Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Immunol ; 204(1): 101-111, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31776202

RESUMEN

Streptococcus pneumoniae is a major cause of pneumonia, wherein infection of respiratory mucosa drives a robust influx of neutrophils. We have previously shown that S. pneumoniae infection of the respiratory epithelium induces the production of the 12-lipoxygenase (12-LOX)-dependent lipid inflammatory mediator hepoxilin A3, which promotes recruitment of neutrophils into the airways, tissue damage, and lethal septicemia. Pneumolysin (PLY), a member of the cholesterol-dependent cytolysin (CDC) family, is a major S. pneumoniae virulence factor that generates ∼25-nm diameter pores in eukaryotic membranes and promotes acute inflammation, tissue damage, and bacteremia. We show that a PLY-deficient S. pneumoniae mutant was impaired in triggering human neutrophil transepithelial migration in vitro. Ectopic production of PLY endowed the nonpathogenic Bacillus subtilis with the ability to trigger neutrophil recruitment across human-cultured monolayers. Purified PLY, several other CDC family members, and the α-toxin of Clostridium septicum, which generates pores with cross-sectional areas nearly 300 times smaller than CDCs, reproduced this robust neutrophil transmigration. PLY non-pore-forming point mutants that are trapped at various stages of pore assembly did not recruit neutrophils. PLY triggered neutrophil recruitment in a 12-LOX-dependent manner in vitro. Instillation of wild-type PLY but not inactive derivatives into the lungs of mice induced robust 12-LOX-dependent neutrophil migration into the airways, although residual inflammation induced by PLY in 12-LOX-deficient mice indicates that 12-LOX-independent pathways also contribute to PLY-triggered pulmonary inflammation. These data indicate that PLY is an important factor in promoting hepoxilin A3-dependent neutrophil recruitment across pulmonary epithelium in a pore-dependent fashion.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Infiltración Neutrófila/inmunología , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/metabolismo , Migración Transendotelial y Transepitelial/inmunología , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/inmunología , Animales , Bacillus subtilis/genética , Bacillus subtilis/patogenicidad , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Línea Celular , Membrana Celular/patología , Clostridium septicum/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/inmunología , Estreptolisinas/genética , Factores de Virulencia/metabolismo
2.
PLoS Pathog ; 7(1): e1001247, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21253576

RESUMEN

Non-typeable Haemophilus influenzae (NTHi), a common commensal of the human pharynx, is also an opportunistic pathogen if it becomes established in the lower respiratory tract (LRT). In comparison to colonizing isolates from the upper airway, LRT isolates, especially those associated with exacerbations of chronic obstructive pulmonary disease, have increased resistance to the complement- and antibody-dependent, bactericidal effect of serum. To define the molecular basis of this resistance, mutants constructed in a serum resistant strain using the mariner transposon were screened for loss of survival in normal human serum. The loci required for serum resistance contribute to the structure of the exposed surface of the bacterial outer membrane. These included loci involved in biosynthesis of the oligosaccharide component of lipooligosaccharide (LOS), and vacJ, which functions with an ABC transporter encoded by yrb genes in retrograde trafficking of phospholipids from the outer to inner leaflet of the cell envelope. Mutations in vacJ and yrb genes reduced the stability of the outer membrane and were associated with increased cell surface hyrophobicity and phospholipid content. Loss of serum resistance in vacJ and yrb mutants correlated with increased binding of natural immunoglobulin M in serum as well as anti-oligosaccharide mAbs. Expression of vacJ and the yrb genes was positively correlated with serum resistance among clinical isolates. Our findings suggest that NTHi adapts to inflammation encountered during infection of the LRT by modulation of its outer leaflet through increased expression of vacJ and yrb genes to minimize recognition by bactericidal anti-oligosaccharide antibodies.


Asunto(s)
Actividad Bactericida de la Sangre/genética , Genes Bacterianos , Infecciones por Haemophilus/genética , Haemophilus influenzae/genética , Interacciones Huésped-Patógeno/genética , Infecciones del Sistema Respiratorio/genética , Actividad Bactericida de la Sangre/inmunología , Variación Genética , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/inmunología , Haemophilus influenzae/patogenicidad , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Pulmón/inmunología , Pulmón/microbiología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología
3.
PLoS Pathog ; 3(8): e118, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17722978

RESUMEN

Airway infection by the Gram-positive pathogen Streptococcus pneumoniae (Sp) leads to recruitment of neutrophils but limited bacterial killing by these cells. Co-colonization by Sp and a Gram-negative species, Haemophilus influenzae (Hi), provides sufficient stimulus to induce neutrophil and complement-mediated clearance of Sp from the mucosal surface in a murine model. Products from Hi, but not Sp, also promote killing of Sp by ex vivo neutrophil-enriched peritoneal exudate cells. Here we identify the stimulus from Hi as its peptidoglycan. Enhancement of opsonophagocytic killing was facilitated by signaling through nucleotide-binding oligomerization domain-1 (Nod1), which is involved in recognition of gamma-D-glutamyl-meso-diaminopimelic acid (meso-DAP) contained in cell walls of Hi but not Sp. Neutrophils from mice treated with Hi or compounds containing meso-DAP, including synthetic peptidoglycan fragments, showed increased Sp killing in a Nod1-dependent manner. Moreover, Nod1(-/-) mice showed reduced Hi-induced clearance of Sp during co-colonization. These observations offer insight into mechanisms of microbial competition and demonstrate the importance of Nod1 in neutrophil-mediated clearance of bacteria in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteína Adaptadora de Señalización NOD1/fisiología , Proteínas Opsoninas/metabolismo , Fagocitosis , Neumonía Neumocócica/metabolismo , Transducción de Señal , Streptococcus pneumoniae/patogenicidad , Animales , Silenciador del Gen , Humanos , Inmunidad Innata/fisiología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Proteínas Opsoninas/genética , Proteínas Opsoninas/inmunología , Peptidoglicano/inmunología , Peptidoglicano/metabolismo , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/microbiología , Streptococcus pneumoniae/inmunología
4.
mBio ; 2(5)2011.
Artículo en Inglés | MEDLINE | ID: mdl-21896678

RESUMEN

UNLABELLED: All fully sequenced strains of Streptococcus pneumoniae possess a version of the blp locus, which is responsible for bacteriocin production and immunity. Activation of the blp locus is stimulated by accumulation of the peptide pheromone, BlpC, following its secretion by the ABC transporter, BlpA. The blp locus is characterized by significant diversity in blpC type and in the region of the locus containing putative bacteriocin and immunity genes. In addition, the blpA gene can represent a single large open reading frame or be divided into several smaller fragments due to the presence of frameshift mutations. In this study, we use a collection of strains with blp-dependent inhibition and immunity to define the genetic changes that bring about phenotypic differences in bacteriocin production or immunity. We demonstrate that alterations in blpA, blpC, and bacteriocin/immunity content likely play an important role in competitive interactions between pneumococcal strains. Importantly, strains with a highly conserved frameshift mutation in blpA are unable to secrete bacteriocins or BlpC, but retain the ability to respond to exogenous peptide pheromone produced by cocolonizing strains, stimulating blp-mediated immunity. These "cheater" strains can only coexist with bacteriocin-producing strains that secrete their cognate BlpC and share the same immunity proteins. The variable outcome of these interactions helps to explain the heterogeneity of the blp pheromone, bacteriocin, and immunity protein content. IMPORTANCE: Streptococcus pneumoniae resides in a polymicrobial environment and competes for limited resources by the elaboration of small antimicrobial peptides called bacteriocins. A conserved cluster of genes in the S. pneumoniae genome is involved in the production of bacteriocins and their associated protective immunity proteins through secretion of a signaling pheromone. In this study, we show that a significant number of strains have lost the ability to secrete bacteriocins and signaling pheromones due to a specific mutation in a dedicated transporter protein. Because the regulatory and immunity portion of the locus is retained, these "cheater" strains can survive in the face of invasion from a bacteriocin-producing strain without the cost of bacteriocin secretion. The outcome of such interactions depends on each strain's repertoire of pheromone, immunity protein, and bacteriocin genes, such that intrastrain competition drives the diversity in bacteriocin, immunity protein, and pheromone content.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Bacteriocinas/metabolismo , Mutación del Sistema de Lectura , Variación Genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Análisis por Conglomerados , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Feromonas/metabolismo , Filogenia , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/aislamiento & purificación , Virulencia
5.
J Immunol ; 180(9): 6246-54, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18424747

RESUMEN

Delivery of Ag to inductive sites, such as nasal-associated lymphoid tissue (NALT) or GALT, is thought to promote mucosal immunity. Host and microbial factors that contribute to this process were investigated during model murine airway colonization by the pathogen Streptococcus pneumoniae. Colonization led to the deposition of released bacterial capsular Ag in the NALT in a manner consistent with trafficking through M cells. This Ag was derived from processing of bacteria in the lumen of the paranasal spaces rather than through invasion or sampling of intact bacteria. Neutrophils, which are recruited to the paranasal spaces where they associate with and may degrade bacteria, were required for efficient Ag delivery. Maximal Ag delivery to the NALT also required expression of the bacterial toxin pneumolysin. Pneumolysin and pneumolysin-expressing bacteria lysed neutrophils through pore formation in vitro. Accordingly, a pneumolysin-dependent loss of neutrophils, which correlated with the increased release of bacterial products, was observed in vivo. Thus, delivery of Ag to the NALT was enhanced by neutrophil-mediated generation of bacterial products together with bacterial-induced lysis of neutrophils. The impaired Ag delivery of pneumolysin-deficient bacteria was associated with diminished clearance from the mucosal surface. This study demonstrates how microbial-host interactions affect Ag delivery and the effectiveness of mucosal immunity.


Asunto(s)
Antígenos Bacterianos/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Mucosa , Neutrófilos/inmunología , Neumonía Neumocócica/inmunología , Streptococcus pneumoniae/fisiología , Estreptolisinas/inmunología , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Transporte Biológico/inmunología , Femenino , Inmunidad Mucosa/genética , Ratones , Ratones Endogámicos BALB C , Neutrófilos/microbiología , Neumonía Neumocócica/microbiología , Estreptolisinas/genética
6.
J Biol Chem ; 282(39): 28700-28708, 2007 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-17650505

RESUMEN

Streptococcus pneumoniae and Haemophilus influenzae are human pathogens that often asymptomatically colonize the mucosal surface of the upper respiratory tract, but also occasionally cause invasive disease. The ability of these species to traverse the epithelium of the airway mucosa was modeled in vitro using polarized respiratory epithelial cells in culture. Migration across the epithelial barrier was preceded by loss of transepithelial resistance. Membrane products of S. pneumoniae that included lipoteichoic acid induced disruption of the epithelial barrier in a Toll-like receptor 2-dependent manner. This result correlates with a recent genetic study that associates increased TLR2 signaling with increased rates of invasive pneumococcal disease in humans. Loss of transepithelial resistance by the TLR2 ligand correlated with activation of p38 MAP kinase and transforming growth factor (TGF)-beta signaling. Activation of p38 MAPK and TGF-beta signaling in epithelial cells upon nasal infection with S. pneumoniae was also demonstrated in vivo. Inhibition of either p38 MAPK or TGF-beta signaling was sufficient to inhibit the migration of S. pneumoniae or H. influenzae. Our data shows that diverse bacteria utilize common mechanisms, including MAPK and TGF-beta signaling pathways to disrupt epithelial barriers and promote invasion.


Asunto(s)
Células Epiteliales/inmunología , Infecciones por Haemophilus/inmunología , Haemophilus influenzae/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Infecciones Neumocócicas/inmunología , Mucosa Respiratoria/inmunología , Streptococcus pneumoniae/inmunología , Factor de Crecimiento Transformador beta/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Animales , Células CACO-2 , Membrana Celular/inmunología , Polaridad Celular/inmunología , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Infecciones por Haemophilus/patología , Haemophilus influenzae/patogenicidad , Humanos , Lipopolisacáridos/inmunología , Ratones , Modelos Biológicos , Infecciones Neumocócicas/patología , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Streptococcus pneumoniae/patogenicidad , Ácidos Teicoicos/inmunología , Receptor Toll-Like 2/inmunología
7.
Antimicrob Agents Chemother ; 51(9): 3225-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17664329

RESUMEN

The occurrence of mutator phenotypes among laboratory-generated and clinical levofloxacin-resistant strains of Streptococcus pneumoniae was determined using fluctuation analysis. The in vitro selection for levofloxacin-resistant mutants of strain D39, each with point mutations in both gyrA and parC or parE, was not associated with a significant change in the mutation rate. Two of eight clinical isolates resistant to levofloxacin (MIC, >8 microg/ml) had estimated mutation rates of 1.2 x 10(-7) and 9.4 x 10(-8) mutations per cell division, indicating potential mutator phenotypes, compared to strain D39, which had an estimated mutation rate of 1.4 x 10(-8) mutations per cell division. The levofloxacin-resistant isolates with the highest mutation rates showed evidence of dysfunctional mismatch repair and contained missense mutations in mut genes at otherwise highly conserved sites. The association of hypermutability in levofloxacin-resistant S. pneumoniae clinical isolates with mutations in DNA mismatch repair genes provides further evidence that mismatch repair mutants may have a selective advantage in the setting of antibiotic pressure, facilitating the development of further antibiotic resistance.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Fluoroquinolonas/farmacología , Mutación/genética , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/genética , Reparación del ADN/efectos de los fármacos , ADN Bacteriano/efectos de los fármacos , ADN Bacteriano/genética , Humanos , Datos de Secuencia Molecular , Mutación Missense/efectos de los fármacos , Mutación Missense/genética , Fenotipo , Infecciones Neumocócicas/microbiología , Quinina/análogos & derivados , Quinina/farmacología
8.
Cell Microbiol ; 9(5): 1343-51, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17474907

RESUMEN

During mucosal colonization, epithelial cells are concurrently exposed to numerous microbial species. Epithelial cytokine production is an early component of innate immunity and contributes to mucosal defence. We have previously demonstrated a synergistic response of respiratory epithelial cells to costimulation by two human pathogens, Streptococcus pneumoniae and Haemophilus influenzae. Here we define a molecular mechanism for the synergistic activation of epithelial signalling during polymicrobial colonization. H. influenzae peptidoglycan synergizes with the pore-forming toxin pneumolysin from S. pneumoniae. Radiolabelled peptidoglycan enters epithelial cells more efficiently in the presence of pneumolysin, consistent with peptidoglycan gaining access to the cytoplasm via toxin pores. Other pore-forming toxins (including anthrolysin O from Bacillus anthracis and Staphylococcus aureus alpha-toxin) can substitute for pneumolysin in the generation of synergistic responses. Consistent with a requirement for pore formation, S. pneumoniae expressing pneumolysin but not an isogenic mutant expressing a non-pore-forming toxoid prime epithelial responses. Nod1, a host cytoplasmic peptidoglycan-recognition molecule, is crucial to the epithelial response. Taken together, these findings demonstrate a role for cytosolic recognition of peptidoglycan in the setting of polymicrobial epithelial stimulation. We conclude that combinations of extracellular organisms can activate innate immune pathways previously considered to be reserved for the detection of intracellular microorganisms.


Asunto(s)
Bacterias/crecimiento & desarrollo , Citoplasma/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Transducción de Señal , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Línea Celular , Línea Celular Tumoral , Citoplasma/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Haemophilus influenzae/crecimiento & desarrollo , Haemophilus influenzae/metabolismo , Humanos , Interleucina-8/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacología , Proteína Adaptadora de Señalización NOD1/genética , Peptidoglicano/metabolismo , Peptidoglicano/farmacología , ARN Interferente Pequeño/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/metabolismo , Estreptolisinas/metabolismo , Estreptolisinas/farmacología , Fosfolipasas de Tipo C/metabolismo , Fosfolipasas de Tipo C/farmacología
9.
J Bacteriol ; 184(16): 4392-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12142409

RESUMEN

Loss-of-function mutations in the following seven pneumococcal genes were detected and analyzed: pspA, spxB, xba, licD2, lytA, nanA, and atpC. Factors associated with these mutations included (i) frameshifts caused by reversible gain and loss of single bases within homopolymeric repeats as short as 6 bases, (ii) deletions caused by recombinational events between nontandem direct repeats as short as 8 bases, and (iii) substitutions of guanine residues caused at an increased frequency by the high levels of hydrogen peroxide (>2 mM) typically generated by this species under aerobic growth conditions. The latter accounted for a frequency as high as 2.8 x 10(-6) for spontaneous mutation to resistance to optochin and was 10- to 200-fold lower in the absence of detectable levels of H2O2. Some of these mutations appear to have been selected for in vivo during pneumococcal infection, perhaps as a consequence of immune pressure or oxidative stress.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Secuencia de Bases , Mutación del Sistema de Lectura/fisiología , Proteínas de Choque Térmico/genética , Datos de Secuencia Molecular , N-Acetil Muramoil-L-Alanina Amidasa/genética , Estrés Oxidativo/genética , Piruvato Oxidasa/genética , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/genética , Secuencias Repetidas en Tándem/genética
10.
Antimicrob Agents Chemother ; 47(1): 1-6, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12499161

RESUMEN

Cationic antimicrobial peptides are believed to exert their primary activities on anionic bacterial cell membranes; however, this model does not adequately account for several important structure-activity relationships. These relationships are likely to be influenced by the bacterial response to peptide challenge. In order to characterize the genomic aspect of this response, transcription profiles were examined for Escherichia coli isolates treated with sublethal and lethal concentrations of the cationic antimicrobial peptide cecropin A. Transcript levels for 26 genes changed significantly following treatment with sublethal peptide concentrations, and half of the transcripts corresponded to protein products with unknown function. The pattern of response is distinct from that following treatment with lethal concentrations and is also distinct from the bacterial response to nutritional, thermal, osmotic, or oxidative stress. These results demonstrate that cecropin A induces a genomic response in E. coli apart from any lethal effects on the membrane and suggest that a complete understanding of its mechanism of action may require a detailed examination of this response.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Escherichia coli/efectos de los fármacos , Hibridación de Ácido Nucleico/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Células Cultivadas , Escherichia coli/genética , Hibridación de Ácido Nucleico/genética , ARN Bacteriano/aislamiento & purificación , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA