Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 38(2): 110202, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021083

RESUMEN

Streptococcus pneumoniae is the primary cause of community-acquired bacterial pneumonia with rates of penicillin and multidrug-resistance exceeding 80% and 40%, respectively. The innate immune response generates a variety of antimicrobial agents to control infection, including zinc stress. Here, we characterize the impact of zinc intoxication on S. pneumoniae, observing disruptions in central carbon metabolism, lipid biogenesis, and peptidoglycan biosynthesis. Characterization of the pivotal peptidoglycan biosynthetic enzyme GlmU indicates a sensitivity to zinc inhibition. Disruption of the sole zinc efflux pathway, czcD, renders S. pneumoniae highly susceptible to ß-lactam antibiotics. To dysregulate zinc homeostasis in the wild-type strain, we investigated the safe-for-human-use ionophore 5,7-dichloro-2-[(dimethylamino)methyl]quinolin-8-ol (PBT2). PBT2 rendered wild-type S. pneumoniae strains sensitive to a range of antibiotics. Using an invasive ampicillin-resistant strain, we demonstrate in a murine pneumonia infection model the efficacy of PBT2 + ampicillin treatment. These findings present a therapeutic modality to break antibiotic resistance in multidrug-resistant S. pneumoniae.


Asunto(s)
Resistencia a la Ampicilina/fisiología , Streptococcus pneumoniae/metabolismo , Zinc/metabolismo , Ampicilina/farmacología , Resistencia a la Ampicilina/genética , Animales , Antibacterianos/farmacología , Clioquinol/análogos & derivados , Clioquinol/farmacología , Modelos Animales de Enfermedad , Femenino , Homeostasis , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Neumonía
2.
mSystems ; 5(3)2020 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-32398278

RESUMEN

Experimental evolution is a powerful technique to understand how populations evolve from selective pressures imparted by the surrounding environment. With the advancement of whole-population genomic sequencing, it is possible to identify and track multiple contending genotypes associated with adaptations to specific selective pressures. This approach has been used repeatedly with model species in vitro, but only rarely in vivo Herein we report results of replicate experimentally evolved populations of Streptococcus pneumoniae propagated by repeated murine nasal colonization with the aim of identifying gene products under strong selection as well as the population genetic dynamics of infection cycles. Frameshift mutations in one gene, dltB, responsible for incorporation of d-alanine into teichoic acids on the bacterial surface, evolved repeatedly and swept to high frequency. Targeted deletions of dltB produced a fitness advantage during initial nasal colonization coupled with a corresponding fitness disadvantage in the lungs during pulmonary infection. The underlying mechanism behind the fitness trade-off between these two niches was found to be enhanced adherence to respiratory cells balanced by increased sensitivity to host-derived antimicrobial peptides, a finding recapitulated in the murine model. Additional mutations that are predicted to affect trace metal transport, central metabolism, and regulation of biofilm production and competence were also selected. These data indicate that experimental evolution can be applied to murine models of pathogenesis to gain insight into organism-specific tissue tropisms.IMPORTANCE Evolution is a powerful force that can be experimentally harnessed to gain insight into how populations evolve in response to selective pressures. Herein we tested the applicability of experimental evolutionary approaches to gain insight into how the major human pathogen Streptococcus pneumoniae responds to repeated colonization events using a murine model. These studies revealed the population dynamics of repeated colonization events and demonstrated that in vivo experimental evolution resulted in highly reproducible trajectories that reflect the environmental niche encountered during nasal colonization. Mutations impacting the surface charge of the bacteria were repeatedly selected during colonization and provided a fitness benefit in this niche that was counterbalanced by a corresponding fitness defect during lung infection. These data indicate that experimental evolution can be applied to models of pathogenesis to gain insight into organism-specific tissue tropisms.

3.
Sci Rep ; 9(1): 19360, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852944

RESUMEN

Asthma is a chronic airways condition that can be exacerbated during respiratory infections. Our previous work, together with epidemiologic findings that asthmatics were less likely to suffer from severe influenza during the 2009 pandemic, suggest that additional complications of influenza such as increased susceptibility to bacterial superinfection, may be mitigated in allergic hosts. To test this hypothesis, we developed a murine model of 'triple-disease' in which mice rendered allergic to Aspergillus fumigatus were co-infected with influenza A virus and Streptococcus pneumoniae seven days apart. Significant alterations to known synergistic effects of co-infection were noted in the allergic mice including reduced morbidity and mortality, bacterial burden, maintenance of alveolar macrophages, and reduced lung inflammation and damage. The lung microbiome of allergic mice differed from that of non-allergic mice during co-infection and antibiotic-induced perturbation to the microbiome rendered allergic animals susceptible to severe morbidity. Our data suggest that responses to co-infection in allergic hosts likely depends on the immune and microbiome states and that antibiotics should be used with caution in individuals with underlying chronic lung disease.


Asunto(s)
Coinfección/microbiología , Coinfección/virología , Hipersensibilidad/microbiología , Inflamación/microbiología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Pulmón/microbiología , Microbiota , Streptococcus pneumoniae/fisiología , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Biodiversidad , Coinfección/tratamiento farmacológico , Coinfección/inmunología , Hipersensibilidad/complicaciones , Hipersensibilidad/tratamiento farmacológico , Hipersensibilidad/inmunología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Modelos Biológicos , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/prevención & control , Neumonía Bacteriana/complicaciones , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/prevención & control
4.
Immunobiology ; 222(12): 1064-1073, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28889999

RESUMEN

Humoral immunity serve dual functions of direct pathogen neutralization and enhancement of leukocyte function. Antibody classes are determined by antigen triggers, and the resulting antibodies can contribute to disease pathogenesis and host defense. Although asthma and influenza are immunologically distinct diseases, since we have found that allergic asthma exacerbation promotes antiviral host responses to influenza A virus, we hypothesized that humoral immunity may contribute to allergic host protection during influenza. C57BL/6J mice sensitized and challenged with Aspergillus fumigatus (or not) were infected with pandemic influenza A/CA/04/2009 virus. Negative control groups included naïve mice, and mice with only 'asthma' or influenza. Concentrations of antibodies were quantified by ELISA, and in situ localization of IgA- and IgE-positive cells in the lungs was determined by immunohistochemistry. The number and phenotype of B cells in spleens and mediastinal lymph nodes were determined by flow cytometry at predetermined timepoints after virus infection until viral clearance. Mucosal and systemic antibodies remained elevated in mice with asthma and influenza with prominent production of IgE and IgA compared to influenza-only controls. B cell expansion was prominent in the mediastinal lymph nodes of allergic mice during influenza where most cells produced IgG1 and IgA. Although allergy-skewed B cell responses dominated in mice with allergic airways inflammation during influenza virus infection, virus-specific antibodies were also induced. Future studies are required to identify the mechanisms involved with B cell activation and function in allergic hosts facing respiratory viral infections.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Asma/inmunología , Linfocitos B/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Anticuerpos Antivirales/sangre , Femenino , Humanos , Inmunidad Humoral , Inmunoglobulina A/metabolismo , Inmunoglobulina E/sangre , Ratones , Ratones Endogámicos C57BL , Neumonía , Bazo/patología
5.
Vaccine ; 32(42): 5405-10, 2014 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25131738

RESUMEN

Pneumonia is the leading killer of children and disproportionately affects developing countries. Vaccination campaigns against Streptococcus pneumoniae, the leading cause of pneumonia, have recently been launched with a new conjugate vaccine in Africa. Using a mouse model, we assessed the potential role that the high burden of helminth infections in the countries targeted for vaccine might have on vaccine effectiveness. Mice vaccinated with either commercial conjugate or purified polysaccharide vaccines had impaired antibody responses if they were chronically infected with Taenia crassiceps. This translated to increased susceptibility to pneumococcal pneumonia and high mortality compared to helminth-negative vaccinated animals, which were fully protected from disease and death. Antibodies taken from Taenia-infected, vaccinated mice were unable to effectively opsonize S. pneumoniae for killing by alveolar macrophages, and did not protect against pneumococcal challenge when adoptively transferred into naïve animals. These data may have implications for vaccination programs in countries endemic with helminths.


Asunto(s)
Helmintiasis/complicaciones , Vacunas Neumococicas/inmunología , Neumonía Neumocócica/prevención & control , Traslado Adoptivo , Animales , Anticuerpos Antibacterianos/sangre , Especificidad de Anticuerpos , Cisticercosis/complicaciones , Modelos Animales de Enfermedad , Femenino , Macrófagos Alveolares/citología , Ratones Endogámicos BALB C , Fagocitosis , Taenia , Insuficiencia del Tratamiento , Vacunas Conjugadas/inmunología
6.
EMBO Mol Med ; 6(1): 141-54, 2014 01.
Artículo en Inglés | MEDLINE | ID: mdl-24408968

RESUMEN

Acute otitis media (AOM) caused by Streptococcus pneumoniae remains one of the most common infectious diseases worldwide despite widespread vaccination. A major limitation of the currently licensed pneumococcal vaccines is the lack of efficacy against mucosal disease manifestations such as AOM, acute bacterial sinusitis and pneumonia. We sought to generate a novel class of live vaccines that (1) retain all major antigenic virulence proteins yet are fully attenuated and (2) protect against otitis media. A live vaccine candidate based on deletion of the signal recognition pathway component ftsY induced potent, serotype-independent protection against otitis media, sinusitis, pneumonia and invasive pneumococcal disease. Protection was maintained in animals coinfected with influenza virus, but was lost if mice were depleted of CD4(+) T cells at the time of vaccination. The live vaccine induced a strong serum IgG2a and IgG2b response that correlated with CD4(+) T-cell mediated class switching. Deletion of genes required for microbial adaptation to the host environment is a novel live attenuated vaccine strategy yielding the first experimental vaccine effective against pneumococcal otitis media.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Otitis Media/prevención & control , Vacunas Neumococicas/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD4-Positivos/citología , Chinchilla , Modelos Animales de Enfermedad , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos BALB C , Otitis Media/mortalidad , Otitis Media/patología , Serotipificación , Sinusitis/microbiología , Sinusitis/mortalidad , Sinusitis/prevención & control , Streptococcus pneumoniae/metabolismo , Tasa de Supervivencia , Vacunas Atenuadas/inmunología , Factores de Virulencia/inmunología
7.
J Virol ; 86(17): 9035-43, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22674997

RESUMEN

A combination of viral, bacterial, and host factors contributes to the severity and overall mortality associated with influenza virus-bacterium superinfections. To date, the virulence associated with the recently identified influenza virus protein PB1-F2 has been largely defined using models of primary influenza virus infection, with only limited assessment in models of Streptococcus pneumoniae superinfection. Specifically, these studies have incorporated isogenic viruses that differ in the PB1-F2 expressed, but there is still knowledge to be gained from evaluation of natural variants derived from a nonhuman host species (swine). Using this rationale, we developed the hypothesis that naturally occurring viruses expressing variants of genes, like the PB1-F2 gene, can be associated with the severity of secondary bacterial infections. To test this hypothesis, we selected viruses expressing variants in PB1-F2 and evaluated outcomes from superinfection with three distinct Gram-positive respiratory pathogens: Streptococcus pneumoniae, Staphylococcus aureus, and Streptococcus pyogenes. Our results demonstrate that the amino acid residues 62L, 66S, 75R, 79R, and 82L, previously proposed as molecular signatures of PB1-F2 virulence for influenza viruses in the setting of bacterial superinfection, are broadly associated with enhanced pathogenicity in swine in a bacterium-specific manner. Furthermore, truncated PB1-F2 proteins can preferentially increase mortality when associated with Streptococcus pyogenes superinfection. These findings support efforts to increase influenza virus surveillance to consider viral genotypes that could be used to predict increased severity of superinfections with specific Gram-positive respiratory pathogens.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/metabolismo , Gripe Humana/microbiología , Gripe Humana/virología , Staphylococcus aureus/fisiología , Streptococcus/fisiología , Sobreinfección/microbiología , Sobreinfección/virología , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H1N1 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/virología , Porcinos , Enfermedades de los Porcinos/virología , Proteínas Virales/química , Proteínas Virales/genética
8.
J Infect Dis ; 203(6): 880-8, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21278211

RESUMEN

Superinfections from Staphylococcus aureus following influenza are an increasing concern. We assessed several laboratory and clinical strains in a mouse coinfection model with influenza virus. A methicillin-resistant USA300 clone and several recent clinical strains from patients with necrotizing pneumonia caused high mortality following influenza virus infection in mice. Both viral and bacterial lung titers were enhanced during coinfections compared with single infections. However, differences in titers did not correspond with differences in disease outcomes in a comparison of superinfections from a highly pathogenic strain with those from a poorly pathogenic strain. These strains did differ, however, in expression of Panton-Valentine leukocidin and in the degree of inflammatory lung damage each engendered. The viral cytotoxin PB1-F2 contributed to the negative outcomes. These data suggest that additional study of specific bacterial virulence factors involved in the pathogenesis of inflammation and lung damage during coinfections is needed.


Asunto(s)
Virus de la Influenza A/patogenicidad , Gripe Humana/complicaciones , Neumonía Estafilocócica/microbiología , Staphylococcus aureus/patogenicidad , Sobreinfección/microbiología , Animales , Citotoxinas , Modelos Animales de Enfermedad , Femenino , Humanos , Virus de la Influenza A/inmunología , Hígado/patología , Hígado/virología , Pulmón/patología , Pulmón/virología , Staphylococcus aureus Resistente a Meticilina , Ratones , Ratones Endogámicos BALB C , Neumonía Estafilocócica/complicaciones , Reacción en Cadena de la Polimerasa , Bazo/patología , Bazo/virología , Sobreinfección/complicaciones , Análisis de Supervivencia
9.
J Infect Dis ; 202(8): 1287-95, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20822454

RESUMEN

The role of respiratory viruses in the transmission of Streptococcus pneumoniae is poorly understood. Key questions, such as which serotypes are most fit for transmission and disease and whether influenza virus alters these parameters in a serotype-specific manner, have not been adequately studied. In a novel model of transmission in ferrets, we demonstrated that pneumococcal transmission and disease were enhanced if donors had previously been infected with influenza virus. Bacterial titers in nasal wash, the incidence of mucosal and invasive disease, and the percentage of contacts that were infected all increased. In contact ferrets, viral infection increased their susceptibility to S. pneumoniae acquisition both in terms of the percentage infected and the distance over which they could acquire infection. These influenza-mediated effects on colonization, transmission, and disease were dependent on the pneumococcal strain. Overall, these data argue that the relationship between respiratory viral infections, acquisition of pneumococci, and development of disease in humans needs further study to be better understood.


Asunto(s)
Hurones , Interacciones Huésped-Patógeno , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/transmisión , Streptococcus pneumoniae/fisiología , Animales , Modelos Animales de Enfermedad , Perros , Hurones/microbiología , Hurones/virología , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/mortalidad , Infecciones Neumocócicas/virología
10.
J Virol ; 84(8): 4105-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20130054

RESUMEN

Secondary bacterial infections contribute to morbidity and mortality from influenza. Vaccine effectiveness is typically assessed using prevention of influenza, not secondary infections, as an endpoint. We vaccinated mice with formalin-inactivated influenza virus vaccine preparations containing disparate HA and NA proteins and demonstrated an ability to induce the appropriate anti-HA and anti-NA immune profiles. Protection from both primary viral and secondary bacterial infection was demonstrated with vaccine-induced immunity directed toward either the HA or the NA. This finding suggests that immunity toward the NA component of the virion is desirable and should be considered in generation of influenza vaccines.


Asunto(s)
Hemaglutininas Virales/inmunología , Vacunas contra la Influenza/inmunología , Neuraminidasa/inmunología , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/inmunología , Neumonía Bacteriana/prevención & control , Proteínas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C
11.
Scand J Infect Dis ; 40(1): 11-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17852951

RESUMEN

Pneumonia caused by Streptococcus pneumoniae is a significant cause of morbidity and mortality during influenza virus epidemics. We had previously advanced the hypothesis that interactions of pneumococcus with the receptor for platelet activating factor (PAFR) in the lung were facilitated by antecedent influenza virus infection and play a major role in the pathogenesis of bacterial superinfections. Although influenza enhanced the adherence of pneumococci to respiratory epithelial cells in vitro, chemical or antibody-mediated blockade of the PAFR did not affect adherence. In agreement with these data, mice lacking PAFR had similar bacterial loads within the lung compartment when compared to heterozygous littermates and were not protected from secondary pneumococcal pneumonia after influenza. Lack of support for this hypothesis and the observation of enhanced inflammation during secondary pneumococcal pneumonia in mice lacking PAFR may moderate enthusiasm for treatment strategies targeting the interaction of bacteria with PAFR.


Asunto(s)
Adhesión Bacteriana , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/complicaciones , Glicoproteínas de Membrana Plaquetaria/fisiología , Neumonía Neumocócica/complicaciones , Receptores Acoplados a Proteínas G/fisiología , Streptococcus pneumoniae/patogenicidad , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Glicoproteínas de Membrana Plaquetaria/genética , Neumonía Neumocócica/virología , Receptores Acoplados a Proteínas G/genética , Sobreinfección
13.
J Virol ; 78(23): 12817-28, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15542634

RESUMEN

The segmented genome of influenza B virus allows exchange of gene segments between cocirculating strains. Through this process of reassortment, diversity is generated by the mixing of genes between viruses that differ in one or more gene segments. Phylogenetic and evolutionary analyses of all 11 genes of 31 influenza B viruses isolated from 1979 to 2003 were used to study the evolution of whole genomes. All 11 genes diverged into two new lineages prior to 1987. All genes except the NS1 gene were undergoing linear evolution, although the rate of evolution and the degree to which nucleotide changes translated into amino acid changes varied between lineages and by gene. Frequent reassortment generated 14 different genotypes distinct from the gene constellation of viruses circulating prior to 1979. Multiple genotypes cocirculated in some locations, and a sequence of reassortment events over time could not be established. The surprising diversity of the viruses, unrestricted mixing of lineages, and lack of evidence for coevolution of gene segments do not support the hypothesis that the reassortment process is driven by selection for functional differences.


Asunto(s)
Virus de la Influenza B/clasificación , Virus Reordenados/genética , Evolución Biológica , Genotipo , Virus de la Influenza B/genética , Filogenia , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA