Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Lung Cancer ; 125: 164-173, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30429016

RESUMO

INTRODUCTION: Local ablative treatment (LAT) improves outcome in lung cancer with oligometastatic disease (OMD) and potentially leads to long term survival. The aim of this retrospective study was to evaluate and quantify the additional benefit of LAT in synchronous OMD and to further identify prognostic factors for survival. PATIENTS AND METHODS: A propensity score-matched pairs analysis was performed on a set of patient and disease variables in 180 patients, treated for synchronous single organ OMD including non small-cell and neuroendocrine lung cancer with ≤4 metastases between 2000 and 2016 in 3 lung cancer centers in Berlin, Germany. Patients either received LAT for all sites of disease (intervention group) by means of surgery or stereotactic radiotherapy, or standard chemotherapy, if necessary combined with a local treatment with palliative intent (control group). RESULTS: Median follow-up time was 32.2 and 18.8 months for the intervention and control group, respectively. Substantial benefits in median progression-free survival (PFS, 25.1 vs. 8.2 months; HR, 0.30; 95% CI, 0.21-0.43; p < 0.001) and overall survival (OS, 60.4 vs. 22.5 months; HR, 0.42; 95% CI, 0.28-0.62; p < 0.001) were associated with LAT. Histology of adenocarcinoma and T1a primaries also predicted a favorable prognosis concerning PFS and OS. More favorable nodal stage (N0-2 vs. 3) and solitary metastases were associated with an extended PFS, whereas initial ECOG-PS (0-1 vs. 2) predicted OS. CONCLUSIONS: LAT was the strongest predictor for PFS and OS in OMD with ≤4 metastases. Survival in the control group identifies OMD as a subset of lung cancer with a generally more favorable prognosis.


Assuntos
Neoplasias Pulmonares/patologia , Adenocarcinoma/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Intervalo Livre de Doença , Feminino , Alemanha , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Pontuação de Propensão , Estudos Retrospectivos
2.
Proc Natl Acad Sci U S A ; 109(5): 1518-23, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22233811

RESUMO

The metabolic state of a cell is a key determinant in the decision to live and proliferate or to die. Consequently, balanced energy metabolism and the regulation of apoptosis are critical for the development and maintenance of differentiated organisms. Hypoxia occurs physiologically during development or exercise and pathologically in vascular disease, tumorigenesis, and inflammation, interfering with homeostatic metabolism. Here, we show that the hypoxia-inducible factor (HIF)-1-regulated glycolytic enzyme hexokinase II (HKII) acts as a molecular switch that determines cellular fate by regulating both cytoprotection and induction of apoptosis based on the metabolic state. We provide evidence for a direct molecular interactor of HKII and show that, together with phosphoprotein enriched in astrocytes (PEA15), HKII inhibits apoptosis after hypoxia. In contrast, HKII accelerates apoptosis in the absence of PEA15 and under glucose deprivation. HKII both protects cells from death during hypoxia and functions as a sensor of glucose availability during normoxia, inducing apoptosis in response to glucose depletion. Thus, HKII-mediated apoptosis may represent an evolutionarily conserved altruistic mechanism to eliminate cells during metabolic stress to the advantage of a multicellular organism.


Assuntos
Astrócitos/metabolismo , Linhagem da Célula , Hexoquinase/metabolismo , Mitocôndrias/enzimologia , Fosfoproteínas/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Catálise , Ratos
3.
J Immunol ; 184(6): 3072-8, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20154210

RESUMO

Chlamydophila pneumoniae infection of the vascular wall as well as activation of the transcription factor IFN regulatory factor (IRF)3 have been linked to development of chronic vascular lesions and atherosclerosis. The innate immune system detects invading pathogens by use of pattern recognition receptors, some of which are able to stimulate IRF3/7 activation and subsequent type I IFN production (e. g., IFN-beta). In this study, we show that infection of human endothelial cells with C. pneumoniae-induced production of IFN-beta, a cytokine that so far has been mainly associated with antiviral immunity. Moreover, C. pneumoniae infection led to IRF3 and IRF7 nuclear translocation in HUVECs and RNA interference experiments showed that IRF3 and IRF7 as well as the mitochondrial antiviral signaling (MAVS) were essential for IFN-beta induction. Finally, C. pneumoniae replication was enhanced in endothelial cells in which IRF3, IRF7, or MAVS expression was inhibited by small interfering RNA and attenuated by IFN-beta treatment. In conclusion, C. pneumoniae infection of endothelial cells activates an MAVS-, IRF3-, and IRF7-dependent signaling, which controls bacterial growth and might modulate development of vascular lesions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Chlamydophila pneumoniae/crescimento & desenvolvimento , Chlamydophila pneumoniae/imunologia , Endotélio Vascular/imunologia , Fator Regulador 3 de Interferon/fisiologia , Fator Regulador 7 de Interferon/fisiologia , Interferon beta/fisiologia , Proteínas Mitocondriais/fisiologia , Interferência de RNA/fisiologia , Células Cultivadas , Regulação para Baixo/imunologia , Endotélio Vascular/microbiologia , Endotélio Vascular/virologia , Humanos , Imunidade Inata , Interferon beta/biossíntese , Interferon beta/genética , Leucemia Experimental/imunologia , Leucemia Experimental/microbiologia , Leucemia Experimental/virologia , Vírus da Leucemia Murina de Moloney/imunologia , RNA Viral/antagonistas & inibidores , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/microbiologia , Infecções por Retroviridae/virologia , Transdução de Sinais/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/microbiologia , Infecções Tumorais por Vírus/virologia
4.
J Immunol ; 181(2): 940-7, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18606645

RESUMO

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.


Assuntos
Células Epiteliais/imunologia , Flagelina/metabolismo , Histonas/metabolismo , Interleucina-8/metabolismo , Legionella pneumophila/imunologia , NF-kappa B/metabolismo , Alvéolos Pulmonares/imunologia , Acetilação , Ácidos Anacárdicos/farmacologia , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Histona Acetiltransferases/metabolismo , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Interleucina-8/genética , Interleucina-8/imunologia , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/metabolismo , NF-kappa B/imunologia , Regiões Promotoras Genéticas , Inibidores da Síntese de Proteínas/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , RNA Polimerase II/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
J Biol Chem ; 281(47): 36173-9, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-16984921

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Epiteliais/citologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Legionella pneumophila/fisiologia , Pulmão/citologia , Pulmão/microbiologia , Transporte Ativo do Núcleo Celular , Diferenciação Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Células Epiteliais/metabolismo , Humanos , Legionella pneumophila/metabolismo , Pulmão/metabolismo , NF-kappa B/metabolismo , RNA/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Respir Res ; 7: 98, 2006 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-16834785

RESUMO

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.


Assuntos
Células Epiteliais/microbiologia , Interleucina-8/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pulmão/microbiologia , Streptococcus pneumoniae , Fator de Transcrição AP-1/metabolismo , Antracenos/farmacologia , Linhagem Celular , DNA/genética , DNA/metabolismo , Ativação Enzimática , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Humanos , Interleucina-8/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Pulmão/imunologia , Pulmão/metabolismo , Fosforilação , Regiões Promotoras Genéticas/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 290(5): L818-26, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16399788

RESUMO

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.


Assuntos
Brônquios/microbiologia , Brônquios/fisiopatologia , Histona Desacetilases/metabolismo , Inflamação/microbiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Moraxella catarrhalis/fisiologia , NF-kappa B/metabolismo , Mucosa Respiratória/fisiopatologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Inibidores de Histona Desacetilases , Humanos , Interleucina-8/fisiologia
8.
J Biol Chem ; 279(51): 53241-7, 2004 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-15485852

RESUMO

Streptococcus pneumoniae is the major cause of community-acquired pneumonia and one of the most common causes of death by infectious disease in industrialized countries. Little is known concerning the mechanisms of target cell activation in this disease. The present study shows that NF-kappaB and p38 MAPK signaling pathways contribute to chemokine synthesis by lung epithelial cells in response to pneumococci. In infected lungs of mice pneumococci stimulate expression of the interleukin (IL)-8 homolog keratinocyte-derived chemokine and granulocyte-macrophage colony-stimulating factor, as well as activate p38 MAPK. Human bronchial epithelium was chosen as a cellular model, because it establishes the first barrier against pathogens, and little is known about its function in innate immunity. Pneumococci infection induces expression of IL-8 and granulocyte-macrophage colony-stimulating factor as well as activation of p38 MAPK in human bronchial epithelial cells (BEAS-2B). Inhibition of p38 MAPK activity by SB202190 and SB203580 blocks pneumococci-induced cytokine release. In mouse lungs in vivo as well as in cultured cells, pneumococci activate NF-kappaBinanIkappaB kinase-dependent manner. Inhibition of p38 MAPK by chemical inhibitors or by RNA interference targeting p38alpha reduces pneumococci-induced NF-kappaB-dependent gene transcription. Blockade of p38 activity did not affect inducible nuclear translocation and recruitment of NF-kappaB/RelA to the IL-8 promotor but did reduce the level of phosphorylated RelA (serine 536) at IL-8 promotor and inhibited pneumococci-mediated recruitment of RNA polymerase II to IL-8 promotor. Thus, p38 MAPK contributes to pneumococci-induced chemokine transcription by modulating p65 NF-kappaB-mediated transactivation.


Assuntos
Interleucina-8/genética , Regiões Promotoras Genéticas , Streptococcus pneumoniae/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Brônquios/citologia , Brônquios/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Imunoprecipitação da Cromatina , Citocinas/biossíntese , Citocinas/metabolismo , Dimerização , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Imidazóis/farmacologia , Inflamação , Interleucina-8/metabolismo , Pulmão/microbiologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Pneumonia/microbiologia , Pneumonia/patologia , Piridinas/farmacologia , Interferência de RNA , RNA Polimerase II/química , RNA Complementar/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina/química , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA