Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(8): e1009735, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34347835

RESUMO

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.


Assuntos
Células Epiteliais Alveolares/microbiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Derrame de Bactérias , Infecções por Bordetella/transmissão , Bordetella bronchiseptica/patogenicidade , Inflamação/patologia , Fatores de Virulência de Bordetella/metabolismo , Animais , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/genética , Infecções por Bordetella/metabolismo , Infecções por Bordetella/microbiologia , Feminino , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fatores de Virulência de Bordetella/genética
3.
PLoS Pathog ; 15(4): e1007696, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30970038

RESUMO

Infection and inflammation of the middle ears that characterizes acute and chronic otitis media (OM), is a major reason for doctor visits and antibiotic prescription, particularly among children. Nasopharyngeal pathogens that are commonly associated with OM in humans do not naturally colonize the middle ears of rodents, and experimental models in most cases involve directly injecting large numbers of human pathogens into the middle ear bullae of rodents, where they induce a short-lived acute inflammation but fail to persist. Here we report that Bordetella pseudohinzii, a respiratory pathogen of mice, naturally, efficiently and rapidly ascends the eustachian tubes to colonize the middle ears, causing acute and chronic histopathological changes with progressive decrease in hearing acuity that closely mimics otitis media in humans. Laboratory mice experimentally inoculated intranasally with very low numbers of bacteria consistently have their middle ears colonized and subsequently transmit the bacterium to cage mates. Taking advantage of the specifically engineered and well characterized immune deficiencies available in mice we conducted experiments to uncover different roles of T and B cells in controlling bacterial numbers in the middle ear during chronic OM. The iconic mouse model provides significant advantages for elucidating aspects of host-pathogen interactions in otitis media that are currently not possible using other animal models. This natural model of otitis media permits the study of transmission between hosts, efficient early colonization of the respiratory tract, ascension of the eustachian tube, as well as colonization, pathogenesis and persistence in the middle ear. It also allows the combination of the powerful tools of mouse molecular immunology and bacterial genetics to determine the mechanistic basis for these important processes.


Assuntos
Infecções por Bordetella/transmissão , Bordetella/patogenicidade , Modelos Animais de Doenças , Tuba Auditiva/microbiologia , Cavidade Nasal/microbiologia , Otite Média/microbiologia , Animais , Infecções por Bordetella/complicações , Infecções por Bordetella/microbiologia , Doença Crônica , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL
4.
J Antimicrob Chemother ; 73(10): 2797-2805, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30107601

RESUMO

Background: Why resistance to specific antibiotics emerges and spreads rapidly in some bacteria confronting these drugs but not others remains a mystery. Resistance to erythromycin in the respiratory pathogens Staphylococcus aureus and Streptococcus pneumoniae emerged rapidly and increased problematically. However, resistance is uncommon amongst the classic Bordetella species despite infections being treated with this macrolide for decades. Objectives: We examined whether the apparent progenitor of the classic Bordetella spp., Bordetella bronchiseptica, is able to rapidly generate de novo resistance to antibiotics and, if so, why such resistance might not persist and propagate. Methods: Independent strains of B. bronchiseptica resistant to erythromycin were generated in vitro by successively passaging them in increasing subinhibitory concentrations of this macrolide. Resistant mutants obtained were evaluated for their capacity to infect mice, and for other virulence properties including adherence, cytotoxicity and induction of cytokines. Results: B. bronchiseptica rapidly developed stable and persistent antibiotic resistance de novo. Unlike the previously reported trade-off in fitness, multiple independent resistant mutants were not defective in their rates of growth in vitro but were consistently defective in colonizing mice and lost a variety of virulence phenotypes. These changes rendered them avirulent but phenotypically similar to the previously described growth phase associated with the ability to survive in soil, water and/or other extra-mammalian environments. Conclusions: These observations raise the possibility that antibiotic resistance in some organisms results in trade-offs that are not quantifiable in routine measures of general fitness such as growth in vitro, but are pronounced in various aspects of infection in the natural host.


Assuntos
Antibacterianos/farmacologia , Infecções por Bordetella/microbiologia , Infecções por Bordetella/patologia , Bordetella bronchiseptica/efeitos dos fármacos , Bordetella bronchiseptica/patogenicidade , Farmacorresistência Bacteriana , Eritromicina/farmacologia , Animais , Aderência Bacteriana , Toxinas Bacterianas/metabolismo , Bordetella bronchiseptica/crescimento & desenvolvimento , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Camundongos , Mutação , Seleção Genética , Inoculações Seriadas , Virulência
5.
PLoS Pathog ; 14(1): e1006804, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29304169

RESUMO

Resistance-nodulation-division (RND) efflux systems are ubiquitous transporters in Gram-negative bacteria that are essential for antibiotic resistance. The RND efflux systems also contribute to diverse phenotypes independent of antimicrobial resistance, but the mechanism by which they affect most of these phenotypes is unclear. This is the case in Vibrio cholerae where the RND systems function in antimicrobial resistance and virulence factor production. Herein, we investigated the linkage between RND efflux and V. cholerae virulence. RNA sequencing revealed that the loss of RND efflux affected the activation state of periplasmic sensing systems including the virulence regulator ToxR. Activation of ToxR in an RND null mutant resulted in ToxR-dependent transcription of the LysR-family regulator leuO. Increased leuO transcription resulted in the repression of the ToxR virulence regulon and attenuated virulence factor production. Consistent with this, leuO deletion restored virulence factor production in an RND-null mutant, but not its ability to colonize infant mice; suggesting that RND efflux was epistatic to virulence factor production for colonization. The periplasmic sensing domain of ToxR was required for the induction of leuO transcription in the RND null mutant, suggesting that ToxR responded to metabolites that accumulated in the periplasm. Our results suggest that ToxR represses virulence factor production in response to metabolites that are normally effluxed from the cell by the RND transporters. We propose that impaired RND efflux results in periplasmic metabolite accumulation, which then activates periplasmic sensors including ToxR and two-component regulatory systems to initiate the expression of adaptive responses.


Assuntos
Adaptação Fisiológica/fisiologia , Proteínas de Bactérias/fisiologia , Farmacorresistência Bacteriana , Proteínas de Membrana Transportadoras/fisiologia , Proteínas Periplásmicas/fisiologia , Vibrio cholerae , Fatores de Virulência/metabolismo , Adaptação Fisiológica/genética , Animais , Animais Recém-Nascidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica , Proteínas de Membrana Transportadoras/genética , Camundongos , Organismos Geneticamente Modificados , Proteínas Periplásmicas/genética , Proteínas Periplásmicas/metabolismo , Vibrio cholerae/genética , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidade , Fatores de Virulência/genética
6.
J Infect Dis ; 216(7): 899-906, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-28973366

RESUMO

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.


Assuntos
Infecções por Bordetella/microbiologia , Infecções por Bordetella/transmissão , Bordetella bronchiseptica/metabolismo , Polissacarídeos Bacterianos/metabolismo , Animais , Feminino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mutação , Polissacarídeos Bacterianos/genética
7.
PLoS Biol ; 15(4): e2000420, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28403138

RESUMO

Multiple lines of evidence suggest that Bordetella species have a significant life stage outside of the mammalian respiratory tract that has yet to be defined. The Bordetella virulence gene (BvgAS) two-component system, a paradigm for a global virulence regulon, controls the expression of many "virulence factors" expressed in the Bvg positive (Bvg+) phase that are necessary for successful respiratory tract infection. A similarly large set of highly conserved genes are expressed under Bvg negative (Bvg-) phase growth conditions; however, these appear to be primarily expressed outside of the host and are thus hypothesized to be important in an undefined extrahost reservoir. Here, we show that Bvg- phase genes are involved in the ability of Bordetella bronchiseptica to grow and disseminate via the complex life cycle of the amoeba Dictyostelium discoideum. Unlike bacteria that serve as an amoeba food source, B. bronchiseptica evades amoeba predation, survives within the amoeba for extended periods of time, incorporates itself into the amoeba sori, and disseminates along with the amoeba. Remarkably, B. bronchiseptica continues to be transferred with the amoeba for months, through multiple life cycles of amoebae grown on the lawns of other bacteria, thus demonstrating a stable relationship that allows B. bronchiseptica to expand and disperse geographically via the D. discoideum life cycle. Furthermore, B. bronchiseptica within the sori can efficiently infect mice, indicating that amoebae may represent an environmental vector within which pathogenic bordetellae expand and disseminate to encounter new mammalian hosts. These data identify amoebae as potential environmental reservoirs as well as amplifying and disseminating vectors for B. bronchiseptica and reveal an important role for the Bvg- phase in these interactions.


Assuntos
Infecções por Bordetella/transmissão , Bordetella bronchiseptica/fisiologia , Dictyostelium/crescimento & desenvolvimento , Animais , Infecções por Bordetella/microbiologia , Bordetella bronchiseptica/patogenicidade , Dictyostelium/microbiologia , Vetores de Doenças , Estágios do Ciclo de Vida , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Fatores de Virulência/genética
8.
Artigo em Inglês | MEDLINE | ID: mdl-29322035

RESUMO

The genus Bordetella comprises several bacterial species that colonize the respiratory tract of mammals. It includes B. pertussis, a human-restricted pathogen that is the causative agent of Whooping Cough. In contrast, the closely related species B. bronchiseptica colonizes a broad range of animals as well as immunocompromised humans. Recent metagenomic studies have identified known and novel bordetellae isolated from different environmental sources, providing a new perspective on their natural history. Using phylogenetic analysis, we have shown that human and animal pathogenic bordetellae have most likely evolved from ancestors that originated from soil and water. Our recent study found that B. bronchiseptica can evade amoebic predation and utilize Dictyostelium discoideum as an expansion and transmission vector, which suggests that the evolutionary pressure to evade the amoebic predator enabled the rise of bordetellae as respiratory pathogens. Interactions with amoeba may represent the starting point for bacterial adaptation to eukaryotic cells. However, as bacteria evolve and adapt to a novel host, they can become specialized and restricted to a specific host. B. pertussis is known to colonize and cause infection only in humans, and this specialization to a closed human-to-human lifecycle has involved genome reduction and the loss of ability to utilize amoeba as an environmental reservoir. The discoveries from studying the interaction of Bordetella species with amoeba will elicit a better understanding of the evolutionary history of these and other important human pathogens.


Assuntos
Adaptação Biológica , Infecções por Bordetella/microbiologia , Bordetella/genética , Bordetella/patogenicidade , Dictyostelium/microbiologia , Microbiologia Ambiental , Infecções Respiratórias/microbiologia , Animais , Bordetella/classificação , Infecções por Bordetella/veterinária , Humanos , Filogenia , Infecções Respiratórias/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...