Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 57(4): 440-450.e7, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-34986324

RESUMEN

Regeneration of adult mammalian central nervous system (CNS) axons is abortive, resulting in inability to recover function after CNS lesion, including spinal cord injury (SCI). Here, we show that the spiny mouse (Acomys) is an exception to other mammals, being capable of spontaneous and fast restoration of function after severe SCI, re-establishing hind limb coordination. Remarkably, Acomys assembles a scarless pro-regenerative tissue at the injury site, providing a unique structural continuity of the initial spinal cord geometry. The Acomys SCI site shows robust axon regeneration of multiple tracts, synapse formation, and electrophysiological signal propagation. Transcriptomic analysis of the spinal cord following transcriptome reconstruction revealed that Acomys rewires glycosylation biosynthetic pathways, culminating in a specific pro-regenerative proteoglycan signature at SCI site. Our work uncovers that a glycosylation switch is critical for axon regeneration after SCI and identifies ß3gnt7, a crucial enzyme of keratan sulfate biosynthesis, as an enhancer of axon growth.


Asunto(s)
Axones/fisiología , Regeneración Nerviosa/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/patología , Animales , Axones/patología , Modelos Animales de Enfermedad , Glicosilación , Ratones , Médula Espinal/fisiología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/fisiopatología , Columna Vertebral/fisiopatología
2.
mBio ; 12(6): e0256921, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34696596

RESUMEN

Streptococcus pneumoniae is an asymptomatic colonizer of the nasopharynx, but it is also one of the most important bacterial pathogens of humans, causing a wide range of mild to life-threatening diseases. The basis of the pneumococcal transition from a commensal to a parasitic lifestyle is not fully understood. We hypothesize that exposure to host catecholamine stress hormones is important for this transition. In this study, we demonstrated that pneumococci preexposed to a hormone released during stress, norepinephrine (NE), have an increased capacity to translocate from the nasopharynx into the lungs compared to untreated pneumococci. Examination of NE-treated pneumococci revealed major alterations in metabolic profiles, cell associations, capsule synthesis, and cell size. By systemically mutating all 12 two-component and 1 orphan regulatory systems, we also identified a unique genetic regulatory circuit involved in pneumococcal recognition and responsiveness to human stress hormones. IMPORTANCE Microbes acquire unique lifestyles under different environmental conditions. Although this is a widespread occurrence, our knowledge of the importance of various host signals and their impact on microbial behavior is not clear despite the therapeutic value of this knowledge. We discovered that catecholamine stress hormones are the host signals that trigger the passage of Streptococcus pneumoniae from a commensal to a parasitic state. We identify that stress hormone treatment of this microbe leads to reductions in cell size and capsule synthesis and renders it more able to migrate from the nasopharynx into the lungs in a mouse model of infection. The microbe requires the TCS09 protein for the recognition and processing of stress hormone signals. Our work has particular clinical significance as catecholamines are abundant in upper respiratory fluids as well as being administered therapeutically to reduce inflammation in ventilated patients, which may explain why intubation in the critically ill is a recognized risk factor for the development of pneumococcal pneumonia.


Asunto(s)
Traslocación Bacteriana , Pulmón/microbiología , Neumonía Neumocócica/microbiología , Streptococcus pneumoniae/fisiología , Animales , Femenino , Humanos , Ratones , Nasofaringe/microbiología , Norepinefrina/metabolismo , Neumonía Neumocócica/metabolismo , Neumonía Neumocócica/fisiopatología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Estrés Fisiológico
3.
PLoS Pathog ; 16(4): e1008464, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32324805

RESUMEN

Streptococcus pneumoniae is a major human pathogen, causing pneumonia and sepsis. Genetic components strongly influence host responses to pneumococcal infections, but the responsible loci are unknown. We have previously identified a locus on mouse chromosome 7 from a susceptible mouse strain, CBA/Ca, to be crucial for pneumococcal infection. Here we identify a responsible gene, Cd22, which carries a point mutation in the CBA/Ca strain, leading to loss of CD22 on B cells. CBA/Ca mice and gene-targeted CD22-deficient mice on a C57BL/6 background are both similarly susceptible to pneumococcal infection, as shown by bacterial replication in the lungs, high bacteremia and early death. After bacterial infections, CD22-deficient mice had strongly reduced B cell populations in the lung, including GM-CSF producing, IgM secreting innate response activator B cells, which are crucial for protection. This study provides striking evidence that CD22 is crucial for protection during invasive pneumococcal disease.


Asunto(s)
Linfocitos B/inmunología , Infecciones Neumocócicas/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Animales , Linfocitos B/microbiología , Bacteriemia/genética , Bacteriemia/inmunología , Bacteriemia/microbiología , Femenino , Interacciones Huésped-Patógeno , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/metabolismo , Neumonía Neumocócica/genética , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/metabolismo , Neumonía Neumocócica/microbiología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/deficiencia , Lectina 2 Similar a Ig de Unión al Ácido Siálico/genética , Streptococcus pneumoniae/patogenicidad
4.
Nat Microbiol ; 3(5): 600-610, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29662129

RESUMEN

Bacterial septicaemia is a major cause of mortality, but its pathogenesis remains poorly understood. In experimental pneumococcal murine intravenous infection, an initial reduction of bacteria in the blood is followed hours later by a fatal septicaemia. These events represent a population bottleneck driven by efficient clearance of pneumococci by splenic macrophages and neutrophils, but as we show in this study, accompanied by occasional intracellular replication of bacteria that are taken up by a subset of CD169+ splenic macrophages. In this model, proliferation of these sequestered bacteria provides a reservoir for dissemination of pneumococci into the bloodstream, as demonstrated by its prevention using an anti-CD169 monoclonal antibody treatment. Intracellular replication of pneumococci within CD169+ splenic macrophages was also observed in an ex vivo porcine spleen, where the microanatomy is comparable with humans. We also showed that macrolides, which effectively penetrate macrophages, prevented septicaemia, whereas beta-lactams, with inefficient intracellular penetration, failed to prevent dissemination to the blood. Our findings define a shift in our understanding of the pneumococcus from an exclusively extracellular pathogen to one with an intracellular phase. These findings open the door to the development of treatments that target this early, previously unrecognized intracellular phase of bacterial sepsis.


Asunto(s)
ADN Bacteriano/genética , Macrófagos/microbiología , Infecciones Neumocócicas/complicaciones , Sepsis/microbiología , Bazo/citología , Streptococcus pneumoniae/fisiología , Animales , Replicación del ADN , Modelos Animales de Enfermedad , Humanos , Macrólidos/farmacología , Macrólidos/uso terapéutico , Ratones , Infecciones Neumocócicas/tratamiento farmacológico , Sepsis/tratamiento farmacológico , Sepsis/etiología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Bazo/microbiología , Streptococcus pneumoniae/patogenicidad , Porcinos
5.
Infect Immun ; 85(11)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28808161

RESUMEN

Listeria monocytogenes is a foodborne pathogen responsible for a number of life-threatening infections of humans. During an infection, it invades epithelial cells before spreading from the intestine to the cells of the liver and spleen. This requires an ability to adapt to varying oxygen levels. Here, we demonstrate that L. monocytogenes has two terminal oxidases, a cytochrome bd-type (CydAB) and a cytochrome aa 3-type menaquinol (QoxAB) oxidase, and that both are used for respiration under different oxygen tensions. Furthermore, we show that possession of both terminal oxidases is important in infection. In air, the CydAB bd-type oxidase is essential for aerobic respiration and intracellular replication, and cydAB mutants are highly attenuated in mice. In contrast, the QoxAB aa 3-type oxidase is required neither for aerobic respiration in air nor for intracellular growth. However, the qoxAB mutants are attenuated in mice, with a delay in the onset of disease signs and with increased survival time, indicating a role for the QoxAB aa 3-type oxidase in the initial stages of infection. Growth of bacteria under defined oxygen conditions revealed that at 1% (vol/vol), both oxidases are functional, and the presence of either is sufficient for aerobic respiration and intracellular replication. However, at 0.2% (vol/vol), both oxidases are necessary for maximum growth. These findings are consistent with the ability of L. monocytogenes to switch between terminal oxidases under different oxygen conditions, providing exquisite adaptation to different conditions encountered within the infected host.

6.
Environ Microbiol ; 19(5): 1868-1880, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28195384

RESUMEN

Air pollution is the world's largest single environmental health risk (WHO). Particulate matter such as black carbon is one of the main components of air pollution. The effects of particulate matter on human health are well established however the effects on bacteria, organisms central to ecosystems in humans and in the natural environment, are poorly understood. We report here for the first time that black carbon drastically changes the development of bacterial biofilms, key aspects of bacterial colonisation and survival. Our data show that exposure to black carbon induces structural, compositional and functional changes in the biofilms of both S. pneumoniae and S. aureus. Importantly, the tolerance of the biofilms to multiple antibiotics and proteolytic degradation is significantly affected. Additionally, our results show that black carbon impacts bacterial colonisation in vivo. In a mouse nasopharyngeal colonisation model, black carbon caused S. pneumoniae to spread from the nasopharynx to the lungs, which is essential for subsequent infection. Therefore our study highlights that air pollution has a significant effect on bacteria that has been largely overlooked. Consequently these findings have important implications concerning the impact of air pollution on human health and bacterial ecosystems worldwide.


Asunto(s)
Contaminación del Aire/efectos adversos , Antibacterianos/farmacología , Biopelículas/crecimiento & desarrollo , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Hollín/farmacología , Streptococcus pneumoniae/crecimiento & desarrollo , Animales , Biopelículas/efectos de los fármacos , Humanos , Pulmón/microbiología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Nasofaringe/microbiología , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/microbiología , Proteolisis/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Streptococcus pneumoniae/efectos de los fármacos
7.
PLoS One ; 10(3): e0121042, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25826206

RESUMEN

The human pathogen Streptococcus pneumoniae is a strictly fermentative organism that relies on glycolytic metabolism to obtain energy. In the human nasopharynx S. pneumoniae encounters glycoconjugates composed of a variety of monosaccharides, which can potentially be used as nutrients once depolymerized by glycosidases. Therefore, it is reasonable to hypothesise that the pneumococcus would rely on these glycan-derived sugars to grow. Here, we identified the sugar-specific catabolic pathways used by S. pneumoniae during growth on mucin. Transcriptome analysis of cells grown on mucin showed specific upregulation of genes likely to be involved in deglycosylation, transport and catabolism of galactose, mannose and N acetylglucosamine. In contrast to growth on mannose and N-acetylglucosamine, S. pneumoniae grown on galactose re-route their metabolic pathway from homolactic fermentation to a truly mixed acid fermentation regime. By measuring intracellular metabolites, enzymatic activities and mutant analysis, we provide an accurate map of the biochemical pathways for galactose, mannose and N-acetylglucosamine catabolism in S. pneumoniae. Intranasal mouse infection models of pneumococcal colonisation and disease showed that only mutants in galactose catabolic genes were attenuated. Our data pinpoint galactose as a key nutrient for growth in the respiratory tract and highlights the importance of central carbon metabolism for pneumococcal pathogenesis.


Asunto(s)
Galactosa/metabolismo , Polisacáridos/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus pneumoniae/metabolismo , Regulación Bacteriana de la Expresión Génica , Mucinas/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Virulencia
8.
J Bacteriol ; 197(5): 807-18, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25512311

RESUMEN

Streptococcus pneumoniae is a common nasopharyngeal resident in healthy people but, at the same time, one of the major causes of infectious diseases such as pneumonia, meningitis, and sepsis. The shift from commensal to pathogen and its interaction with host cells are poorly understood. One of the major limitations for research on pneumococcal-host interactions is the lack of suitable tools for live-cell imaging. To address this issue, we developed a generally applicable strategy to create genetically stable, highly fluorescent bacteria. Our strategy relies on fusing superfolder green fluorescent protein (GFP) or a far-red fluorescent protein (RFP) to the abundant histone-like protein HlpA. Due to efficient translation and limited cellular diffusion of these fusions, the cells are 25-fold brighter than those of the currently best available imaging S. pneumoniae strain. These novel bright pneumococcal strains are fully virulent, and the GFP reporter can be used for in situ imaging in mouse tissue. We used our reporter strains to study the effect of the polysaccharide capsule, a major pneumococcal virulence factor, on different stages of infection. By dual-color live-cell imaging experiments, we show that unencapsulated pneumococci adhere significantly better to human lung epithelial cells than encapsulated strains, in line with previous data obtained by classical approaches. We also confirm with live-cell imaging that the capsule protects pneumococci from neutrophil phagocytosis, demonstrating the versatility and usability of our reporters. The described imaging tools will pave the way for live-cell imaging of pneumococcal infection and help further understanding of the mechanisms of pneumococcal pathogenesis.


Asunto(s)
Rastreo Celular/métodos , Interacciones Huésped-Patógeno , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/química , Streptococcus pneumoniae/fisiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Fluorescencia , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Pulmón/microbiología , Ratones , Streptococcus pneumoniae/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
9.
PLoS One ; 9(3): e89831, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24594938

RESUMEN

Streptococcus pneumoniae is an important human pathogen responsible for high mortality and morbidity worldwide. The susceptibility to pneumococcal infections is controlled by as yet unknown genetic factors. To elucidate these factors could help to develop new medical treatments and tools to identify those most at risk. In recent years genome wide association studies (GWAS) in mice and humans have proved successful in identification of causal genes involved in many complex diseases for example diabetes, systemic lupus or cholesterol metabolism. In this study a GWAS approach was used to map genetic loci associated with susceptibility to pneumococcal infection in 26 inbred mouse strains. As a result four candidate QTLs were identified on chromosomes 7, 13, 18 and 19. Interestingly, the QTL on chromosome 7 was located within S. pneumoniae resistance QTL (Spir1) identified previously in a linkage study of BALB/cOlaHsd and CBA/CaOlaHsd F2 intercrosses. We showed that only a limited number of genes encoded within the QTLs carried phenotype-associated polymorphisms (22 genes out of several hundred located within the QTLs). These candidate genes are known to regulate TGFß signalling, smooth muscle and immune cells functions. Interestingly, our pulmonary histopathology and gene expression data demonstrated, lung vasculature plays an important role in resistance to pneumococcal infection. Therefore we concluded that the cumulative effect of these candidate genes on vasculature and immune cells functions as contributory factors in the observed differences in susceptibility to pneumococcal infection. We also propose that TGFß-mediated regulation of fibroblast differentiation plays an important role in development of invasive pneumococcal disease. Gene expression data submitted to the NCBI Gene Expression Omnibus Accession No: GSE49533 SNP data submitted to NCBI dbSNP Short Genetic Variation http://www.ncbi.nlm.nih.gov/projects/SNP/snp_viewTable.cgi?handle=MUSPNEUMONIA.


Asunto(s)
Estudio de Asociación del Genoma Completo , Pulmón/irrigación sanguínea , Infecciones Neumocócicas/inmunología , Animales , Humanos , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Sitios de Carácter Cuantitativo , Transcriptoma
10.
BMC Genomics ; 14: 242, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23577770

RESUMEN

BACKGROUND: Streptococcus pneumoniae causes over one million deaths worldwide annually, despite recent developments in vaccine and antibiotic therapy. Host susceptibility to pneumococcal infection and disease is controlled by a combination of genetic and environmental influences, but current knowledge remains limited. RESULTS: In order to identify novel host genetic variants as predictive risk factors or as potential targets for prophylaxis, we have looked for quantitative trait loci in a mouse model of invasive pneumococcal disease. We describe a novel locus, called Streptococcus pneumoniae infection resistance 2 (Spir2) on Chr4, which influences time to morbidity and the development of bacteraemia post-infection. CONCLUSIONS: The two quantitative trait loci we have identified (Spir1 and Spir2) are linked significantly to both bacteraemia and survival time. This may mean that the principle cause of death, in our model of pneumonia, is bacteraemia and the downstream inflammatory effects it precipitates in the host.


Asunto(s)
Cromosomas de los Mamíferos , Predisposición Genética a la Enfermedad , Proteínas de Microfilamentos/genética , Infecciones Neumocócicas/genética , Sitios de Carácter Cuantitativo , Streptococcus pneumoniae , Animales , Bacteriemia/genética , Bacteriemia/microbiología , Cruzamiento , Femenino , Genotipo , Haplotipos , Escala de Lod , Masculino , Ratones , Fenotipo , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/mortalidad , Polimorfismo de Nucleótido Simple
11.
PLoS Pathog ; 8(4): e1002660, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22563306

RESUMEN

Streptococcus pneumoniae is an important human pathogen responsible for a spectrum of diseases including pneumonia. Immunological and pro-inflammatory processes induced in the lung during pneumococcal infection are well documented, but little is known about the role played by immunoregulatory cells and cytokines in the control of such responses. We demonstrate considerable differences in the immunomodulatory cytokine transforming growth factor (TGF)-ß between the pneumococcal pneumonia resistant BALB/c and susceptible CBA/Ca mouse strains. Immunohistochemistry and flow cytometry reveal higher levels of TGF-ß protein in BALB/c lungs during pneumococcal pneumonia that correlates with a rapid rise in lung Foxp3(+)Helios(+) T regulatory cells. These cells have protective functions during pneumococcal pneumonia, because blocking their induction with an inhibitor of TGF-ß impairs BALB/c resistance to infection and aids bacterial dissemination from lungs. Conversely, adoptive transfer of T regulatory cells to CBA/Ca mice, prior to infection, prolongs survival and decreases bacterial dissemination from lungs to blood. Importantly, strong T regulatory cell responses also correlate with disease-resistance in outbred MF1 mice, confirming the importance of immunoregulatory cells in controlling protective responses to the pneumococcus. This study provides exciting new evidence for the importance of immunomodulation during pulmonary pneumococcal infection and suggests that TGF-ß signalling is a potential target for immunotherapy or drug design.


Asunto(s)
Neumonía Neumocócica/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Proteínas de Unión al ADN/inmunología , Susceptibilidad a Enfermedades/inmunología , Sistemas de Liberación de Medicamentos , Femenino , Factores de Transcripción Forkhead/inmunología , Ratones , Ratones Endogámicos BALB C , Neumonía Neumocócica/tratamiento farmacológico , Especificidad de la Especie , Streptococcus pneumoniae/inmunología , Factores de Transcripción/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
12.
J Immunol ; 186(10): 5907-15, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21460207

RESUMEN

Neutrophils and T cells play an important role in host protection against pulmonary infection caused by Streptococcus pneumoniae. However, the role of the integrins in recruitment of these cells to infected lungs is not well understood. In this study we used the twin approaches of mAb blockade and gene-deficient mice to investigate the relative impact of specific integrins on cellular recruitment and bacterial loads following pneumococcal infection. We find that both Mac-1 (CD11b/CD18) and α(4)ß(1) (CD49d/CD29) integrins, but surprisingly not LFA-1 (CD11a/CD18), contribute to two aspects of the response. In terms of recruitment from the circulation into lungs, neutrophils depend on Mac-1 and α(4)ß(1), whereas the T cells are entirely dependent on α(4)ß(1). Second, immunohistochemistry results indicate that adhesion also plays a role within infected lung tissue itself. There is widespread expression of ICAM-1 within lung tissue. Use of ICAM-1(-/-) mice revealed that neutrophils make use of this Mac-1 ligand, not for lung entry or for migration within lung tissue, but for combating the pneumococcal infection. In contrast to ICAM-1, there is restricted and constitutive expression of the α(4)ß(1) ligand, VCAM-1, on the bronchioles, allowing direct access of the leukocytes to the airways via this integrin at an early stage of pneumococcal infection. Therefore, integrins Mac-1 and α(4)ß(1) have a pivotal role in prevention of pneumococcal outgrowth during disease both in regulating neutrophil and T cell recruitment into infected lungs and by influencing their behavior within the lung tissue itself.


Asunto(s)
Integrina alfa4beta1/inmunología , Antígeno de Macrófago-1/inmunología , Infiltración Neutrófila , Neutrófilos/inmunología , Neumonía Neumocócica/inmunología , Streptococcus pneumoniae/inmunología , Linfocitos T/inmunología , Administración Intranasal , Animales , Anticuerpos Monoclonales , Movimiento Celular , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Pulmón/inmunología , Pulmón/microbiología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Streptococcus pneumoniae/patogenicidad
13.
PLoS Pathog ; 6(11): e1001191, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21085613

RESUMEN

Pneumolysin (PLY) is a key Streptococcus pneumoniae virulence factor and potential candidate for inclusion in pneumococcal subunit vaccines. Dendritic cells (DC) play a key role in the initiation and instruction of adaptive immunity, but the effects of PLY on DC have not been widely investigated. Endotoxin-free PLY enhanced costimulatory molecule expression on DC but did not induce cytokine secretion. These effects have functional significance as adoptive transfer of DC exposed to PLY and antigen resulted in stronger antigen-specific T cell proliferation than transfer of DC exposed to antigen alone. PLY synergized with TLR agonists to enhance secretion of the proinflammatory cytokines IL-12, IL-23, IL-6, IL-1ß, IL-1α and TNF-α by DC and enhanced cytokines including IL-17A and IFN-γ by splenocytes. PLY-induced DC maturation and cytokine secretion by DC and splenocytes was TLR4-independent. Both IL-17A and IFN-γ are required for protective immunity to pneumococcal infection and intranasal infection of mice with PLY-deficient pneumococci induced significantly less IFN-γ and IL-17A in the lungs compared to infection with wild-type bacteria. IL-1ß plays a key role in promoting IL-17A and was previously shown to mediate protection against pneumococcal infection. The enhancement of IL-1ß secretion by whole live S. pneumoniae and by PLY in DC required NLRP3, identifying PLY as a novel NLRP3 inflammasome activator. Furthermore, NLRP3 was required for protective immunity against respiratory infection with S. pneumoniae. These results add significantly to our understanding of the interactions between PLY and the immune system.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/farmacología , Receptor Toll-Like 4/metabolismo , Animales , Proteínas Bacterianas/farmacología , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/microbiología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/microbiología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Infecciones Neumocócicas/metabolismo , Infecciones Neumocócicas/microbiología , Bazo/inmunología , Bazo/metabolismo , Bazo/microbiología , Streptococcus pneumoniae/inmunología
14.
Part Fibre Toxicol ; 7: 30, 2010 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-20958976

RESUMEN

BACKGROUND: Epidemiological studies suggest that inhalation of carbonaceous particulate matter from biomass combustion increases susceptibility to bacterial pneumonia. In vitro studies report that phagocytosis of carbon black by alveolar macrophages (AM) impairs killing of Streptococcus pneumoniae. We have previously reported high levels of black carbon in AM from biomass smoke-exposed children and adults. We therefore aimed to use a mouse model to test the hypothesis that high levels of carbon loading of AM in vivo increases susceptibility to pneumococcal pneumonia. METHODS: Female outbred mice were treated with either intranasal phosphate buffered saline (PBS) or ultrafine carbon black (UF-CB in PBS; 500 µg on day 1 and day 4), and then infected with S. pneumoniae strain D39 on day 5. Survival was assessed over 72 h. The effect of UF-CB on AM carbon loading, airway inflammation, and a urinary marker of pulmonary oxidative stress was assessed in uninfected animals. RESULTS: Instillation of UF-CB in mice resulted a pattern of AM carbon loading similar to that of biomass-smoke exposed humans. In uninfected animals, UF-CB treated animals had increased urinary 8-oxodG (P = 0.055), and an increased airway neutrophil differential count (P < 0.01). All PBS-treated mice died within 72 h after infection with S. pneumoniae, whereas morbidity and mortality after infection was reduced in UF-CB treated animals (median survival 48 h vs. 30 h, P < 0.001). At 24 hr post-infection, UF-CB treated mice had lower lung and the blood S. pneumoniae colony forming unit counts, and lower airway levels of keratinocyte-derived chemokine/growth-related oncogene (KC/GRO), and interferon gamma. CONCLUSION: Acute high level loading of AM with ultrafine carbon black particles per se does not increase the susceptibility of mice to pneumococcal infection in vivo.


Asunto(s)
Neumonía Neumocócica/fisiopatología , Hollín/toxicidad , Streptococcus pneumoniae , Administración por Inhalación , Animales , Líquido del Lavado Bronquioalveolar , Quimiocinas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Interferón gamma/metabolismo , Pulmón/microbiología , Macrófagos Alveolares/metabolismo , Ratones , Tamaño de la Partícula , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/microbiología
15.
J Immunol ; 184(12): 7108-15, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20483738

RESUMEN

Mast cells are emerging as contributors to innate immunity. Mouse mast cells have a pivotal role in protection against bacterial infection, and human cord blood-derived mast cells reduce bacterial viability in culture. The objectives of this study were to determine whether human lung mast cells (HLMCs) might be protective against pneumococcal lung infection through direct antimicrobial activity. Tissue-derived HLMCs and the human mast cell lines HMC-1 and LAD2 were cocultured with wild-type and mutant pneumococci, and viability and functional assays were performed. Mast cells were also stimulated with purified pneumolysin. HLMCs killed wild-type serotype-2 (D39) pneumococci in coculture but had no effect on an isogenic pneumolysin-deficient (PLN-A) pneumococcus. D39 wild-type, but not PLN-A pneumococci, induced the release of leukotriene C4 from human mast cells in a dose-dependent manner, which was not accompanied by histamine release. Stimulation of mast cells with sublytic concentrations of purified pneumolysin replicated this effect. Furthermore, pneumolysin induced the release of the cathelicidin LL-37 from HLMCs, purified LL-37 reduced pneumococcal viability, and neutralizing Ab to LL-37 attenuated mast cell-dependent pneumococcal killing. In addition, at high concentrations, all pneumococcal strains tested reduced HLMC viability through a combination of pneumolysin and H2O2-dependent mechanisms. HLMCs exhibit direct antimicrobial activity to pneumococci through their activation by pneumolysin. This antimicrobial activity is mediated, in part, by the release of LL-37 from HLMCs. This suggests that mast cells provide an early warning system and potentially limit pneumococcal dissemination early in the course of invasive pulmonary pneumococcal disease.


Asunto(s)
Pulmón/inmunología , Mastocitos/inmunología , Streptococcus pneumoniae/inmunología , Estreptolisinas/inmunología , Proteínas Bacterianas/inmunología , Degranulación de la Célula/inmunología , Células Cultivadas , Técnicas de Cocultivo , Humanos , Pulmón/citología , Mastocitos/microbiología , Infecciones Neumocócicas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...