Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 467(1): 46-52, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26417692

RESUMEN

BACKGROUND: Bacterial colonisation with Moraxella catarrhalis may partly sustain chronic inflammation in the lower airways of patients with chronic obstructive pulmonary disease (COPD). In addition, this bacterium causes infectious exacerbations of COPD, which often necessitate treatment with antibiotics. Antimicrobial peptides are the body's own antibiotic substances with bactericidal and bacteriostatic, as well as immunomodulatory function. In particular, human beta-defensin 3 (hBD-3) exerts an antimicrobial effect against an extraordinarily broad spectrum of pathogens. We therefore investigated the role of hBD-3 in infections of pulmonary epithelial cells with M. catarrhalis. METHODS: The antimicrobial activity of hBD-3 vs. M. catarrhalis was evaluated in an antimicrobial susceptibility assay. We analyzed hBD-3 secretion of M. catarrhalis-infected pulmonary epithelial cells using ELISA. The role of M. catarrhalis-specific virulence factors, toll-like receptors (TLR) 2 and 4, MAPK pathways, and transcription factors AP-1 and NF-κB in the induction and regulation of hBD-3 expression were explored with specific inhibitors, small interference RNA, Western Blot, and chromatin immunoprecipitation (ChIP) assays. RESULTS: HBD-3 exhibited a strong bactericidal effect against M. catarrhalis. M. catarrhalis induced hBD-3 expression in pulmonary epithelial cells, which was dependent on M. catarrhalis membranous lipoolygosaccharide (LOS), while the surface proteins UspA1 and UspA2 were not involved. Gene silencing of TLR2, but not TLR4, led to a reduced hBD-3 secretion after stimulation with M. catarrhalis or M. catarrhalis LOS. Inhibition of MAPKs ERK1/2 and JNK, but not p38, reduced hBD-3 secretion. HBD-3 expression was mediated through the recruitment of AP-1 to the hBD-3 gene promoter and was independent of NF-κB. CONCLUSION: The immune response of pulmonary epithelial cells towards M. catarrhalis involves secretion of hBD-3, which has a bactericidal effect against this pathogen. Binding of M. catarrhalis virulence factor LOS to TLR2 causes an ERK1/2- and JNK-dependent induction of AP-1-related transcription of the hBD-3 gene, resulting in the production and secretion of hBD-3.


Asunto(s)
Moraxella catarrhalis/patogenicidad , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , beta-Defensinas/metabolismo , Línea Celular , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Regulación de la Expresión Génica , Humanos , Lipopolisacáridos/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Sistema de Señalización de MAP Quinasas , Moraxella catarrhalis/inmunología , Infecciones por Moraxellaceae/complicaciones , FN-kappa B/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Mucosa Respiratoria/inmunología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Transcripción AP-1/metabolismo , beta-Defensinas/genética
2.
Biochem Biophys Res Commun ; 450(2): 1038-44, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24978309

RESUMEN

BACKGROUND: Chronic lower airway inflammation is considered to be a major cause of pathogenesis and disease progression in chronic obstructive pulmonary disease (COPD). Moraxella catarrhalis is a COPD-associated pathogen causing exacerbations and bacterial colonization in the lower airways of patients, which may contribute to chronic inflammation. Increasing evidence suggests that the epidermal growth factor receptor (EGFR) modulates inflammatory processes in the human airways. The goal of this study was to investigate the role of EGFR in the M. catarrhalis-induced pro-inflammatory immune response in airway epithelial cells. METHODS: The effects of inhibition and gene silencing of EGFR on M. catarrhalis-dependent pro-inflammatory cytokine expression in human primary bronchial epithelial cells (NHBEs), as well as the pulmonary epithelial cell lines BEAS-2B and A549 were analyzed. We also assessed the involvement of EGFR-dependent ERK and NF-κB signaling pathways. RESULTS: The M. catarrhalis-induced pro-inflammatory immune response depends, at least in part, on the phosphorylation and activation of the EGF receptor. Interaction of M. catarrhalis with EGFR increases the secretion of pro-inflammatory cytokines, which is mediated via ERK and NF-κB activation. CONCLUSION: The interaction between M. catarrhalis and EGFR increases airway inflammation caused by this pathogen. Our data suggest that the inhibition of EGFR signaling in COPD could be an interesting target for reducing M. catarrhalis-induced airway inflammation.


Asunto(s)
Células Epiteliales/inmunología , Receptores ErbB/metabolismo , Moraxella catarrhalis/inmunología , Mucosa Respiratoria/inmunología , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Silenciador del Gen , Humanos , Interleucina-8/metabolismo , Viabilidad Microbiana , FN-kappa B/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Transducción de Señal , Especificidad de la Especie
3.
Exp Lung Res ; 38(2): 100-10, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22296408

RESUMEN

Streptococcus pneumoniae is an important causative agent of pneumonia in humans. Pulmonary epithelial surfaces constitutes not only a mechanical barrier against invading pathogens but also essentially contribute to innate immunity by producing antimicrobial peptides such as human ß-defensin-2 (hBD-2) and -3 (hBD-3). In this study the authors demonstrated that pneumococci induced hBD-2 and hBD-3 expression in human pulmonary epithelial cells. Further analysis indicated an essential role of Toll-like receptor 2 (TLR2) for the expression of both peptides in infected pulmonary epithelial cells. Whereas the hBD-2 release was controlled by the phosphoinositide 3-kinase (PI3K) and the transcription factor nuclear factor kappa B (NF-κB), hBD-3 was triggered via the c-Jun N-terminal kinase (JNK)-activator protein 1 (AP-1) pathway. Additionally, the authors showed that exogenous hBD-2 as well as hBD-3 elicited a strong antimicrobial effect on S. pneumoniae. Thus, differential regulation of the expression of hBD-2 and hBD-3 might play an important role in pneumococci pneumonia.


Asunto(s)
Células Epiteliales Alveolares/microbiología , Pulmón/microbiología , Streptococcus pneumoniae/patogenicidad , beta-Defensinas/biosíntesis , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/metabolismo , Humanos , Inmunidad Innata , Pulmón/inmunología , Pulmón/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 2/metabolismo , Factor de Transcripción AP-1/metabolismo
4.
PLoS One ; 7(1): e30379, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22276187

RESUMEN

Chlamydophila pneumoniae causes acute respiratory tract infections and has been associated with development of asthma and atherosclerosis. The production of IL-1ß, a key mediator of acute and chronic inflammation, is regulated on a transcriptional level and additionally on a posttranslational level by inflammasomes. In the present study we show that C. pneumoniae-infected human mononuclear cells produce IL-1ß protein depending on an inflammasome consisting of NLRP3, the adapter protein ASC and caspase-1. We further found that the small GTPase Rac1 is activated in C. pneumoniae-infected cells. Importantly, studies with specific inhibitors as well as siRNA show that Rac1 regulates inflammasome activation in C. pneumoniae-infected cells. In conclusion, C. pneumoniae infection of mononuclear cells stimulates IL-1ß production dependent on a NLRP3 inflammasome-mediated processing of proIL-1ß which is controlled by Rac1.


Asunto(s)
Proteínas Portadoras/metabolismo , Chlamydophila pneumoniae/inmunología , Chlamydophila pneumoniae/fisiología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Monocitos/metabolismo , Monocitos/microbiología , Proteína de Unión al GTP rac1/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Proteína de Unión al GTP rac1/genética
5.
Pulm Pharmacol Ther ; 24(4): 377-85, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21419858

RESUMEN

Lung barrier protection by Sphingosine-1 Phosphate (S1P) has been demonstrated experimentally, but recent evidence suggests barrier disruptive properties of high systemic S1P concentrations. The S1P analog FTY720 recently gained an FDA approval for treatment of multiple sclerosis. In case of FTY720 treated patients experiencing multiple organ dysfunction syndrome the drug may accumulate due to liver failure, and the patients may receive ventilator therapy. Whereas low doses of FTY720 enhanced endothelial barrier function, data on effects of increased FTY720 concentrations are lacking. We measured transcellular electrical resistance (TER) of human umbilical vein endothelial cell (HUVEC) monolayers, performed morphologic analysis and measured apoptosis by TUNEL staining and procaspase-3 degradation in HUVECs stimulated with FTY720 (0.01-100 µM). Healthy C57BL/6 mice and mice ventilated with 17 ml/kg tidal volume and 100% oxygen for 2 h were treated with 0.1 or 2 mg/kg FTY720 or solvent, and lung permeability, oxygenation and leukocyte counts in BAL and blood were quantified. Further, electron microscopic analysis of lung tissue was performed. We observed barrier protective effects of FTY720 on HUVEC cell layers at concentrations up to 1 µM while higher concentrations induced irreversible barrier breakdown accompanied by induction of apoptosis. Low FTY720 concentrations (0.1 mg/kg) reduced lung permeability in mechanically ventilated mice, but 2 mg/kg FTY720 increased pulmonary vascular permeability in ventilated mice accompanied by endothelial apoptosis, while not affecting permeability in non-ventilated mice. Moreover, hyperoxic mechanical ventilation sensitized the pulmonary vasculature to a barrier disrupting effect of FTY720, resulting in worsening of ventilator induced lung injury. In conclusion, the current data suggest FTY720 induced endothelial barrier dysfunction, which was probably caused by proapoptotic effects and enhanced by mechanical ventilation.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Pulmón/efectos de los fármacos , Glicoles de Propileno/toxicidad , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/análogos & derivados , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Animales , Apoptosis/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Clorhidrato de Fingolimod , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Esfingosina/toxicidad
6.
J Immunol ; 185(1): 597-604, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20525885

RESUMEN

The release of potent proinflammatory mediators is not only central for mounting an efficient host response, but also bears the risk for deleterious excessive tissue-damaging inflammation. This is highlighted in severe pneumococcal pneumonia, in which the delicate balance between a robust inflammatory response to kill pneumococci and loss of organ function determines the outcome of disease. In this study, we tested the hypothesis that Krüppel-like factor (KLF)2 counterregulates pneumococci- and pattern recognition receptor-related human lung cell activation. Pneumococci induced KLF2 expression in vitro and in a murine pneumonia model. Activation of TLR2- and nucleotide-binding oligomerization domain protein 2-related signaling induced KLF2 expression in a PI3K-dependent manner. Overexpression of KLF2 downregulated pneumococci-, TLR2-, and nucleotide-binding oligomerization domain protein 2-related NF-kappaB-dependent gene expression and IL-8 release, whereas small interfering RNA-based silencing of KLF2 provoked an enhanced inflammatory response. KLF2-dependent downregulation of NF-kappaB activity is partly reversible by overexpression of the histone acetylase p300/CREB-binding protein-associated factor. In conclusion, KLF2 may act as a counterregulatory transcription factor in pneumococci- and pattern recognition receptor-related proinflammatory activation of lung cells, thereby preventing lung hyperinflammation and subsequent organ failure.


Asunto(s)
Regulación hacia Abajo/inmunología , Regulación de la Expresión Génica/inmunología , Mediadores de Inflamación/fisiología , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/fisiología , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Proteína Adaptadora de Señalización NOD2/fisiología , Neumonía Neumocócica/inmunología , Receptor Toll-Like 2/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/inmunología , Insuficiencia Multiorgánica/prevención & control , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Neumonía Neumocócica/genética , Neumonía Neumocócica/prevención & control , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/genética
7.
Am J Physiol Lung Cell Mol Physiol ; 298(5): L687-95, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20154223

RESUMEN

Legionella pneumophila is an important causative agent of severe pneumonia in humans. Human alveolar epithelium is an effective barrier for inhaled microorganisms and actively participates in the initiation of innate host defense. Induction of antimicrobial peptide human ß-defensin-2 (hBD-2) by various stimuli in epithelial cells has been reported. However, the mechanisms by which bacterial infections enhance hBD-2 expression remain poorly understood. In this study, we investigated the effect of the pulmonary pathogen L. pneumophila on induction of hBD-2 in human pulmonary epithelial cells. Infection with L. pneumophila markedly increased hBD-2 production, and the response was attenuated in Toll-like receptor (TLR) 2 and TLR5 transient knockdown cells. Furthermore, pretreatment with SB-202190 (an inhibitor of p38 MAPK) and JNK II (an inhibitor of c-Jun NH(2)-terminal kinase), but not U0126 (an inhibitor of ERK), reduced L. pneumophila-induced hBD-2 release in A549 cells. L. pneumophila-induced hBD-2 liberation was mediated via recruitment of NF-κB and AP-1 to the hBD-2 gene promoter. Additionally, we showed that exo- and endogenous hBD-2 elicited a strong antimicrobial effect towards L. pneumophila. Together, these results suggest that L. pneumophila induces hBD-2 release in A549 cells, and the induction seems to be mediated through TLR2 and TLR5 as well as activation of p38 MAPK, JNK, NF-κB, and AP-1.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/microbiología , Legionella pneumophila/patogenicidad , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 5/metabolismo , beta-Defensinas/biosíntesis , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/inmunología , Secuencia de Bases , Línea Celular , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Legionella pneumophila/efectos de los fármacos , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/metabolismo , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/farmacología , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/genética , Receptor Toll-Like 5/antagonistas & inhibidores , Receptor Toll-Like 5/genética , Factor de Transcripción AP-1/metabolismo , beta-Defensinas/inmunología , beta-Defensinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Cell Microbiol ; 10(12): 2579-88, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18771559

RESUMEN

Intracellular bacteria and cytosolic stimulation with DNA activate type I IFN responses independently of Toll-like receptors, most Nod-like receptors and RIG-like receptors. A recent study suggested that ZBP1 (DLM-1/DAI) represents the long anticipated pattern recognition receptor which mediates IFNalpha/beta responses to cytosolic DNA in mice. Here we show that Legionella pneumophila infection, and intracellular challenge with poly(dA-dT), but not with poly(dG-dC), induced expression of IFNbeta, full-length hZBP1 and a prominent splice variant lacking the first Zalpha domain (hZBP1DeltaZalpha) in human cells. Overexpression of hZBP1 but not hZBP1DeltaZalpha slightly amplified poly(dA-dT)-stimulated IFNbeta reporter activation in HEK293 cells, but had no effect on IFNbeta and IL-8 production induced by bacteria or poly(dA-dT) in A549 cells. We found that mZBP1 siRNA impaired poly(dA-dT)-induced IFNbeta responses in mouse L929 fibroblasts at a later time point, while multiple hZBP1 siRNAs did not suppress IFNbeta or IL-8 expression induced by poly(dA-dT) or bacterial infection in human cells. In contrast, IRF3 siRNA strongly impaired the IFNbeta responses to poly(dA-dT) or bacterial infection. In conclusion, intracellular bacteria and cytosolic poly(dA-dT) activate IFNbeta responses in different human cells without requiring human ZBP1.


Asunto(s)
ADN Bacteriano/inmunología , Proteínas de Unión al ADN/metabolismo , Interferón beta/biosíntesis , Legionella pneumophila/inmunología , Animales , Línea Celular , Humanos , Interleucina-8/biosíntesis , Ratones , Proteínas de Unión al ARN
9.
J Immunol ; 181(4): 2664-71, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684957

RESUMEN

The nucleotide-binding domain and leucine-rich repeat containing protein NOD2 serves as a cytoplasmic pattern recognition molecule sensing bacterial muramyl dipeptide (MDP), whereas TLR2 mediates cell surface recognition of bacterial lipopeptides. In this study, we show that NOD2 stimulation activated Rac1 in human THP-1 cells and primary human monocytes. Rac1 inhibition or knock-down, or actin cytoskeleton disruption increased MDP-stimulated IL-8 secretion and NF-kappaB activation, whereas TLR2-dependent cell activation was suppressed by Rac1 inhibition. p21-activated kinase [Pak]-interacting exchange factor (beta-PIX) plays a role in this negative regulation, because knock-down of beta-PIX also led to increased NOD2-mediated but not TLR2-mediated IL-8 secretion, and coimmunoprecipitation experiments demonstrated that NOD2 interacted with beta-PIX as well as Rac1 upon MDP stimulation. Moreover, knock-down of beta-PIX or Rac1 abrogated membrane recruitment of NOD2, and interaction of NOD2 with its negative regulator Erbin. Overall, our data indicate that beta-PIX and Rac1 mediate trafficking and negative regulation of NOD2-dependent signaling which is different from Rac1's positive regulatory role in TLR2 signaling.


Asunto(s)
Regulación hacia Abajo , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteína Adaptadora de Señalización NOD2/antagonistas & inhibidores , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteína de Unión al GTP rac1/fisiología , Línea Celular , Células Cultivadas , Regulación hacia Abajo/inmunología , Activación Enzimática/inmunología , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Proteína Adaptadora de Señalización NOD2/agonistas , Proteína Adaptadora de Señalización NOD2/fisiología , Transporte de Proteínas/inmunología , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal/inmunología , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética
10.
J Immunol ; 181(2): 940-7, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606645

RESUMEN

Legionella pneumophila causes severe pneumonia. Acetylation of histones is thought to be an important regulator of gene transcription, but its impact on L. pneumophila-induced expression of proinflammatory cytokines is unknown. L. pneumophila strain 130b induced the expression of the important chemoattractant IL-8 and genome-wide histone modifications in human lung epithelial A549 cells. We analyzed the IL-8-promoter and found that histone H4 was acetylated and H3 was phosphorylated at Ser(10) and acetylated at Lys(14), followed by transcription factor NF-kappaB. Recruitment of RNA polymerase II to the IL-8 promoter corresponded with increases in gene transcription. Histone modification and IL-8 release were dependent on p38 kinase and NF-kappaB pathways. Legionella-induced IL-8 expression was decreased by histone acetylase (HAT) inhibitor anacardic acid and enhanced by histone deacetylase (HDAC) inhibitor trichostatin A. After Legionella infection, HATs p300 and CREB-binding protein were time-dependently recruited to the IL-8 promoter, whereas HDAC1 and HDAC5 first decreased and later reappeared at the promoter. Legionella specifically induced expression of HDAC5 but not of other HDACs in lung epithelial cells, but knockdown of HDAC1 or 5 did not alter IL-8 release. Furthermore, Legionella-induced cytokine release, promoter-specific histone modifications, and RNA polymerase II recruitment were reduced in infection with flagellin-deletion mutants. Legionella-induced histone modification as well as HAT-/HDAC-dependent IL-8 release could also be shown in primary lung epithelial cells. In summary, histone acetylation seems to be important for the regulation of proinflammatory gene expression in L. pneumophila infected lung epithelial cells. These pathways may contribute to the host response in Legionnaires' disease.


Asunto(s)
Células Epiteliales/inmunología , Flagelina/metabolismo , Histonas/metabolismo , Interleucina-8/metabolismo , Legionella pneumophila/inmunología , FN-kappa B/metabolismo , Alveolos Pulmonares/inmunología , Acetilación , Ácidos Anacárdicos/farmacología , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Interleucina-8/genética , Interleucina-8/inmunología , Legionella pneumophila/efectos de los fármacos , Legionella pneumophila/metabolismo , FN-kappa B/inmunología , Regiones Promotoras Genéticas , Inhibidores de la Síntesis de la Proteína/farmacología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/microbiología , ARN Polimerasa II/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Immunol ; 180(10): 6808-15, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18453601

RESUMEN

In mice, different alleles of the mNAIP5 (murine neuronal apoptosis inhibitory protein-5)/mBirc1e gene determine whether macrophages restrict or support intracellular replication of Legionella pneumophila, and whether a mouse is resistant or (moderately) susceptible to Legionella infection. In the resistant mice strains, the nucleotide-binding oligomerization domain (Nod)-like receptor (NLR) family member mNAIP5/mBirc1e, as well as the NLR protein mIpaf (murine ICE protease-activating factor), are involved in recognition of Legionella flagellin and in restriction of bacterial replication. Human macrophages and lung epithelial cells support L. pneumophila growth, and humans can develop severe pneumonia (Legionnaires disease) after Legionella infection. The role of human orthologs to mNAIP5/mBirc1e and mIpaf in this bacterial infection has not been elucidated. Herein we demonstrate that flagellin-deficient L. pneumophila replicate more efficiently in human THP-1 macrophages, primary monocyte-derived macrophages, and alveolar macrophages, and in A549 lung epithelial cells compared with wild-type bacteria. Additionally, we note expression of the mNAIP5 ortholog hNAIP in all cell types examined, and expression of hIpaf in human macrophages. Gene silencing of hNAIP or hIpaf in macrophages or of hNAIP in lung epithelial cells leads to an enhanced bacterial growth, and overexpression of both molecules strongly reduces Legionella replication. In contrast to experiments with wild-type L. pneumophila, hNAIP or hIpaf knock-down affects the (enhanced) replication of flagellin-deficient Legionella only marginally. In conclusion, hNAIP and hIpaf mediate innate intracellular defense against flagellated Legionella in human cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Células Epiteliales/microbiología , Legionella pneumophila/crecimiento & desarrollo , Enfermedad de los Legionarios/inmunología , Macrófagos/microbiología , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Western Blotting , Línea Celular , Proliferación Celular , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Flagelina/genética , Flagelina/metabolismo , Humanos , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/metabolismo , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología
12.
Circ Res ; 102(8): 888-95, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18309103

RESUMEN

Inflammatory activation of the endothelium by Chlamydophila pneumoniae infection has been implicated in the development of chronic vascular lesions and coronary heart disease by seroepidemiological and animal studies. We tested the hypothesis that C. pneumoniae induced inflammatory gene expression is regulated by Rho-GTPase-related histone modifications. C. pneumoniae infection induced the liberation of proinflammatory interleukin-6, interleukin-8, granulocyte colony-stimulating factor, macrophage inflammatory protein-1beta, granulocyte/macrophage colony-stimulating factor, and interferon-gamma by human endothelial cells. Cytokine secretion was reduced by simvastatin and the specific Rac1 inhibitor NSC23766 but was synergistically enhanced by inhibitors of histone deacetylases trichostatin A and suberoylanilide hydroxamic acid. Infection of endothelial cells with viable C. pneumoniae, but not exposure to heat-inactivated C. pneumoniae or infection with C. trachomatis, induced acetylation of histone H4 and phosphorylation and acetylation of histone H3. Pretreatment of C. pneumoniae-infected cells with simvastatin or NSC23766 reduced global histone modifications as well as specific modifications at the il8 gene promoter, as shown by chromatin immunoprecipitation. Reduced recruitment of nuclear factor kappaB p65/RelA as well as of RNA polymerase II was observed in statin-treated cells. Taken together, Rac1-mediated histone modifications seem to play an important role in C. pneumoniae-induced cytokine production by human endothelial cells.


Asunto(s)
Chlamydophila pneumoniae/fisiología , Histonas/metabolismo , Simvastatina/farmacología , Células Cultivadas , Chlamydophila pneumoniae/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/metabolismo , Células Endoteliales/microbiología , Expresión Génica/efectos de los fármacos , Humanos , Proteína de Unión al GTP rac1/metabolismo
13.
J Infect Dis ; 195(11): 1651-60, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17471435

RESUMEN

Moraxella catarrhalis is a major cause of exacerbations of chronic obstructive pulmonary disease (COPD) and emphysema. M. catarrhalis-specific UspA1 and the epithelial carcinoembryonic antigen-related cell adhesion molecule (CEACAM1) were required to induce apoptosis. M. catarrhalis-induced apoptosis was significantly enhanced in HeLa cells stably transfected with CEACAM1, compared with HeLa cells not expressing CEACAM1. Infected cells showed increased activity of caspases 3, 6, and 9 but not of caspase 8. Reduced expression of Bcl-2, translocation of Bax into the mitochondria, and cytosolic increase of apoptosis-inducing factor in M. catarrhalis-infected cells implicated the involvement of mitochondrial death pathways. In conclusion, M. catarrhalis induced apoptosis in pulmonary epithelial cells--a process that was triggered by interaction between CEACAM1 and UspA1. Thus, M. catarrhalis-induced apoptosis of pulmonary epithelial cells may contribute to the development of COPD and emphysema.


Asunto(s)
Antígenos CD/metabolismo , Apoptosis/fisiología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/microbiología , Moraxella catarrhalis/patogenicidad , Alveolos Pulmonares/microbiología , Línea Celular , Células HeLa , Humanos , Moraxella catarrhalis/metabolismo , Alveolos Pulmonares/citología
14.
Am J Physiol Lung Cell Mol Physiol ; 292(1): L267-77, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17012371

RESUMEN

Legionella pneumophila causes community- and hospital-acquired pneumonia. Lung airway and alveolar epithelial cells comprise an important barrier against airborne pathogens. Cyclooxygenase (COX) and microsomal PGE(2) synthase-1 (mPGES-1)-derived prostaglandins like prostaglandin E(2) (PGE(2)) are considered as important regulators of lung function. Herein we tested the hypothesis that L. pneumophila induced COX-2 and mPGES-1-dependent PGE(2) production in pulmonary epithelial cells. Legionella induced the release of PGE(2) in primary human small airway epithelial cells and A549 cells. This was accompanied by an increased expression of COX-2 and mPGES-1 as well as an increased PLA(2) activity in infected cells. Deletion of the type IV secretion system Dot/Icm did not impair Legionella-related COX-2 expression or PGE(2) release in A549 cells. L. pneumophila induced the degradation of IkappaBalpha and activated NF-kappaB. Inhibition of IKK blocked L. pneumophila-induced PGE(2) release and COX-2 expression. We noted activation of p38 and p42/44 MAP kinase in Legionella-infected A549 cells. Moreover, membrane translocation and activation of PKCalpha was observed in infected cells. PKCalpha and p38 and p42/44 MAP kinase inhibitors reduced PGE(2) release and COX-2 expression. In summary, PKCalpha and p38 and p42/44 MAP kinase controlled COX-2 expression and subsequent PGE(2) release by Legionella-infected lung epithelial cells. These pathways may significantly contribute to the host response in Legionnaires' disease.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Legionella pneumophila/patogenicidad , Pulmón/metabolismo , Pulmón/microbiología , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Proteína Quinasa C-alfa/metabolismo , Secuencia de Bases , Línea Celular , Células Cultivadas , ADN Complementario/genética , Dinoprostona/biosíntesis , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Oxidorreductasas Intramoleculares/genética , Enfermedad de los Legionarios/etiología , Enfermedad de los Legionarios/metabolismo , Pulmón/citología , Fosfolipasas A/metabolismo , Prostaglandina-E Sintasas
15.
J Biol Chem ; 281(47): 36173-9, 2006 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-16984921

RESUMEN

Legionella pneumophila, a Gram-negative facultative intracellular bacterium, causes severe pneumonia (Legionnaires' disease). Type I interferons (IFNs) were so far associated with antiviral immunity, but recent studies also indicated a role of these cytokines in immune responses against (intracellular) bacteria. Here we show that wild-type L. pneumophila and flagellin-deficient Legionella, but not L. pneumophila lacking a functional type IV secretion system Dot/Icm, or heat-inactivated Legionella induced IFNbeta expression in human lung epithelial cells. We found that factor (IRF)-3 and NF-kappaB-p65 translocated into the nucleus and bound to the IFNbeta gene enhancer after L. pneumophila infection of lung epithelial cells. RNA interference demonstrated that in addition to IRF3, the caspase recruitment domain (CARD)-containing adapter molecule IPS-1 (interferon-beta promoter stimulator 1) is crucial for L. pneumophila-induced IFNbeta expression, whereas other CARD-possessing molecules, such as RIG-I (retinoic acid-inducible protein I), MDA5 (melanoma differentiation-associated gene 5), Nod27 (nucleotide-binding oligomerization domain protein 27), and ASC (apoptosis-associated speck-like protein containing a CARD) seemed not to be involved. Finally, bacterial multiplication assays in small interfering RNA-treated cells indicated that IPS-1, IRF3, and IFNbeta were essential for the control of intracellular replication of L. pneumophila in lung epithelial cells. In conclusion, we demonstrated a critical role of IPS-1, IRF3, and IFNbeta in Legionella infection of lung epithelium.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Epiteliales/citología , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Legionella pneumophila/fisiología , Pulmón/citología , Pulmón/microbiología , Transporte Activo de Núcleo Celular , Diferenciación Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Células Epiteliales/metabolismo , Humanos , Legionella pneumophila/metabolismo , Pulmón/metabolismo , FN-kappa B/metabolismo , ARN/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Respir Res ; 7: 98, 2006 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-16834785

RESUMEN

BACKGROUND: Although pneumococcal pneumonia is one of the most common causes of death due to infectious diseases, little is known about pneumococci-lung cell interaction. Herein we tested the hypothesis that pneumococci activated pulmonary epithelial cell cytokine release by c-Jun-NH2-terminal kinase (JNK) METHODS: Human bronchial epithelial cells (BEAS-2B) or epithelial HEK293 cells were infected with S. pneumoniae R6x and cytokine induction was measured by RT-PCR, ELISA and Bioplex assay. JNK-phosphorylation was detected by Western blot and nuclear signaling was assessed by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP). JNK was modulated by the small molecule inhibitor SP600125 and AP1 by transfection of a dominant negative mutant. RESULTS: S. pneumoniae induced the release of distinct CC and CXC, as well as Th1 and Th2 cytokines and growth factors by human lung epithelial cell line BEAS-2B. Furthermore, pneumococci infection resulted in JNK phosphorylation in BEAS-2B cells. Inhibition of JNK by small molecule inhibitor SP600125 reduced pneumococci-induced IL-8 mRNA expression and release of IL-8 and IL-6. One regulator of the il8 promoter is JNK-phosphorylated activator protein 1 (AP-1). We showed that S. pneumoniae time-dependently induced DNA binding of AP-1 and its phosphorylated subunit c-Jun with a maximum at 3 to 5 h after infection. Recruitment of Ser63/73-phosphorylated c-Jun and RNA polymerase II to the endogenous il8 promoter was found 2 h after S. pneumoniae infection by chromatin immunoprecipitation. AP-1 repressor A-Fos reduced IL-8 release by TLR2-overexpressing HEK293 cells induced by pneumococci but not by TNFalpha. Antisense-constructs targeting the AP-1 subunits Fra1 and Fra2 had no inhibitory effect on pneumococci-induced IL-8 release. CONCLUSION: S. pneumoniae-induced IL-8 expression by human epithelial BEAS-2B cells depended on activation of JNK and recruitment of phosphorylated c-Jun to the il8 promoter.


Asunto(s)
Células Epiteliales/microbiología , Interleucina-8/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/microbiología , Streptococcus pneumoniae , Factor de Transcripción AP-1/metabolismo , Antracenos/farmacología , Línea Celular , ADN/genética , ADN/metabolismo , Activación Enzimática , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Interleucina-8/genética , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Pulmón/inmunología , Pulmón/metabolismo , Fosforilación , Regiones Promotoras Genéticas/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-jun/metabolismo
17.
Am J Physiol Lung Cell Mol Physiol ; 290(6): L1131-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16414978

RESUMEN

Streptococcus pneumoniae is a major cause of community-acquired pneumonia and death from infectious diseases in industrialized countries. Lung airway and alveolar epithelial cells comprise an important barrier against airborne pathogens. Cyclooxygenase (COX)-derived prostaglandins, such as PGE(2), are considered to be important regulators of lung function. Herein, we tested the hypothesis that pneumococci induced COX-2-dependent PGE(2) production in pulmonary epithelial cells. Pneumococci-infected human pulmonary epithelial BEAS-2B cells released PGE(2). Expression of COX-2 but not COX-1 was dose and time dependently increased in S. pneumoniae-infected BEAS-2B cells as well as in lungs of mice with pneumococcal pneumonia. S. pneumoniae induced degradation of IkappaBalpha and DNA binding of NF-kappaB. A specific peptide inhibitor of the IkappaBalpha kinase complex blocked pneumococci-induced PGE(2) release and COX-2 expression. In addition, we noted activation of p38 MAPK and JNK in pneumococci-infected BEAS-2B cells. PGE(2) release and COX-2 expression were reduced by p38 MAPK inhibitor SB-202190 but not by JNK inhibitor SP-600125. We analyzed interaction of kinase pathways and NF-kappaB activation: dominant-negative mutants of p38 MAPK isoforms alpha, beta(2), gamma, and delta blocked S. pneumoniae-induced NF-kappaB activation. In addition, recruitment of NF-kappaB subunit p65/RelA and RNA polymerase II to the cox2 promoter depended on p38 MAPK but not on JNK activity. In summary, p38 MAPK- and NF-kappaB-controlled COX-2 expression and subsequent PGE(2) release by lung epithelial cells may contribute significantly to the host response in pneumococcal pneumonia.


Asunto(s)
Ciclooxigenasa 2/genética , Pulmón/citología , FN-kappa B/metabolismo , Mucosa Respiratoria/enzimología , Streptococcus pneumoniae/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Secuencia de Bases , Línea Celular , Cartilla de ADN , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Imidazoles/farmacología , Plásmidos , Infecciones Neumocócicas/fisiopatología , Piridinas/farmacología , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/genética
18.
Am J Physiol Lung Cell Mol Physiol ; 290(5): L818-26, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16399788

RESUMEN

Moraxella catarrhalis is a major cause of infectious exacerbations of chronic obstructive lung disease (COPD) and may also contribute to the pathogenesis of COPD. Little is known about M. catarrhalis-bronchial epithelium interaction. We investigated activation of M. catarrhalis infected bronchial epithelial cells and characterized the signal transduction pathways. Moreover, we tested the hypothesis that the M. catarrhalis-induced cytokine expression is regulated by acetylation of histone residues and controlled by histone deacetylase activity (HDAC). We demonstrated that M. catarrhalis induced a strong time- and dose-dependent inflammatory response in the bronchial epithelial cell line (BEAS-2B), characterized by the release of IL-8 and GM-CSF. For this cytokine liberation activation of the ERK and p38 mitogen-activated protein (MAP) kinases and transcription factor NF-kappaB was required. Furthermore, M. catarrhalis-infected bronchial epithelial cells showed an enhanced acetylation of histone H3 and H4 globally and at the promoter of the il8 gene. Preventing histone deacetylation by the histone deacetylase inhibitor trichostatin A augmented the M. catarrhalis-induced IL-8 response. After exposure to M. catarrhalis, we found a decrease in global histone deacetylase expression and activity. Our findings suggest that M. catarrhalis-induced activation of il8 gene transcription was caused by interference with epigenetic mechanisms regulating il8 gene accessibility. Our findings provide insight into important molecular and cellular mechanisms of M. catarrhalis-induced activation of human bronchial epithelium.


Asunto(s)
Bronquios/microbiología , Bronquios/fisiopatología , Histona Desacetilasas/metabolismo , Inflamación/microbiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Moraxella catarrhalis/fisiología , FN-kappa B/metabolismo , Mucosa Respiratoria/fisiopatología , Línea Celular , Inhibidores Enzimáticos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Inhibidores de Histona Desacetilasas , Humanos , Interleucina-8/fisiología
19.
Thromb Haemost ; 94(2): 295-303, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16113818

RESUMEN

Streptococcus pneumoniae is the major pathogen of community-acquired pneumonia and a common cause of otitis, meningitis and sepsis. During pneumococci infection accompanied with bacterial invasion and hematogenous spreading, the endothelium is directly targeted by pneumococci and their virulence factors. Therefore, we tested the hypothesis that pneumococci induced endothelial apoptosis. Unencapsulated R6x pneumococci strongly induced apoptosis of human endothelial cells both from lung microvasculature and umbilical vein, whereas an encapsulated strain D39 mainly led to necrotic cell death. Deletion of the gene coding for pneumolysin reduced pneumococci-induced apoptosis in HUVEC. Furthermore, N-acetyl-L-cysteine, an antioxidant thiol, significantly reduced apoptosis caused by R6x, and LDH release induced by D39, pointing to a role for reactive oxygen species in the pathogenesis. Apoptotic cells showed increased cleavage and activity of caspases 6 and 9 but only late activation of caspase 3. Programmed cell death could be strongly reduced by pan-caspase inhibitor zVAD. Reduced levels of Bcl2 and cytosolic increase of apoptosis-inducing factor in pneumococci-infected cells implicated involvement of mitochondrial death pathways. Caspase activation and apoptosis were abolished by cAMP elevation. Moreover, p38 mitogen-activated protein kinase and c-Jun NH(2)-terminal kinase were activated in pneumococci-infected cells and inhibitors of both kinases strongly reduced pneumococci-induced caspase activation and apoptosis. Hence, kinase- and caspase-dependence of pneumococci-induced endothelial apoptosis may bear relevance to novel therapeutic approaches to pneumococci-related disease.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/microbiología , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Acetilcisteína/farmacología , Anexina A5/farmacología , Antioxidantes/farmacología , Proteínas Bacterianas/genética , Western Blotting , Caspasa 6 , Caspasa 9 , Muerte Celular , Células Cultivadas , AMP Cíclico/metabolismo , Citosol/metabolismo , Fragmentación del ADN , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Eliminación de Gen , Humanos , Peróxido de Hidrógeno/farmacología , L-Lactato Deshidrogenasa/metabolismo , Pulmón/irrigación sanguínea , Pulmón/microbiología , Microscopía Fluorescente , Mitocondrias/metabolismo , Necrosis , Fosforilación , Propidio/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno , Estreptolisinas/genética , Compuestos de Sulfhidrilo/química , Factores de Tiempo , Venas Umbilicales/citología , Venas Umbilicales/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA