Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 7(1): e30379, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22276187

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Chlamydophila pneumoniae/imunologia , Chlamydophila pneumoniae/fisiologia , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Monócitos/metabolismo , Monócitos/microbiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Reação em Cadeia da Polimerase , Interferência de RNA , Proteínas rac1 de Ligação ao GTP/genética
2.
J Immunol ; 185(1): 597-604, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20525885

RESUMO

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.


Assuntos
Regulação para Baixo/imunologia , Regulação da Expressão Gênica/imunologia , Mediadores da Inflamação/fisiologia , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/fisiologia , Subunidade p50 de NF-kappa B/antagonistas & inibidores , Proteína Adaptadora de Sinalização NOD2/fisiologia , Pneumonia Pneumocócica/imunologia , Receptor 2 Toll-Like/fisiologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Regulação para Baixo/genética , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/prevenção & controle , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Pneumonia Pneumocócica/genética , Pneumonia Pneumocócica/prevenção & controle , Streptococcus pneumoniae/imunologia , Receptor 2 Toll-Like/biossíntese , Receptor 2 Toll-Like/genética
3.
Am J Respir Crit Care Med ; 181(12): 1294-309, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20167850

RESUMO

Diseases of the respiratory tract are among the leading causes of death in the world population. Increasing evidence points to a key role of the innate immune system with its pattern recognition receptors (PRRs) in both infectious and noninfectious lung diseases, which include pneumonia, chronic obstructive pulmonary disease, acute lung injury, pneumoconioses, and asthma. PRRs are capable of sensing different microbes as well as endogenous molecules that are released after cell damage. This PRR engagement is the prerequisite for the initiation of immune responses to infections and tissue injuries which can be beneficial or detrimental to the host. PRRs include the Toll-like receptors, NOD-like receptors, RIG-I-like receptors, and cytosolic DNA sensors. The PRRs and their signaling pathways represent promising targets for prophylactic and therapeutic interventions in various lung diseases.


Assuntos
Imunidade Inata/imunologia , Pneumopatias/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Infecções Respiratórias/imunologia , Lesão Pulmonar Aguda/imunologia , Animais , Asma/imunologia , Humanos , Pneumopatias/microbiologia , Pneumopatias/virologia , Camundongos , Doença Pulmonar Obstrutiva Crônica/imunologia , Infecções Respiratórias/microbiologia , Infecções Respiratórias/virologia , Receptores Toll-Like/imunologia
4.
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
5.
Front Microbiol ; 1: 149, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21607087

RESUMO

Listeria monocytogenes is an intracellular, Gram-positive bacterium that can cause life-threatening illness especially in immunocompromised individuals and newborns. The pathogen propagates within the cytosol of various host cells after escaping from the phagosomal compartment depending on the cytolysin listeriolysin O. While L. monocytogenes can manipulate the endocytic and many host-cell signaling cascades to its advantage, host cells are however capable of detecting Listeria infection at different cellular compartments by expressing innate immune receptors that trigger antibacterial defense pathways. These receptors include the Toll-like receptors, NOD-like receptors (NLRs), and cytosolic DNA sensors. Some NLRs as well as the DNA sensor AIM2 form multiprotein complexes called inflammasomes. Inflammasomes regulate caspase-1-dependent production of the key inflammatory cytokines IL-1ß and IL-18 as well as pyroptotic cell death in L. monocytogenes-infected cells. This review describes the current knowledge about innate immune sensing and inflammasome activation in Listeria infection.

6.
J Immunol ; 184(2): 922-30, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20008285

RESUMO

Different NOD-like receptors, including NLRP1, NLRP3, and NLRC4, as well as the recently identified HIN-200 protein, AIM2, form multiprotein complexes called inflammasomes, which mediate caspase-1-dependent processing of pro-IL-1beta. Listeria monocytogenes is an intracellular pathogen that is actively phagocytosed by monocytes/macrophages and subsequently escapes from the phagosome into the host cell cytosol, depending on its pore-forming toxin listeriolysin O (LLO). In this study, we demonstrate that human PBMCs produced mature IL-1beta when infected with wild-type L. monocytogenes or when treated with purified LLO. L. monocytogenes mutants lacking LLO or expressing a noncytolytic LLO as well as the avirulent Listeria innocua induced strongly impaired IL-1beta production. RNA interference and inhibitor experiments in human PBMCs as well as experiments in Nlrp3 and Rip2 knockout bone marrow-derived macrophages demonstrated that the Listeria-induced IL-1beta release was dependent on ASC, caspase-1, and NLRP3, whereas NOD2, Rip2, NLRP1, NLRP6, NLRP12, NLRC4, and AIM2 appeared to be dispensable. We found that L. monocytogenes-induced IL-1beta production was largely dependent on phagosomal acidification and cathepsin B release, whereas purified LLO activated an IL-1beta production independently of these mechanisms. Our results indicate that L. monocytogenes-infected human PBMCs produced IL-1beta, largely depending on an LLO-mediated phagosomal rupture and cathepsin B release, which is sensed by Nlrp3. In addition, an LLO-dependent but cathepsin B-independent NLRP3 activation might contribute to some extent to the IL-1beta production in L. monocytogenes-infected cells.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Interleucina-1beta/biossíntese , Leucócitos Mononucleares/microbiologia , Listeria monocytogenes/imunologia , Animais , Toxinas Bacterianas , Catepsina B/metabolismo , Linhagem Celular , Humanos , Leucócitos Mononucleares/metabolismo , Listeria monocytogenes/patogenicidade , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fagossomos
7.
Thromb Haemost ; 102(6): 1103-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19967140

RESUMO

Bacteraemia and viraemia are characterised by pathogens entering the bloodstream. Endothelial cells are among the first cells coming into contact with the microbes and also some endogenous molecules which are released by tissue damage. As part of the innate immune system, endothelial cells respond to these contacts by producing inflammatory mediators and expressing surface molecules. The initial sensing of microbial and endogenous danger-associated molecules is mediated by so-called pattern recognition receptors (PRRs). PRRs can be classified in different protein families such as the Toll-like receptors, the NOD-like receptors and the RIG-I-like receptors. By activating inflammatory gene transcription and posttranslational processing, PRRs control the immediate innate immune reaction and also the subsequent adaptive immune response. Here we describe the current knowledge of extra- and intracellular PRRs in endothelial cells and their potential role in sepsis and vascular diseases.


Assuntos
RNA Helicases DEAD-box/fisiologia , Células Endoteliais/imunologia , Proteínas Adaptadoras de Sinalização NOD/fisiologia , Sepse/imunologia , Receptores Toll-Like/fisiologia , Imunidade Adaptativa , Bacteriemia/imunologia , Humanos , Imunidade Inata , Modelos Imunológicos , Viremia/imunologia
8.
Cell Microbiol ; 11(12): 1782-801, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19681907

RESUMO

The outer membrane proteins YadA and invasin of Yersinia pseudotuberculosis promote invasion into mammalian cells through beta(1)-integrins and trigger the production of interleukin (IL)-8. FAK, c-Src and the PI3 kinase were previously found to be important for both YadA- and invasin-promoted uptake. Here, we demonstrate that two different downstream effectors of PI3 kinase, Akt and phospholipase Cgamma1 are required for efficient cell invasion. Inhibition of Akt or phospholipase C-gamma (PLC-gamma)1 by pharmaceutical agents as well as reduced expression of the isoforms Akt1 and Akt2, and of PLC-gamma1 by RNA interference decreased entry of YadA- and Inv-expressing bacteria significantly. In addition, we report that the conventional protein kinases C (PKC)alpha and -beta, positioned downstream of PLC-gamma1, are activated upon Inv- or YadA-promoted cell entry. They colocalize with intracellular bacteria and their depletion by siRNA treatment also resulted in a strong reduction of cell entry. In contrast, neither Akt nor PLC-gamma1, and the PKCs are essential for YadA- and Inv-mediated IL-8 synthesis and release. We conclude that YadA and invasin of Y. pseudotuberculosis both trigger similar signal transduction pathways during integrin-mediated phagocytosis into epithelial cells, which lead to the activation of Akt, PLC-gamma1, PKCalpha and -beta downstream of PI3 kinase, separate from the MAPK-dependent pathway that triggers IL-8 production.


Assuntos
Adesinas Bacterianas/metabolismo , Proteína Oncogênica v-akt/metabolismo , Fosfolipase C gama/metabolismo , Proteína Quinase C/metabolismo , Infecções por Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/metabolismo , Linhagem Celular , Inibidores Enzimáticos/administração & dosagem , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Interleucina-8/biossíntese , Proteína Oncogênica v-akt/antagonistas & inibidores , Proteína Oncogênica v-akt/genética , Fagocitose , Fosfolipase C gama/antagonistas & inibidores , Fosfolipase C gama/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Yersinia pseudotuberculosis/genética , Infecções por Yersinia pseudotuberculosis/microbiologia
9.
Arterioscler Thromb Vasc Biol ; 29(3): 380-6, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19122173

RESUMO

OBJECTIVE: Activation of the endothelium by oxidized low-density lipoprotein (oxLDL) has been implicated in the development of atherosclerosis. Histone modifications impact on the transcriptional activity state of genes. We tested the hypothesis that oxLDL-induced inflammatory gene expression is regulated by histone modifications and experienced the effect of statins on these alterations. METHODS AND RESULTS: OxLDL-related interleukin-8 (IL-8) and monocyte-chemoattractant protein-1 (MCP-1) secretion in endothelial cells was reduced by statins but enhanced by histone deacetylase inhibitors. OxLDL induced lectin-like oxidized LDL receptor-1 (LOX-1) and extracellular regulated kinases (ERK1/2)-dependent acetylation of histone H3 and H4 as well as phosphorylation of histone H3, both globally and on the promoters of il8 and mcp1. Pretreatment of oxLDL-exposed cells with statins reduced the above mentioned histone modification, as well as recruitment of CREB binding protein (CBP) 300, NF-kappaB, and of RNA polymerase II but prevented loss of binding of histone deacetylase (HDAC)-1 and -2 at the il8 and mcp1 gene promoters. OxLDL reduced HDAC1 and 2 expression, and statins partly restored global HDAC-activity. Statin-related effects were reverted with mevalonate. In situ experiments indicated decreased expression of HDAC2 in endothelial cells in atherosclerotic plaques of human coronary arteries. CONCLUSIONS: Histone modifications seem to play an important role in atherosclerosis.


Assuntos
Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Histonas/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Mediadores da Inflamação/metabolismo , Lipoproteínas LDL/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Vasos Coronários/enzimologia , Citocinas/genética , Células Endoteliais/enzimologia , Células Endoteliais/imunologia , Ácidos Graxos Monoinsaturados/farmacologia , Fluvastatina , Regulação da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 1 , Histona Desacetilase 2 , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , Interleucina-8/metabolismo , Ácido Mevalônico/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Receptores Depuradores Classe E/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , Vorinostat
10.
Nat Immunol ; 9(11): 1270-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18836450

RESUMO

Although Moraxella catarrhalis and Neisseria meningitidis are important human pathogens, they often colonize the human respiratory tract without causing overt clinical symptoms. Both pathogens express structurally unrelated proteins that share the ability to stimulate the adhesion molecule CEACAM1 expressed on human cells. Here we demonstrate that the interaction of CEACAM1 with ubiquitous surface protein A1 expressed on M. catarrhalis or with opacity-associated proteins on N. meningitidis resulted in reduced Toll-like receptor 2-initiated transcription factor NF-kappaB-dependent inflammatory responses of primary pulmonary epithelial cells. These inhibitory effects were mediated by tyrosine phosphorylation of the immunoreceptor tyrosine-based inhibitory motif of CEACAM1 and by recruitment of the phosphatase SHP-1, which negatively regulated Toll-like receptor 2-dependent activation of the phosphatidylinositol 3-OH kinase-Akt kinase pathway. Our results identify a CEACAM1-dependent immune-evasion strategy.


Assuntos
Antígenos CD/imunologia , Brônquios/imunologia , Moléculas de Adesão Celular/imunologia , Moraxella catarrhalis/imunologia , Neisseria meningitidis/imunologia , Mucosa Respiratória/imunologia , Receptor 2 Toll-Like/imunologia , Motivos de Aminoácidos/fisiologia , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Antígenos CD/química , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Brônquios/metabolismo , Brônquios/microbiologia , Moléculas de Adesão Celular/química , Células Cultivadas , Citocinas/metabolismo , Regulação para Baixo , Humanos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/metabolismo
11.
Cell Microbiol ; 10(12): 2579-88, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18771559

RESUMO

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.


Assuntos
DNA Bacteriano/imunologia , Proteínas de Ligação a DNA/metabolismo , Interferon beta/biossíntese , Legionella pneumophila/imunologia , Animais , Linhagem Celular , Humanos , Interleucina-8/biossíntese , Camundongos , Proteínas de Ligação a RNA
12.
J Immunol ; 181(4): 2664-71, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18684957

RESUMO

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.


Assuntos
Regulação para Baixo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteína Adaptadora de Sinalização NOD2/antagonistas & inibidores , Proteína Adaptadora de Sinalização NOD2/metabolismo , Proteínas rac1 de Ligação ao GTP/fisiologia , Linhagem Celular , Células Cultivadas , Regulação para Baixo/imunologia , Ativação Enzimática/imunologia , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Proteína Adaptadora de Sinalização NOD2/agonistas , Proteína Adaptadora de Sinalização NOD2/fisiologia , Transporte Proteico/imunologia , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais/imunologia , Proteínas rac1 de Ligação ao GTP/deficiência , Proteínas rac1 de Ligação ao GTP/genética
13.
J Immunol ; 180(10): 6808-15, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18453601

RESUMO

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.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Células Epiteliais/microbiologia , Legionella pneumophila/crescimento & desenvolvimento , Doença dos Legionários/imunologia , Macrófagos/microbiologia , Proteína Inibidora de Apoptose Neuronal/metabolismo , Western Blotting , Linhagem Celular , Proliferação de Células , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Flagelina/genética , Flagelina/metabolismo , Humanos , Imunidade Inata , Macrófagos/imunologia , Macrófagos/metabolismo , Microscopia Confocal , Reação em Cadeia da Polimerase , Interferência de RNA , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia
14.
Histochem Cell Biol ; 130(1): 165-76, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18365236

RESUMO

Streptococcus pneumoniae is the leading causative agent of community-acquired pneumonia. Induction of apoptosis in pulmonary epithelial cells by bacteria during pneumonia might be harmful to the host. Interleukin-15 (IL-15) has been demonstrated as an effective inhibitor of apoptosis and is expressed in lung epithelium on the mRNA and protein level. Therefore, we characterized the sub-cellular expression pattern of the short and long IL-15 isoforms in lung epithelial cells in vitro as well as its role in pneumococci-related lung epithelial cell apoptosis. We found an expression pattern for both IL-15 signal peptides in the pulmonary epithelial cell lines A549 and Beas-2B. Moreover, a strong co-localization of IL-15 and IL-15Ralpha was detected on cell surfaces. Compared to pro-inflammatory cytokine stimulation, neither IL-15 nor its trimeric receptor complex was up-regulated after pneumococcal infection. However, overexpression of IL-15 isoforms revealed IL-15LSP and IL-15Vkl as inhibitors of pneumococci induced apoptosis in pulmonary epithelial cells. Thus, IL-15 may act as an anti-apoptotic molecule in pneumococci infection, thereby suggesting IL-15 as a benefical cytokine in pulmonary host defense against infection.


Assuntos
Interleucina-15/metabolismo , Pulmão/imunologia , Pulmão/patologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/patologia , Receptores de Interleucina-15/metabolismo , Streptococcus pneumoniae/imunologia , Apoptose , Linhagem Celular Tumoral , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Humanos , Pulmão/microbiologia , Isoformas de Proteínas/metabolismo , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Transfecção
15.
Circ Res ; 102(8): 888-95, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18309103

RESUMO

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.


Assuntos
Chlamydophila pneumoniae/fisiologia , Histonas/metabolismo , Sinvastatina/farmacologia , Células Cultivadas , Chlamydophila pneumoniae/efeitos dos fármacos , Citocinas/biossíntese , Citocinas/metabolismo , Células Endoteliais/microbiologia , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas rac1 de Ligação ao GTP/metabolismo
16.
Thromb Haemost ; 98(2): 319-26, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17721613

RESUMO

The innate immune system represents the principal sensor of infections in multicellular organisms and might also mediate responses to some endogenous molecules. In this context, endothelial cells are among the first cells coming into contact with microbial or endogenous (danger-associated) molecules or whole pathogens entering the bloodstream. Since many bacteria and viruses invade the endothelium, endothelial cells are equipped with both extracellular and cytosolic surveillance systems capable of sensing microbial components, and endogenous danger-associated molecules. The receptor molecules, called pattern recognition receptors (PRRs), are classified as transmembrane or cytosolic molecules. While the transmembrane PRRs recognize extracellular and membrane-enclosed foreign organisms, the cytosolic PRRs appear to sense intracellular infections. Here we focus on both PRR classes in general, and outline the current knowledge of extra- and intracellular pattern recognition in endothelial cells and its potential role in vascular diseases and sepsis.


Assuntos
Células Endoteliais/imunologia , Imunidade Inata , Proteínas Adaptadoras de Sinalização CARD/imunologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Humanos , Receptores de Reconhecimento de Padrão/imunologia , Sepse/imunologia , Receptores Toll-Like/imunologia
17.
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
18.
Biochem Pharmacol ; 72(11): 1367-74, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-16884694

RESUMO

Infection of endothelial cells by Listeria monocytogenes is an essential step in the pathogenesis of listeriosis. Small GTPases of the Rho family act as molecular switches in signal transduction. We tested the hypothesis that Rho GTPases contribute to the regulation of cytokine expression following L. monocytogenes infection. L. monocytogenes induced release of distinct CC and CXC, as well as Th1 and Th2 cytokines and growth factors by endothelial cells and activated RhoA and Rac1. Inhibition of Rac1 by inhibitor Nsc23766 reduced cytokine expression, and slightly yet significantly the uptake of bacteria. Blocking of Rho proteins by Clostridium difficile toxin B-10463 (TcdB) reduced Listeria-dependent cytokine expression, whereas activating Rho proteins by Escherichia coli CNF1 increased it. We analyzed regulation of IL-8 expression in more detail: Listeria-induced IL-8 release was reduced by inhibition of RhoA, Rac1 and Cdc42 (TcdB) or Rac1 while blocking of RhoA/B/C by Clostridium limosum C3 fusion toxin (C3FT) or Rho kinase by Y27632 reduced cytokine expression only slightly. Activation of RhoA, Rac1 and Cdc42 (CNF1), but not of RhoA alone (CNF(Y)), enhanced Listeria-dependent IL-8 release significantly. Furthermore, inhibition of RhoA, Rac1 and Cdc42 (TcdB) and Rac1 (Nsc23766), but not of RhoA (C3FT) reduced Listeria-related recruitment of NF-kappaB/p65 and RNA polymerase II to the il8 promoter, as well as acetylation of histone H4 and Ser10/Lys14-phosphorylation/acetylation of histone H3 at the il8 gene promoter in HUVEC. In conclusion, Rac1 contributed to L. monocytogenes-induced cytokine expression by human endothelial cells.


Assuntos
Endotélio Vascular/enzimologia , Listeria monocytogenes/patogenicidade , Transdução de Sinais , Veias Umbilicais/enzimologia , Proteínas rac1 de Ligação ao GTP/biossíntese , Aminoquinolinas/farmacologia , Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Células Cultivadas , Citocinas/biossíntese , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/microbiologia , Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/farmacologia , Humanos , Pirimidinas/farmacologia , Veias Umbilicais/efeitos dos fármacos , Veias Umbilicais/microbiologia
19.
Cell Microbiol ; 7(1): 63-77, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15617524

RESUMO

The YadA protein of Yersinia pseudotuberculosis promotes tight adhesion and invasion into mammalian cells through beta(1)-integrins. In this work, we demonstrate that YadA also triggers the production of interleukin-8 (IL-8) in host cells and we identify intracellular signal transduction mechanisms involved in YadA-initiated cell invasion and/or IL-8 synthesis. Tyrosine protein kinases, including the focal adhesion kinase (FAK) and c-Src, as well as the small GTPase Ras, were shown to play a significant role in both YadA-promoted cell processes. YadA-mediated cell contact led to autophosphorylation of FAK at position Tyr397 and induced GTP-loading of Ras. Furthermore, IL-8 production and invasion induced by YadA were strongly reduced in FAK- and c-Src-deficient cells and in cells overexpressing dominant interfering forms of FAK, c-Src or Ras. We also demonstrate that YadA activates the Ras-dependent Raf-MEK1/2-ERK1/2 pathway and mitogen-activated protein kinases (MAPKs) p38 and JNK. Moreover, inhibition of ERK1/2 by pharmacological agents or overexpression of dominant negative FAK, c-Src or Ras abrogated IL-8 release, whereas invasion remained unaffected. In contrast, actin polymerization and phosphatidylinositol 3-kinase (PI3K) activity is essential for YadA-promoted cell entry, but not for cytokine secretion. We conclude that YadA triggers FAK-Src complex formation and subsequent Ras activation, which leads to the stimulation of MAPKs-dependent IL-8 production or to PI3K-dependent invasion.


Assuntos
Adesinas Bacterianas/fisiologia , Interleucina-8/biossíntese , Transdução de Sinais , Yersinia pseudotuberculosis/patogenicidade , Animais , Proteína Tirosina Quinase CSK , Linhagem Celular , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/análise , MAP Quinase Quinase 1/metabolismo , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/análise , Quinases raf/metabolismo , Proteínas ras/genética , Proteínas ras/fisiologia , Quinases da Família src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...