Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
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
2.
Emerg Infect Dis ; 27(6): 1561-1566, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34014152

RESUMO

Recent reemergence of pertussis (whooping cough) in highly vaccinated populations and rapid expansion of Bordetella pertussis strains lacking pertactin (PRN), a common acellular vaccine antigen, have raised the specter of vaccine-driven evolution and potential return of what was once the major killer of children. The discovery that most circulating B. pertussis strains in the United States have acquired new and independent disruptive mutations in PRN is compelling evidence of strong selective pressure. However, the other 4 antigens included in acellular vaccines do not appear to be selected against so rapidly. We consider 3 aspects of PRN that distinguish it from other vaccine antigens, which might, individually or collectively, explain why only this antigen is being precipitously eliminated. An understanding of the increase in PRN-deficient strains should provide useful information for the current search for new protective antigens and provide broader lessons for the design of improved subunit vaccines.


Assuntos
Bordetella pertussis , Coqueluche , Proteínas da Membrana Bacteriana Externa , Criança , Humanos , Vacina contra Coqueluche , Fatores de Virulência de Bordetella
3.
Emerg Infect Dis ; 27(8): 2107-2116, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34286682

RESUMO

Conventional pertussis animal models deliver hundreds of thousands of Bordetella pertussis bacteria deep into the lungs, rapidly inducing severe pneumonic pathology and a robust immune response. However, human infections usually begin with colonization and growth in the upper respiratory tract. We inoculated only the nasopharynx of mice to explore the course of infection in a more natural exposure model. Nasopharyngeal colonization resulted in robust growth in the upper respiratory tract but elicited little immune response, enabling prolonged and persistent infection. Immunization with human acellular pertussis vaccine, which prevents severe lung infections in the conventional pneumonic infection model, had little effect on nasopharyngeal colonization. Our infection model revealed that B. pertussis can efficiently colonize the mouse nasopharynx, grow and spread within and between respiratory organs, evade robust host immunity, and persist for months. This experimental approach can measure aspects of the infection processes not observed in the conventional pneumonic infection model.


Assuntos
Infecções por Bordetella , Coqueluche , Animais , Bordetella pertussis , Evasão da Resposta Imune , Camundongos , Nasofaringe , Vacina contra Coqueluche , Coqueluche/prevenção & controle
5.
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
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.
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
9.
Appl Environ Microbiol ; 84(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29269496

RESUMO

Francisella tularensis subsp. holarctica is found in North America and much of Europe and causes the disease tularemia in humans and animals. An aquatic cycle has been described for this subspecies, which has caused waterborne outbreaks of tularemia in at least 10 countries. In this study, we sought to identify the mechanosensitive channel(s) required for the bacterium to survive the transition from mammalian hosts to freshwater, which is likely essential for the transmission of the bacterium between susceptible hosts. A single 165-amino-acid MscS-type mechanosensitive channel (FtMscS) was found to protect F. tularensis subsp. holarctica from hypoosmotic shock, despite lacking much of the cytoplasmic vestibule domain found in well-characterized MscS proteins from other organisms. The deletion of this channel did not affect virulence within the mammalian host; however, FtMscS was required to survive the transition from the host niche to freshwater. The deletion of FtMscS did not alter the sensitivity of F. tularensis subsp. holarctica to detergents, H2O2, or antibiotics, suggesting that the role of FtMscS is specific to protection from hypoosmotic shock. The deletion of FtMscS also led to a reduced average cell size without altering gross cell morphology. The mechanosensitive channel identified and characterized in this study likely contributes to the transmission of tularemia between hosts by allowing the bacterium to survive the transition from mammalian hosts to freshwater.IMPORTANCE The contamination of freshwater by Francisella tularensis subsp. holarctica has resulted in a number of outbreaks of tularemia. Invariably, the contamination originates from the carcasses or excreta of infected animals and thus involves an abrupt osmotic downshock as the bacteria enter freshwater. How F. tularensis survives this drastic change in osmolarity has not been clear, but here we report that a single mechanosensitive channel protects the bacterium from osmotic downshock. This channel is functional despite lacking much of the cytoplasmic vestibule domain that is present in better-studied organisms such as Escherichia coli; this report builds on previous studies that have suggested that parts of this domain are dispensable for downshock protection. These findings extend our understanding of the aquatic cycle and ecological persistence of F. tularensis, with further implications for mechanosensitive channel biology.


Assuntos
Francisella tularensis/fisiologia , Água Doce , Mecanotransdução Celular/fisiologia , Estresse Salino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Organismos Livres de Patógenos Específicos
10.
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
11.
Can J Urol ; 24(4): 8883-8889, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28832305

RESUMO

INTRODUCTION: To determine if a povidone iodine rectal preparation (PIRP) reduces rates of bacteriuria and bacteremia following transrectal ultrasound guided prostate needle biopsy (TRUS PNB). MATERIALS AND METHODS: Men undergoing TRUS PNB were prospectively enrolled in a study comparing the impact of PIRP versus standard of care (two pills of ciprofloxacin 500 mg). Urine, blood, and rectal cultures were obtained 30 minutes post-procedure with colony forming units (CFUs) determined after 48 hours. Patients were called 7 and 30 days post-procedure to evaluate for infections. RESULTS: A total of 150 men were accrued into this study including 95 receiving PIRP and 55 the standard of care. Two-thirds of patients were undergoing an initial biopsy, 19% used antibiotics within the previous 6 months, and median number of biopsy cores was 14. There were no differences between the two cohorts with respect to baseline or biopsy characteristics. In the PIRP cohort, rectal cultures before and after PIRP administration noted a 97.2% reduction in microorganism colonies (2.4 x 10 5 CFU/mL versus 6.7 x 10³CFU/mL, p < 0.001). Mean urine bacterial counts following TRUS PNB were 1 CFU/mL for PIRP versus 7 CFU/mL for standard cohort (p < 0.001). Mean serum bacterial counts following TRUS PNB were 0 CFU/mL for PIRP versus 3 CFU/mL for standard of care (p = 0.01). One patient in the PIRP cohort (1.1%) developed post-biopsy sepsis while 3 (5.5%) in the standard cohort had an infectious complication (1 UTI, 2 sepsis). CONCLUSION: A PIRP regimen reduced bacteruria and bacteremia following TRUS PNB.


Assuntos
Anti-Infecciosos Locais/administração & dosagem , Bacteriemia/prevenção & controle , Bacteriúria/prevenção & controle , Complicações Pós-Operatórias/microbiologia , Complicações Pós-Operatórias/prevenção & controle , Povidona-Iodo/administração & dosagem , Próstata/patologia , Neoplasias da Próstata/patologia , Administração Tópica , Bacteriemia/epidemiologia , Bacteriúria/epidemiologia , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/epidemiologia , Estudos Prospectivos , Reto
12.
Antimicrob Agents Chemother ; 60(6): 3276-82, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26953190

RESUMO

Bacteria require at least one pathway to rescue ribosomes stalled at the ends of mRNAs. The primary pathway for ribosome rescue is trans-translation, which is conserved in >99% of sequenced bacterial genomes. Some species also have backup systems, such as ArfA or ArfB, which can rescue ribosomes in the absence of sufficient trans-translation activity. Small-molecule inhibitors of ribosome rescue have broad-spectrum antimicrobial activity against bacteria grown in liquid culture. These compounds were tested against the tier 1 select agent Francisella tularensis to determine if they can limit bacterial proliferation during infection of eukaryotic cells. The inhibitors KKL-10 and KKL-40 exhibited exceptional antimicrobial activity against both attenuated and fully virulent strains of F. tularensis in vitro and during ex vivo infection. Addition of KKL-10 or KKL-40 to macrophages or liver cells at any time after infection by F. tularensis prevented further bacterial proliferation. When macrophages were stimulated with the proinflammatory cytokine gamma interferon before being infected by F. tularensis, addition of KKL-10 or KKL-40 reduced intracellular bacteria by >99%, indicating that the combination of cytokine-induced stress and a nonfunctional ribosome rescue pathway is fatal to F. tularensis Neither KKL-10 nor KKL-40 was cytotoxic to eukaryotic cells in culture. These results demonstrate that ribosome rescue is required for F. tularensis growth at all stages of its infection cycle and suggest that KKL-10 and KKL-40 are good lead compounds for antibiotic development.


Assuntos
Antibacterianos/farmacologia , Francisella tularensis/efeitos dos fármacos , Oxidiazóis/farmacologia , Ribossomos/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Interferon gama/farmacologia , Fígado/microbiologia , Macrófagos/microbiologia , Camundongos , Testes de Sensibilidade Microbiana , Células RAW 264.7 , Virulência/efeitos dos fármacos
14.
World J Urol ; 32(4): 905-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24682238

RESUMO

PURPOSE: The purpose of the study was to evaluate whether a peri-procedural povidone-iodine rectal preparation (PIRP) prior to transrectal ultrasound-guided prostate needle biopsy (TRUS PNB) can reduce microorganism colony counts and infectious complications. METHODS: Our institutional TRUS PNB database was reviewed to identify infectious post-biopsy complications (defined as fever >38.5 °C with positive culture). The last 570 biopsy patients were divided into those administered only preoperative oral and/or parenteral antibiotics (n = 456; chronologically cohorts A-D) versus men receiving peri-procedural PIRP in conjunction with standard preoperative antibiotics (n = 114; cohort E). Rectal cultures were obtained in the PIRP cohort to quantify changes in microorganism colony counts. RESULTS: Mean baseline PSA for patients was 11.6 ng/ml, 63 % were undergoing an initial biopsy, and 17 % had documented use of antibiotic therapy within the previous 6 months. A reduction in infectious complications was observed when comparing the conventional antibiotic (cohorts A-D) versus PIRP (cohort E) group (1.8 vs. 0 %), with the largest magnitude of decline occurring in the concurrent contemporary cohorts (cohort D-5.3 % vs. cohort E-0 %, p = 0.03). Rectal cultures obtained in 92 men before and after PIRP administration noted a 97 % reduction in microorganism colonies (2.1 × 10(5) vs. 6.3 × 10(3) CFU/ml, p < 0.001). No adverse reactions to the PIRP were reported by patients 7 days post-biopsy. CONCLUSIONS: Peri-procedural PIRP decreased microorganism colony counts and effectively reduced infectious complications following TRUS PNB. This safe, cheap, and simple strategy may be a reasonable alternative to systemic or targeted antibiotic therapy to reduce post-biopsy infections.


Assuntos
Infecções Bacterianas/prevenção & controle , Biópsia por Agulha/métodos , Povidona-Iodo/uso terapêutico , Próstata/microbiologia , Próstata/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anti-Infecciosos Locais/uso terapêutico , Bactérias/isolamento & purificação , Infecções Bacterianas/epidemiologia , Humanos , Biópsia Guiada por Imagem , Incidência , Masculino , Pessoa de Meia-Idade , Assistência Perioperatória/métodos , Próstata/diagnóstico por imagem , Estudos Retrospectivos , Ultrassonografia
15.
Int J Pediatr Otorhinolaryngol ; 176: 111814, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38101097

RESUMO

OBJECTIVE: To review and summarize recently published key articles on the topics of animal models, cell culture studies, tissue biomedical engineering and regeneration, and new models in relation to otitis media (OM). DATA SOURCE: Electronic databases: PubMed, National Library of Medicine, Ovid Medline. REVIEW METHODS: Key topics were assigned to the panel participants for identification and detailed evaluation. The PubMed reviews were focused on the period from June 2019 to June 2023, in any of the objective subject(s) or keywords listed above, noting the relevant references relating to these advances with a global overview and noting areas of recommendation(s). The final manuscript was prepared with input from all panel members. CONCLUSIONS: In conclusion, ex vivo and in vivo OM research models have seen great advancements in the past 4 years. From the usage of novel genetic and molecular tools to the refinement of in vivo inducible and spontaneous mouse models, to the introduction of a wide array of reliable middle ear epithelium (MEE) cell culture systems, the next five years are likely to experience exponential growth in OM pathophysiology discoveries. Moreover, advances in these systems will predictably facilitate rapid means for novel molecular therapeutic studies.


Assuntos
Otite Média , Animais , Camundongos , Humanos , Otite Média/tratamento farmacológico , Orelha Média , Modelos Animais de Doenças , Engenharia Biomédica , Técnicas de Cultura de Células
16.
Front Cell Infect Microbiol ; 13: 1288057, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38125908

RESUMO

The efficacy of the adaptive immune system in the middle ear (ME) is well established, but the mechanisms are not as well defined as those of gastrointestinal or respiratory tracts. While cellular elements of the adaptive response have been detected in the MEs following infections (or intranasal immunizations), their specific contributions to protecting the organ against reinfections are unknown. How immune protection mechanisms of the MEs compares with those in the adjacent and attached upper and lower respiratory airways remains unclear. To address these knowledge gaps, we used an established mouse respiratory infection model that we recently showed also involves ME infections. Bordetella bronchiseptica delivered to the external nares of mice in tiny numbers very efficiently infects the respiratory tract and ascends the Eustachian tube to colonize and infect the MEs, where it causes severe but acute inflammation resembling human acute otitis media (AOM). Since this AOM naturally resolves, we here examine the immunological mechanisms that clear infection and protect against subsequent infection, to guide efforts to induce protective immunity in the ME. Our results show that once the MEs are cleared of a primary B. bronchiseptica infection, the convalescent organ is strongly protected from reinfection by the pathogen despite its persistence in the upper respiratory tract, suggesting important immunological differences in these adjacent and connected organs. CD4+ and CD8+ T cells trafficked to the MEs following infection and were necessary to robustly protect against secondary challenge. Intranasal vaccination with heat killed B. bronchiseptica conferred robust protection against infection to the MEs, even though the nasopharynx itself was only partially protected. These data establish the MEs as discrete effector sites of adaptive immunity and shows that effective protection in the MEs and the respiratory tract is significantly different. This model system allows the dissection of immunological mechanisms that can prevent bacteria in the nasopharynx from ascending the ET to colonize the ME.


Assuntos
Infecções por Bordetella , Bordetella bronchiseptica , Otite Média , Infecções Respiratórias , Humanos , Animais , Camundongos , Infecções por Bordetella/microbiologia , Sistema Respiratório , Infecções Respiratórias/microbiologia , Otite Média/prevenção & controle , Orelha Média
17.
mSphere ; 7(1): e0089221, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35196124

RESUMO

Acute pathogens such as Bordetella pertussis can cause severe disease but are ultimately cleared by the immune response. This has led to the accepted paradigm that convalescent immunity is optimal and therefore broadly accepted as the "gold standard" against which vaccine candidates should be compared. However, successful pathogens like B. pertussis have evolved multiple mechanisms for suppressing and evading host immunity, raising the possibility that disruption of these mechanisms could result in substantially stronger or better immunity. Current acellular B. pertussis vaccines, delivered in a 5-dose regimen, induce only short-term immunity against disease and even less against colonization and transmission. Importantly, they provide modest protection against other Bordetella species that cause substantial human disease. A universal vaccine that protects against the three classical Bordetella spp. could decrease the burden of whooping cough-like disease in humans and other animals. Our recent work demonstrated that Bordetella spp. suppress host inflammatory responses and that disrupting the regulation of immunosuppressive mechanisms can allow the host to generate substantially stronger sterilizing immunity against the three classical Bordetella spp. Here, we identify immune parameters impacted by Bordetella species immunomodulation, including the generation of robust Th17 and B cell memory responses. Disrupting immunomodulation augmented the immune response, providing strong protection against the prototypes of all three classical Bordetella spp. as well as recent clinical isolates. Importantly, the protection in mice lasted for at least 15 months and was associated with recruitment of high numbers of B and T cells in the lungs as well as enhanced Th17 mucosal responses and persistently high titers of antibodies. These findings demonstrate that disrupting bacterial immunomodulatory pathways can generate much stronger and more protective immune responses to infection, with important implications for the development of better vaccines. IMPORTANCE Infectious diseases are a major cause of morbidity and mortality in the United States, accounting for over 40 million hospitalizations since 1998. Therefore, novel vaccine strategies are imperative, which can be improved with a better understanding of the mechanisms that bacteria utilize to suppress host immunity, a key mechanism for establishing colonization. Bordetella spp., the causative agents of whooping cough, suppress host immunity, which allows for persistent colonization. We discovered a regulator of a bacterial immunosuppressive pathway, which, when mutated in Bordetella spp., allows for rapid clearance of infection and subsequent generation of protective immunity for at least 15 months. After infection with the mutant strain, mice exhibited sterilizing immunity against the three classical Bordetella spp., suggesting that the immune response can be both stronger and cross-protective. This work presents a strategy for vaccine development that can be applied to other immunomodulatory pathogens.


Assuntos
Coqueluche , Animais , Bordetella pertussis , Modelos Animais de Doenças , Camundongos , Vacina contra Coqueluche , Vacinas Atenuadas , Coqueluche/microbiologia , Coqueluche/prevenção & controle
18.
Front Cell Infect Microbiol ; 12: 798317, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35223538

RESUMO

A variety of bacteria have evolved the ability to interact with environmental phagocytic predators such as amoebae, which may have facilitated their subsequent interactions with phagocytes in animal hosts. Our recent study found that the animal pathogen Bordetella bronchiseptica can evade predation by the common soil amoeba Dictyostelium discoideum, survive within, and hijack its complex life cycle as a propagation and dissemination vector. However, it is uncertain whether the mechanisms allowing interactions with predatory amoebae are conserved among Bordetella species, because divergence, evolution, and adaptation to different hosts and ecological niches was accompanied by acquisition and loss of many genes. Here we tested 9 diverse Bordetella species in three assays representing distinct aspects of their interactions with D. discoideum. Several human and animal pathogens retained the abilities to survive within single-celled amoeba, to inhibit amoebic plaque expansion, and to translocate with amoebae to the fruiting body and disseminate along with the fruiting body. In contrast, these abilities were partly degraded for the bird pathogen B. avium, and for the human-restricted species B. pertussis and B. parapertussis. Interestingly, a different lineage of B. parapertussis only known to infect sheep retained the ability to interact with D. discoideum, demonstrating that these abilities were lost in multiple lineages independently, correlating with niche specialization and recent rapid genome decay apparently mediated by insertion sequences. B. petrii has been isolated sporadically from diverse human and environmental sources, has acquired insertion sequences, undergone genome decay and has also lost the ability to interact with amoebae, suggesting some specialization to some unknown niche. A genome-wide association study (GWAS) identified a set of genes that are potentially associated with the ability to interact with D. discoideum. These results suggest that massive gene loss associated with specialization of some Bordetella species to a closed life cycle in a particular host was repeatedly and independently accompanied by loss of the ability to interact with amoebae in an environmental niche.


Assuntos
Amoeba , Bordetella bronchiseptica , Bordetella , Dictyostelium , Amoeba/microbiologia , Animais , Bordetella/genética , Bordetella bronchiseptica/genética , Dictyostelium/microbiologia , Estudo de Associação Genômica Ampla , Ovinos/genética
19.
Front Cell Infect Microbiol ; 12: 795230, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35360099

RESUMO

Chronic otitis media (COM) is the long-term infection and inflammation of the middle ears typically caused by upper respiratory tract pathogens that are able to ascend the Eustachian tube. Our understanding of contributing factors is limited because human otopathogens cannot naturally colonize or persist in the middle ears of mice. We recently described a natural COM in mice caused by Bordetella pseudohinzii and proposed this as an experimental system to study bacterial mechanisms of immune evasion that allow persistent infection of the middle ear. Here we describe a novel pertussis toxin (PTx)-like factor unique to B. pseudohinzii, apparently acquired horizontally, that is associated with its particularly efficient persistence and pathogenesis. The catalytic subunit of this toxin, PsxA, has conserved catalytic sites and substantial predicted structural homology to pertussis toxin catalytic subunit PtxA. Deletion of the gene predicted to encode the catalytic subunit, psxA, resulted in a significant decrease in persistence in the middle ears. The defect was not observed in mice lacking T cells, indicating that PsxA is necessary for persistence only when T cells are present. These results demonstrate the role of a novel putative toxin in the persistence of B. pseudohinzii and its generation of COM. This PsxA-mediated immune evasion strategy may similarly be utilized by human otopathogens, via other PTx-like toxins or alternative mechanisms to disrupt critical T cell functions necessary to clear bacteria from the middle ear. This work demonstrates that this experimental system can allow for the detailed study of general strategies and specific mechanisms that otopathogens use to evade host immune responses to persist in the middle ear to cause COM.


Assuntos
Otite Média , Animais , Bactérias , Orelha Média/microbiologia , Inflamação , Camundongos , Otite Média/microbiologia , Toxina Pertussis
20.
Dis Model Mech ; 15(5)2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35311902

RESUMO

Pertussis (whooping cough) is a highly transmissible human respiratory disease caused by Bordetella pertussis, a human-restricted pathogen. Animal models generally involve pneumonic infections induced by depositing large numbers of bacteria in the lungs of mice. These models have informed us about the molecular pathogenesis of pertussis and guided development of vaccines that successfully protect against severe disease. However, they bypass the catarrhal stage of the disease, when bacteria first colonize and initially grow in the upper respiratory tract. This is a critical and highly transmissible stage of the infection that current vaccines do not prevent. Here, we demonstrate a model system in which B. pertussis robustly and persistently infects the nasopharynx of TLR4-deficient mice, inducing localized inflammation, neutrophil recruitment and mucus production as well as persistent shedding and occasional transmission to cage mates. This novel experimental system will allow the study of the contributions of bacterial factors to colonization of and shedding from the nasopharynx, as occurs during the catarrhal stage of pertussis, and interventions that might better control the ongoing circulation of pertussis.


Assuntos
Infecções Respiratórias , Coqueluche , Animais , Bordetella pertussis , Pulmão/microbiologia , Camundongos , Vacina contra Coqueluche , Coqueluche/microbiologia , Coqueluche/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA