Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Elife ; 122023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37222419

RESUMEN

Streptococcus pneumoniae is a major pathogen in children, elderly subjects, and immunodeficient patients. Pentraxin 3 (PTX3) is a fluid-phase pattern recognition molecule (PRM) involved in resistance to selected microbial agents and in regulation of inflammation. The present study was designed to assess the role of PTX3 in invasive pneumococcal infection. In a murine model of invasive pneumococcal infection, PTX3 was strongly induced in non-hematopoietic (particularly, endothelial) cells. The IL-1ß/MyD88 axis played a major role in regulation of the Ptx3 gene expression. Ptx3-/- mice presented more severe invasive pneumococcal infection. Although high concentrations of PTX3 had opsonic activity in vitro, no evidence of PTX3-enhanced phagocytosis was obtained in vivo. In contrast, Ptx3-deficient mice showed enhanced recruitment of neutrophils and inflammation. Using P-selectin-deficient mice, we found that protection against pneumococcus was dependent upon PTX3-mediated regulation of neutrophil inflammation. In humans, PTX3 gene polymorphisms were associated with invasive pneumococcal infections. Thus, this fluid-phase PRM plays an important role in tuning inflammation and resistance against invasive pneumococcal infection.


Asunto(s)
Inflamación , Infecciones Neumocócicas , Animales , Ratones , Inflamación/metabolismo , Neutrófilos/metabolismo , Fagocitosis , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae
2.
Biomed Res Int ; 2021: 9963700, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34604389

RESUMEN

The study is aimed at observing the influence of microribonucleic acid- (miRNA-) 30a-50p on the pulmonary fibrosis in mice with Streptococcus pneumoniae infection through the regulation of autophagy by Beclin-1. Specific pathogen-free mice were instilled with Streptococcus pneumoniae through the trachea to establish the pulmonary fibrosis model. Then, they were divided into the miRNA-30a-50p mimics group (mimics group, n = 10) and miRNA-30a-5p inhibitors group (inhibitors group, n = 10), with the control group (n = 10) also set. Pulmonary tissue wet weight/dry weight (W/D) was detected. The content of tumor necrosis factor-α (TNF-α), interleukin- (IL-) 6, and myeloperoxidase (MPO) was determined using enzyme-linked immunosorbent assay (ELISA). Besides, the changes in the pulmonary function index dynamic lung compliance (Cdyn), plateau pressure (Pplat), and peak airway pressure (Ppeak) were monitored, and the gene and protein expression levels were measured via quantitative PCR (qPCR) and Western blotting. The expression level of miRNA-30a-5p was substantially raised in the mimics group (p < 0.05), but extremely low in the inhibitors group (p < 0.05). The mimics group had obviously raised levels of serum aminotransferase (AST), glutamic-pyruvic transaminase (GPT), alkaline phosphatase (ALP), and pulmonary tissue W/D (p < 0.05). Additionally, the expression levels of TNF-α, IL-6, and MPO were notably elevated in the mimics group, while their expression levels showed the opposite conditions in the inhibitors group (p < 0.05). According to the HE staining results, the inhibitors group had arranged orderly cells, while the mimics group exhibited lung injury, pulmonary edema, severe inflammatory response, and alveolar congestion. In the inhibitors group, Cdyn was remarkably elevated, but Pplat and Ppeak declined considerably (p < 0.05). Besides, the inhibitors group exhibited elevated messenger RNA (mRNA) levels of Beclin-1 and LC3, lowered mRNA levels of α-SMA and p62, a raised protein level of Beclin-1, and a markedly decreased protein level of p62 (p < 0.05). Silencing miRNA-30a-5p expression can promote the expression of Beclin-1 to accelerate the occurrence of autophagy, thereby treating pulmonary fibrosis in mice with Streptococcus pneumoniae infection.


Asunto(s)
Autofagia/genética , Beclina-1/metabolismo , MicroARNs/metabolismo , Infecciones Neumocócicas/genética , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/microbiología , Actinas/metabolismo , Fosfatasa Alcalina/sangre , Animales , Aspartato Aminotransferasas/sangre , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Ratones , MicroARNs/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Tamaño de los Órganos , Pruebas de Función Respiratoria , Proteína Sequestosoma-1/metabolismo , Transaminasas/sangre
3.
Front Immunol ; 12: 786164, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35003103

RESUMEN

Family history is one key in diagnosing inborn errors of immunity (IEI); however, disease status is difficult to determine in deceased relatives. X-linked anhidrotic ectodermal dysplasia with immunodeficiency is one of the hyper IgM syndromes that is caused by a hypomorphic variant in the nuclear factor kappa beta essential modulator. We identified a novel IKBKG variant in a 7-month-old boy with pneumococcal rib osteomyelitis and later found that his mother has incontinentia pigmenti. Genetic analysis of preserved umbilical cords revealed the same variant in two of his deceased maternal uncles. Analysis of preserved umbilical cord tissue from deceased relatives can provide important information for diagnosing IEI in their descendants.


Asunto(s)
Displasia Ectodérmica/diagnóstico , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Quinasa I-kappa B/genética , Osteomielitis/diagnóstico , Infecciones Neumocócicas/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Cordón Umbilical/patología , Análisis Mutacional de ADN , Diagnóstico Tardío , Displasia Ectodérmica/complicaciones , Displasia Ectodérmica/genética , Displasia Ectodérmica/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Humanos , Lactante , Masculino , Osteomielitis/genética , Osteomielitis/inmunología , Osteomielitis/microbiología , Linaje , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/inmunología , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/aislamiento & purificación
4.
Nat Microbiol ; 6(2): 257-269, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33349663

RESUMEN

Streptococcus pneumoniae is a natural colonizer of the human respiratory tract and an opportunistic pathogen. Although epithelial cells are among the first to encounter pneumococci, the cellular processes and contribution of epithelial cells to the host response are poorly understood. Here, we show that a S. pneumoniae serotype 6B ST90 strain, which does not cause disease in a murine infection model, induces a unique NF-κB signature response distinct from an invasive-disease-causing isolate of serotype 4 (TIGR4). This signature is characterized by activation of p65 and requires a histone demethylase KDM6B. We show, molecularly, that the interaction of the 6B strain with epithelial cells leads to chromatin remodelling within the IL-11 promoter in a KDM6B-dependent manner, where KDM6B specifically demethylates histone H3 lysine 27 dimethyl. Remodelling of the IL-11 locus facilitates p65 access to three NF-κB sites that are otherwise inaccessible when stimulated by IL-1ß or TIGR4. Finally, we demonstrate through chemical inhibition of KDM6B with GSK-J4 inhibitor and through exogenous addition of IL-11 that the host responses to the 6B ST90 and TIGR4 strains can be interchanged both in vitro and in a murine model of infection in vivo. Our studies therefore reveal how a chromatin modifier governs cellular responses during infection.


Asunto(s)
Ensamble y Desensamble de Cromatina , Interacciones Huésped-Patógeno/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/patogenicidad , Células A549 , Células Epiteliales Alveolares , Animales , Benzazepinas/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos , Células Epiteliales/microbiología , Regulación de la Expresión Génica , Humanos , Interleucina-11/genética , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , FN-kappa B/farmacología , Infecciones Neumocócicas/enzimología , Infecciones Neumocócicas/genética , Regiones Promotoras Genéticas , Pirimidinas/farmacología
6.
Biosens Bioelectron ; 151: 111969, 2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-31999579

RESUMEN

We report a peptide-based sensor that involves a multivalent interaction with L-ascorbate 6-phosphate lactonase (UlaG), a protein marker of Streptococcus pneumonia. By integrating the antifouling feature of the sensor, we significantly improved the signal-to-noise ratio of UlaG detection. The antifouling surface was fabricated via electrodeposition using an equivalent mixture of 4-amino-N,N,N-trimethylanilinium and 4-aminobenzenesulfonate. This antifouling layer not only effectively reduces the non-specific adsorption on the biosensor but also decreases the charge transfer resistance (Rct) of the screen-printed carbon electrode. The aniline-modified S7 peptide, an UlaG-binding peptide, was pre-synthesized and further electrochemically modified to bind onto the antifouling layer. Bio-electrochemical analysis confirms that the antifouling S7-peptide sensor binds strongly to the UlaG with a dissociation constant (Kd) = 0.5 nM. This strong interaction can be attributed to a multivalent interaction between the biosensor and the heximeric form of UlaG. To demonstrate the potential for clinical application, further detection of Streptococcus pneumonia from 50 to 5×104 CFU/mL were successfully performed in 25% human serum.


Asunto(s)
Biomarcadores/sangre , Técnicas Biosensibles , Infecciones Neumocócicas/sangre , Streptococcus pneumoniae/aislamiento & purificación , Aptámeros de Nucleótidos/química , Oro/química , Humanos , Péptidos/genética , Péptidos/aislamiento & purificación , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad
7.
Mol Med Rep ; 20(6): 5345-5352, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31638229

RESUMEN

Streptococcus pneumoniae­induced pneumonia is a common disease and major cause of community­acquired pneumonia. Previous studies have shown that Follistatin­like protein 1 (FSTL­1) serves important roles in regulating the inflammatory response. The present study aimed to investigate the effect of FSTL­1 on the inflammatory response during S. pneumoniae infection using in vitro and in vivo models. ELISAs were used to detect the production of interleukin (IL)­1ß, tumor necrosis factor­α and IL­6. Western blotting and reverse transcription­quantitative PCR were performed to determine the protein and mRNA expression of these factors. The results of the present study indicated that S. pneumoniae infection triggered a strong proinflammatory response and a high level of FSTL­1 expression in mouse bone marrow­derived macrophages. Moreover, FSTL­1 may be required for the production of inflammatory factors during S. pneumoniae infection by regulating nucleotide oligomerization domain­like receptor protein 3 in vitro and in vivo. In addition, it was found that the Toll­like receptor 4/nuclear factor­κB signaling pathway was involved in the inflammatory response regulated by FSTL­1. The findings of the present study suggested that FSTL­1 plays an important role in the inflammatory response during S. pneumoniae infection, providing a potential therapeutic target for reducing morbidity and mortality in patients with pneumonia.


Asunto(s)
Proteínas Relacionadas con la Folistatina/genética , Regulación de la Expresión Génica , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Masculino , Ratones Noqueados , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Transducción de Señal
8.
Front Immunol ; 10: 615, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31019504

RESUMEN

Pore-forming toxin (PFT) induced necroptosis exacerbates pulmonary injury during bacterial pneumonia. However, its role during asymptomatic nasopharyngeal colonization and toward the development of protective immunity was unknown. Using a mouse model of Streptococcus pneumoniae (Spn) asymptomatic colonization, we determined that nasopharyngeal epithelial cells (nEC) died of pneumolysin (Ply)-dependent necroptosis. Mice deficient in MLKL, the necroptosis effector, or challenged with Ply-deficient Spn showed less nEC sloughing, increased neutrophil infiltration, and altered IL-1α, IL-33, CXCL2, IL-17, and IL-6 levels in nasal lavage fluid (NALF). Activated MLKL correlated with increased presence of CD11c+ antigen presenting cells in Spn-associated submucosa. Colonized MLKL KO mice and wildtype mice colonized with Ply-deficient Spn produced less antibody against the bacterial surface protein PspA, were delayed in bacterial clearance, and were more susceptible to a lethal secondary Spn challenge. We conclude that PFT-induced necroptosis is instrumental in the natural development of protective immunity against opportunistic PFT-producing bacterial pathogens.


Asunto(s)
Inmunidad Adaptativa , Necroptosis/inmunología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/inmunología , Animales , Formación de Anticuerpos/inmunología , Apoptosis/inmunología , Proteínas Bacterianas/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Femenino , Humanos , Inmunidad Innata , Masculino , Ratones , Necroptosis/genética , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/metabolismo , Estreptolisinas/inmunología
9.
Infect Immun ; 87(6)2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30910792

RESUMEN

CD4+ T-cell mechanisms are implied in protection against pneumococcal colonization; however, their target antigens and function are not well defined. In contrast to high-throughput protein arrays for serology, basic antigen tools for CD4+ T-cell studies are lacking. Here, we evaluate the potential of a bioinformatics tool for in silico prediction of immunogenicity as a method to reveal domains of pneumococcal proteins targeted by human CD4+ T cells. For 100 pneumococcal proteins, CD4+ T-cell immunogenicity was predicted based on HLA-DRB1 binding motifs. For 20 potentially CD4+ T-cell immunogenic proteins, epitope regions were verified by testing synthetic peptides in T-cell assays using peripheral blood mononuclear cells from healthy adults. Peptide pools of 19 out of 20 proteins evoked T-cell responses. The most frequent responses (detectable in ≥20% of donors tested) were found to SP_0117 (PspA), SP_0468 (putative sortase), SP_0546 (BlpZ), SP_1650 (PsaA), SP_1923 (Ply), SP_2048 (conserved hypothetical protein), SP_2216 (PscB), and SPR_0907 (PhtD). Responding donors had diverging recognition patterns and profiles of signature cytokines (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], interleukin-13 [IL-13], and/or IL-17A) against single-epitope regions. Natural HLA-DR-restricted presentation and recognition of a predicted SP_1923-derived epitope were validated through the isolation of a CD4+ T-cell clone producing IFN-γ, TNF-α, and IL-17A in response to the synthetic peptide, whole protein, and heat-inactivated pneumococcus. This proof of principle for a bioinformatics tool to identify pneumococcal protein epitopes targeted by human CD4+ T cells provides a peptide-based strategy to study cell-mediated immune mechanisms for the pneumococcal proteome, advancing the development of immunomonitoring assays and targeted vaccine approaches.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/inmunología , Proteínas Bacterianas/genética , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Leucocitos Mononucleares/inmunología , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/inmunología , Dominios Proteicos , Streptococcus pneumoniae/química , Streptococcus pneumoniae/genética
10.
Front Immunol ; 9: 1954, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30214444

RESUMEN

Activating transcription factor-3 (ATF3) in the ER stress pathway induces cytokine production and promotes survival during gram-positive bacterial infection. IL-17A is a critical cytokine that is essential for clearance of Streptococcus pneumoniae. However, the mechanism by which ATF3 induces IL-17A production remains unknown. Here, we show that ATF3 induces IL-17A production via NLRP3 inflammasome-dependent IL-1ß secretion. Survival rates were comparable in IL-17A-depleted and ATF3 KO mice but were lower than in WT mice treated with isotype control, indicating that ATF3 positively regulated IL-17A production. Indeed, ATF3 KO mice showed a marked reduction in IL-17A protein and mRNA expression compared to levels in WT mice. Moreover, mitochondrial IL-1ß production by bone marrow-derived macrophages was significantly reduced in ATF3 KO mice as a result of the disruption of cellular ROS and Ca2+ homeostasis. Accordingly, ATF3 KO mice displayed diminished survival and bacterial clearance following S. pneumoniae infection. Taken together, these data suggest a mechanism in which macrophage ATF3 promotes IL-17A production in γδ T cells to rapidly induce host defenses during early S. pneumoniae infection.


Asunto(s)
Factor de Transcripción Activador 3/inmunología , Señalización del Calcio/inmunología , Interleucina-17/inmunología , Interleucina-1beta/inmunología , Infecciones Neumocócicas/inmunología , Especies Reactivas de Oxígeno/inmunología , Streptococcus pneumoniae/inmunología , Factor de Transcripción Activador 3/genética , Animales , Calcio/inmunología , Señalización del Calcio/genética , Femenino , Interleucina-17/genética , Interleucina-1beta/genética , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Infecciones Neumocócicas/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Linfocitos T/patología
11.
Sci Rep ; 8(1): 5723, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636524

RESUMEN

The recruitment of myeloid cells to the lung is of utmost importance for the elimination of invading pathogens. We investigated the Streptococcus pneumoniae-dependent induction mechanism of KLF4 in macrophages as a potential regulator of the macrophage immune response. We demonstrated that only viable pneumococci, which have direct contact to the host cells and release LytA-dependent DNA, induced KLF4. Exogenous supplementation of pneumococcal, other bacterial, eukaryotic foreign (human) or self (mouse) DNA to autolysis-deficient pneumococci restored (at least in part) pneumococci-related KLF4 induction. Experiments using TLR9, TRIF and MyD88 knockout macrophages revealed that TLR9, TRIF and MyD88 were partly involved in the S. pneumoniae-induced KLF4 expression. BMMs missing important DNA receptor related molecules (ASC-/-, STING-/-) showed no differences in pneumococci-related KLF4 expression. Similar results were observed with IFNAR-/- BMMs and Type I IFN stimulated cells. LyzMcre mediated knockdown of KLF4 in BMMs resulted in a decreased secretion of proinflammatory cytokines and enhanced IL-10 release. In summary, we showed that pneumococci-related KLF4 induction in macrophages is mediated via a PAMP-DAMP induction mechanism involving a hitherto unknown host cell DNA sensor leading to a more proinflammatory macrophage phenotype.


Asunto(s)
ADN Bacteriano/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Macrófagos/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/fisiología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Comunicación Autocrina , Cápsulas Bacterianas/inmunología , Citocinas/metabolismo , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Factor 4 Similar a Kruppel , Macrófagos/inmunología , Ratones , Comunicación Paracrina , Fagocitosis/inmunología , Infecciones Neumocócicas/inmunología , Receptor Toll-Like 9/metabolismo
12.
Cell Immunol ; 325: 14-22, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29366563

RESUMEN

Streptococcus pneumoniae is a leading cause of bacterial pneumonia. Our previous study suggested that S. pneumoniae autolysis-dependently releases intracellular pneumolysin, which subsequently leads to lung injury. In this study, we hypothesized that pneumococcal autolysis induces the leakage of additional intracellular molecules that could increase the pathogenicity of S. pneumoniae. Liquid chromatography tandem-mass spectrometry analysis identified that chaperone protein DnaK, elongation factor Tu (EF-Tu), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were released with pneumococcal DNA by autolysis. We demonstrated that recombinant (r) DnaK, rEF-Tu, and rGAPDH induced significantly higher levels of interleukin-6 and tumor necrosis factor production in peritoneal macrophages and THP-1-derived macrophage-like cells via toll-like receptor 4. Furthermore, the DNA-binding activity of these proteins was confirmed by surface plasmon resonance assay. We demonstrated that pneumococcal DnaK, EF-Tu, and GAPDH induced the production of proinflammatory cytokines in macrophages, and might cause host tissue damage and affect the development of pneumococcal diseases.


Asunto(s)
Autólisis/metabolismo , Proteínas de Unión al ADN/metabolismo , Streptococcus pneumoniae/metabolismo , Animales , Proteínas Bacterianas , Cromatografía Liquida/métodos , Citocinas/metabolismo , Proteínas de Unión al ADN/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Chaperonas Moleculares/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , Infecciones Neumocócicas/genética , Streptococcus pneumoniae/genética , Células THP-1 , Espectrometría de Masas en Tándem/métodos , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
13.
Infect Immun ; 86(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29263110

RESUMEN

Streptococcus pneumoniae is a frequent colonizer of the upper respiratory tract and a leading cause of bacterial pneumonia. The innate immune system senses pneumococcal cell wall components, toxin, and nucleic acids, which leads to production of inflammatory mediators to initiate and control antibacterial defense. Here, we show that the cGAS (cyclic GMP-AMP [cGAMP] synthase)-STING pathway mediates detection of pneumococcal DNA in mouse macrophages to primarily stimulate type I interferon (IFN) responses. Cells of human individuals carrying HAQ TMEM173, which encodes a common hypomorphic variant of STING, were largely or partly defective in inducing type I IFNs and proinflammatory cytokines upon infection. Subsequent analyses, however, revealed that STING was dispensable for restricting S. pneumoniae during acute pneumonia in mice. Moreover, explorative analyses did not find differences in the allele frequency of HAQ TMEM173 in nonvaccinated pneumococcal pneumonia patients and healthy controls or an association of HAQ TMEM173 carriage with disease severity. Together, our results indicate that the cGAS/STING pathway senses S. pneumoniae but plays no major role in antipneumococcal immunity in mice and humans.


Asunto(s)
Proteínas de la Membrana/inmunología , Nucleotidiltransferasas/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Adulto , Anciano , Animales , Estudios de Cohortes , Femenino , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Macrófagos/inmunología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Nucleotidiltransferasas/genética , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética
14.
PLoS Pathog ; 13(8): e1006582, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28841717

RESUMEN

For over 130 years, invasive pneumococcal disease has been associated with the presence of extracellular planktonic pneumococci, i.e. diplococci or short chains in affected tissues. Herein, we show that Streptococcus pneumoniae that invade the myocardium instead replicate within cellular vesicles and transition into non-purulent biofilms. Pneumococci within mature cardiac microlesions exhibited salient biofilm features including intrinsic resistance to antibiotic killing and the presence of an extracellular matrix. Dual RNA-seq and subsequent principal component analyses of heart- and blood-isolated pneumococci confirmed the biofilm phenotype in vivo and revealed stark anatomical site-specific differences in virulence gene expression; the latter having major implications on future vaccine antigen selection. Our RNA-seq approach also identified three genomic islands as exclusively expressed in vivo. Deletion of one such island, Region of Diversity 12, resulted in a biofilm-deficient and highly inflammogenic phenotype within the heart; indicating a possible link between the biofilm phenotype and a dampened host-response. We subsequently determined that biofilm pneumococci released greater amounts of the toxin pneumolysin than did planktonic or RD12 deficient pneumococci. This allowed heart-invaded wildtype pneumococci to kill resident cardiac macrophages and subsequently subvert cytokine/chemokine production and neutrophil infiltration into the myocardium. This is the first report for pneumococcal biofilm formation in an invasive disease setting. We show that biofilm pneumococci actively suppress the host response through pneumolysin-mediated immune cell killing. As such, our findings contradict the emerging notion that biofilm pneumococci are passively immunoquiescent.


Asunto(s)
Biopelículas , Macrófagos/inmunología , Miocarditis/inmunología , Miocarditis/microbiología , Infecciones Neumocócicas/inmunología , Transcriptoma , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Infecciones Neumocócicas/genética , Análisis de Componente Principal , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/inmunología , Virulencia/genética , Virulencia/inmunología
15.
Infect Immun ; 85(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28808157

RESUMEN

Pulmonary infection by Streptococcus pneumoniae is characterized by a robust alveolar infiltration of neutrophils (polymorphonuclear cells [PMNs]) that can promote systemic spread of the infection if not resolved. We previously showed that 12-lipoxygenase (12-LOX), which is required to generate the PMN chemoattractant hepoxilin A3 (HXA3) from arachidonic acid (AA), promotes acute pulmonary inflammation and systemic infection after lung challenge with S. pneumoniae As phospholipase A2 (PLA2) promotes the release of AA, we investigated the role of PLA2 in local and systemic disease during S. pneumoniae infection. The group IVA cytosolic isoform of PLA2 (cPLA2α) was activated upon S. pneumoniae infection of cultured lung epithelial cells and was critical for AA release from membrane phospholipids. Pharmacological inhibition of this enzyme blocked S. pneumoniae-induced PMN transepithelial migration in vitro Genetic ablation of the cPLA2 isoform cPLA2α dramatically reduced lung inflammation in mice upon high-dose pulmonary challenge with S. pneumoniae The cPLA2α-deficient mice also suffered no bacteremia and survived a pulmonary challenge that was lethal to wild-type mice. Our data suggest that cPLA2α plays a crucial role in eliciting pulmonary inflammation during pneumococcal infection and is required for lethal systemic infection following S. pneumoniae lung challenge.


Asunto(s)
Células Epiteliales/inmunología , Fosfolipasas A2 Grupo IV/inmunología , Interacciones Huésped-Patógeno , Pulmón/inmunología , Infecciones Neumocócicas/inmunología , Neumonía Bacteriana/inmunología , Animales , Ácido Araquidónico/inmunología , Ácido Araquidónico/metabolismo , Bacteriemia/genética , Bacteriemia/inmunología , Bacteriemia/prevención & control , Línea Celular Tumoral , Factores Quimiotácticos/inmunología , Factores Quimiotácticos/metabolismo , Clorobenzoatos/farmacología , Cinamatos/farmacología , Ciclohexanonas/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/microbiología , Fosfolipasas A2 Grupo IV/antagonistas & inhibidores , Fosfolipasas A2 Grupo IV/deficiencia , Fosfolipasas A2 Grupo IV/genética , Humanos , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/microbiología , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/mortalidad , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/mortalidad , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Análisis de Supervivencia , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/inmunología , ortoaminobenzoatos/farmacología
16.
Virulence ; 8(8): 1631-1642, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-28605238

RESUMEN

Streptococcus pneumoniae is a bacterial pathogen that commonly resides in the human nasopharynx, typically without causing any disease. However, in some cases these bacteria migrate from the nasopharynx to other sites of the body such as the lungs and bloodstream causing pneumonia and sepsis, respectively. This study used a mouse model of infection to investigate the potential role of Mucin 1 (MUC1), a cell membrane-associated glycoprotein known for playing a key barrier role at mucosal surfaces, in regulating this process. Wildtype (WT) and MUC1-deficient (Muc1-/-) mice were infected intranasally with an invasive strain of S. pneumoniae and bacterial loads in the nasopharynx, lungs, and blood were analyzed. Lungs were graded histologically for inflammation and cytokine profiles in the lungs analyzed by ELISA. While there was no difference in pneumococcal colonization of the nasopharynx between WT and Muc1-/- mice, infected Muc1-/- mice showed high pneumococcal loads in their lungs 16 hours post-infection, as well as bacteremia. In contrast, infected WT mice cleared the pneumococci from their lungs and remained asymptomatic. Infection in Muc1-/- mice was associated with an elevation in lung inflammation, with cellular recruitment especially of monocytes/macrophages. While MUC1-deficiency has been shown to increase phagocytosis of Pseudomonas aeruginosa, macrophages from Muc1-/- mice exhibited a reduced capacity to phagocytose S. pneumoniae indicating diverse and bacterial-specific effects. In conclusion, these findings indicate that MUC1 plays an important role in protection against severe pneumococcal disease, potentially mediated by facilitating macrophage phagocytosis.


Asunto(s)
Mucina-1/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mucina-1/genética , Nasofaringe/inmunología , Nasofaringe/microbiología , Fagocitosis , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética
17.
Blood ; 129(23): 3087-3099, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28325862

RESUMEN

ß-Thalassemia is associated with several abnormalities of the innate immune system. Neutrophils in particular are defective, predisposing patients to life-threatening bacterial infections. The molecular and cellular mechanisms involved in impaired neutrophil function remain incompletely defined. We used the Hbbth3/+ ß-thalassemia mouse and hemoglobin E (HbE)/ß-thalassemia patients to investigate dysregulated neutrophil activity. Mature neutrophils from Hbbth3/+ mice displayed a significant reduction in chemotaxis, opsonophagocytosis, and production of reactive oxygen species, closely mimicking the defective immune functions observed in ß-thalassemia patients. In Hbbth3/+ mice, the expression of neutrophil CXCR2, CD11b, and reduced NAD phosphate oxidase components (p22phox, p67phox, and gp91phox) were significantly reduced. Morphological analysis of Hbbth3/+ neutrophils showed that a large percentage of mature phenotype neutrophils (Ly6GhiLy6Clow) appeared as band form cells, and a striking expansion of immature (Ly6GlowLy6Clow) hyposegmented neutrophils, consisting mainly of myelocytes and metamyelocytes, was noted. Intriguingly, expression of an essential mediator of neutrophil terminal differentiation, the ets transcription factor PU.1, was significantly decreased in Hbbth3/+ neutrophils. In addition, in vivo infection with Streptococcus pneumoniae failed to induce PU.1 expression or upregulate neutrophil effector functions in Hbbth3/+ mice. Similar changes to neutrophil morphology and PU.1 expression were observed in splenectomized and nonsplenectomized HbE/ß-thalassemia patients. This study provides a mechanistic insight into defective neutrophil maturation in ß-thalassemia patients, which contributes to deficiencies in neutrophil effector functions.


Asunto(s)
Neutrófilos/inmunología , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Talasemia beta/genética , Talasemia beta/inmunología , Adulto , Animales , Antígeno CD11b/metabolismo , Estudios de Casos y Controles , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Activación Neutrófila , Neutrófilos/metabolismo , Neutrófilos/patología , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/inmunología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-8B/metabolismo , Transactivadores/deficiencia , Transactivadores/inmunología , Adulto Joven , Talasemia beta/patología
18.
Infect Immun ; 85(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28193634

RESUMEN

Insights into the host-microbial virulence factor interaction, especially the immune signaling mechanisms, could provide novel prevention and treatment options for pneumococcal diseases. Streptococcus pneumoniae endopeptidase O (PepO) is a newly discovered and ubiquitously expressed pneumococcal virulence protein. A PepO-mutant strain showed impaired adherence to and invasion of host cells compared with the isogenic wild-type strain. It is still unknown whether PepO is involved in the host defense response to pneumococcal infection. Here, we demonstrated that PepO could enhance phagocytosis of Streptococcus pneumoniae and Staphylococcus aureus by peritoneal exudate macrophages (PEMs). Further studies showed that PepO stimulation upregulated the expression of microRNA-155 (miR-155) in PEMs in a time- and dose-dependent manner. PepO-induced enhanced phagocytosis was decreased in cells transfected with an inhibitor of miR-155, while it was increased in cells transfected with a mimic of miR-155. We also revealed that PepO-induced upregulation of miR-155 in PEMs was mediated by Toll-like receptor 2 (TLR2)-NF-κB signaling and that the increased expression of miR-155 downregulated expression of SHIP1. Taken together, these results indicate that PepO induces upregulation of miR-155 in PEMs, contributing to enhanced phagocytosis and host defense response to pneumococci and Staphylococcus aureus.


Asunto(s)
Proteínas Bacterianas/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Metaloendopeptidasas/inmunología , MicroARNs/genética , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 2/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Fagocitosis , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Infecciones Neumocócicas/microbiología , Interferencia de ARN , Transducción de Señal , Staphylococcus aureus/inmunología
19.
Proc Natl Acad Sci U S A ; 112(51): E7128-37, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26621750

RESUMEN

This paper reviews the developments that have occurred in the field of human genetics of infectious diseases from the second half of the 20th century onward. In particular, it stresses and explains the importance of the recently described monogenic inborn errors of immunity underlying resistance or susceptibility to specific infections. The monogenic component of the genetic theory provides a plausible explanation for the occurrence of severe infectious diseases during primary infection. Over the last 20 y, increasing numbers of life-threatening infectious diseases striking otherwise healthy children, adolescents, and even young adults have been attributed to single-gene inborn errors of immunity. These studies were inspired by seminal but neglected findings in plant and animal infections. Infectious diseases typically manifest as sporadic traits because human genotypes often display incomplete penetrance (most genetically predisposed individuals remain healthy) and variable expressivity (different infections can be allelic at the same locus). Infectious diseases of childhood, once thought to be archetypal environmental diseases, actually may be among the most genetically determined conditions of mankind. This nascent and testable notion has interesting medical and biological implications.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Infecciones/genética , Infecciones/inmunología , Adolescente , Candidiasis Mucocutánea Crónica/genética , Candidiasis Mucocutánea Crónica/inmunología , Niño , Proteínas del Sistema Complemento/genética , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/inmunología , Epidermodisplasia Verruciforme/genética , Epidermodisplasia Verruciforme/inmunología , Predisposición Genética a la Enfermedad , Humanos , Gripe Humana/genética , Gripe Humana/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/inmunología , Malaria/genética , Malaria/inmunología , Modelos Genéticos , Modelos Inmunológicos , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/inmunología , Neisseria/inmunología , Neisseria/patogenicidad , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/inmunología , Tiña/genética , Tiña/inmunología , Adulto Joven
20.
PLoS One ; 10(11): e0142773, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26566142

RESUMEN

Streptococcus pneumoniae (Spn) is a major causative organism of empyema, an inflammatory condition occurring in the pleural sac. In this study, we used human and Spn cDNA microarrays to characterize the transcriptional responses occurring during initial contact between Spn and a human pleural mesothelial cell line (PMC) in vitro. Using stringent filtering criteria, 42 and 23 Spn genes were up-and down-regulated respectively. In particular, genes encoding factors potentially involved in metabolic processes and Spn adherence to eukaryotic cells were up-regulated e.g. glnQ, glnA, aliA, psaB, lytB and nox. After Spn initial contact, 870 human genes were differentially regulated and the largest numbers of significant gene expression changes were found in canonical pathways for eukaryotic initiation factor 2 signaling (60 genes out of 171), oxidative phosphorylation (32/103), mitochondrial dysfunction (37/164), eIF4 and p70S6K signaling (28/142), mTOR signaling (27/182), NRF2-mediated oxidative stress response (20/177), epithelial adherens junction remodeling (11/66) and ubiquitination (22/254). The cellular response appeared to be directed towards host cell survival and defense. Spn did not activate NF-kB or phosphorylate p38 MAPK or induce cytokine production from PMC. Moreover, Spn infection of TNF-α pre-stimulated PMC inhibited production of IL-6 and IL-8 secretion by >50% (p<0.01). In summary, this descriptive study provides datasets and a platform for examining further the molecular mechanisms underlying the pathogenesis of empyema.


Asunto(s)
Empiema/microbiología , Epitelio/metabolismo , Epitelio/microbiología , Regulación de la Expresión Génica , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética , Algoritmos , Adhesión Celular , Línea Celular , Citocinas/metabolismo , ADN Complementario/metabolismo , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Humanos , Inflamación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Infecciones Neumocócicas/genética , ARN Mensajero/metabolismo , Transducción de Señal , Transcriptoma , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA