in protecting against infection than the reactogenic whole-cell pertussis vaccines (wPV), which elicit primarily a Th1/Th17 response. An important goal for the field is to devise aPV that will induce immune responses similar to those of wPV. We show that Bordetella colonization factor A (BcfA), an outer membrane protein from Bordetella bronchiseptica, has strong adjuvant function and elicits cellular and humoral immune responses to heterologous and Bordetella pertussis antigens. Addition of BcfA to a commercial aPV resulted in greater reduction of B. pertussis numbers from the lungs than that elicited by aPV alone. The more-efficient pathogen clearance was accompanied by increased interleukin-17 (IL-17) and reduced IL-5 and an increased ratio of IgG2/IgG1 antibodies. Thus, our results suggest that BcfA improves aPV-induced responses by modifying the alum-induced Th2-biased aPV response toward Th1/Th17. A redesigned aPV containing BcfA may allow better control of pertussis reemergence by reshaping immune responses to resemble those elicited by wPV immunization.


Asunto(s)
Bordetella pertussis/fisiología , Pulmón/microbiología , Vacuna contra la Tos Ferina/inmunología , Vitamina B 12/análogos & derivados , Tos Ferina/microbiología , Inmunidad Adaptativa , Adyuvantes Inmunológicos , Compuestos de Alumbre , Animales , Bordetella pertussis/inmunología , Regulación hacia Abajo , Inmunidad Innata , Ratones , Vitamina B 12/inmunología , Tos Ferina/prevención & control
17.
J Bacteriol ; 200(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29581411

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bordetella bronchiseptica/crecimiento & desarrollo , Bordetella bronchiseptica/metabolismo , Regulación Bacteriana de la Expresión Génica , Niacina/metabolismo , Proteínas Bacterianas/genética , Secuencia de Bases , Bordetella bronchiseptica/genética , Eliminación de Gen , Genes Reguladores , Transcripción Genética
18.
Med Microbiol Immunol ; 207(1): 3-26, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29164393

RESUMEN

Despite high vaccine coverage, whooping cough caused by Bordetella pertussis remains one of the most common vaccine-preventable diseases worldwide. Introduction of whole-cell pertussis (wP) vaccines in the 1940s and acellular pertussis (aP) vaccines in 1990s reduced the mortality due to pertussis. Despite induction of both antibody and cell-mediated immune (CMI) responses by aP and wP vaccines, there has been resurgence of pertussis in many countries in recent years. Possible reasons hypothesised for resurgence have ranged from incompliance with the recommended vaccination programmes with the currently used aP vaccine to infection with a resurged clinical isolates characterised by mutations in the virulence factors, resulting in antigenic divergence with vaccine strain, and increased production of pertussis toxin, resulting in dampening of immune responses. While use of these vaccines provide varying degrees of protection against whooping cough, protection against infection and transmission appears to be less effective, warranting continuation of efforts in the development of an improved pertussis vaccine formulations capable of achieving this objective. Major approaches currently under evaluation for the development of an improved pertussis vaccine include identification of novel biofilm-associated antigens for incorporation in current aP vaccine formulations, development of live attenuated vaccines and discovery of novel non-toxic adjuvants capable of inducing both antibody and CMI. In this review, the potential roles of different accredited virulence factors, including novel biofilm-associated antigens, of B. pertussis in the evolution, formulation and delivery of improved pertussis vaccines, with potential to block the transmission of whooping cough in the community, are discussed.


Asunto(s)
Antígenos Bacterianos/inmunología , Bordetella pertussis/inmunología , Transmisión de Enfermedad Infecciosa/prevención & control , Vacuna contra la Tos Ferina/inmunología , Factores de Virulencia/inmunología , Tos Ferina/prevención & control , Bordetella pertussis/patogenicidad , Descubrimiento de Drogas/tendencias , Humanos , Vacuna contra la Tos Ferina/aislamiento & purificación , Tos Ferina/epidemiología
19.
Infect Immun ; 85(12)2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28893915

RESUMEN

Pertussis, or whooping cough, caused by the obligate human pathogen Bordetella pertussis is undergoing a worldwide resurgence. The majority of studies of this pathogen are conducted with laboratory-adapted strains which may not be representative of the species as a whole. Biofilm formation by B. pertussis plays an important role in pathogenesis. We conducted a side-by-side comparison of the biofilm-forming abilities of the prototype laboratory strains and the currently circulating isolates from two countries with different vaccination programs. Compared to the reference strain, all strains examined herein formed biofilms at high levels. Biofilm structural analyses revealed country-specific differences, with strains from the United States forming more structured biofilms. Bacterial hyperaggregation and reciprocal expression of biofilm-promoting and -inhibitory factors were observed in clinical isolates. An association of increased biofilm formation with augmented epithelial cell adhesion and higher levels of bacterial colonization in the mouse nose and trachea was detected. To our knowledge, this work links for the first time increased biofilm formation in bacteria with a colonization advantage in an animal model. We propose that the enhanced biofilm-forming capacity of currently circulating strains contributes to their persistence, transmission, and continued circulation.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Bordetella pertussis/fisiología , Tos Ferina/microbiología , Animales , Adhesión Bacteriana , Bordetella pertussis/aislamiento & purificación , Bordetella pertussis/patogenicidad , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Humanos , Ratones , Nariz/microbiología , Tráquea/microbiología , Virulencia
20.
Infect Immun ; 85(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28559403

RESUMEN

Bordetella bronchiseptica is pervasive in swine populations and plays multiple roles in respiratory disease. Additionally, B. bronchiseptica is capable of establishing long-term or chronic infections in swine. Bacterial biofilms are increasingly recognized as important contributors to chronic bacterial infections. Recently the polysaccharide locus bpsABCD has been demonstrated to serve a critical role in the development of mature biofilms formed by the sequenced laboratory strain of B. bronchiseptica We hypothesized that swine isolates would also have the ability to form mature biofilms and the bpsABCD locus would serve a key role in this process. A mutant containing an in-frame deletion of the bpsABCD structural genes was constructed in a wild-type swine isolate and found to be negative for poly-N-acetylglucosamine (PNAG)-like material by immunoblot assay. Further, the bpsABCD locus was found to be required for the development and maintenance of the three-dimensional structures under continuous-flow conditions. To investigate the contribution of the bpsABCD locus to the pathogenesis of B. bronchiseptica in swine, the KM22Δbps mutant was compared to the wild-type swine isolate for the ability to colonize and cause disease in pigs. The bpsABCD locus was found to not be required for persistence in the upper respiratory tract of swine. Additionally, the bpsABCD locus did not affect the development of anti-Bordetella humoral immunity, did not contribute to disease severity, and did not mediate protection from complement-mediated killing. However, the bpsABCD locus was found to enhance survival in the lower respiratory tract of swine.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Infecciones por Bordetella/microbiología , Bordetella bronchiseptica/patogenicidad , Polisacáridos Bacterianos/metabolismo , Tráquea/microbiología , Animales , Proteínas Bacterianas/genética , Infecciones por Bordetella/inmunología , Bordetella bronchiseptica/química , Bordetella bronchiseptica/genética , Bordetella bronchiseptica/inmunología , Bronquios/microbiología , Regulación Bacteriana de la Expresión Génica , Mutación , Nariz/microbiología , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...