Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36835297

RESUMO

Legionella pneumophila is an intracellular pathogen that can cause severe pneumonia after the inhalation of contaminated aerosols and replication in alveolar macrophages. Several pattern recognition receptors (PRRs) have been identified that contribute to the recognition of L. pneumophila by the innate immune system. However, the function of the C-type lectin receptors (CLRs), which are mainly expressed by macrophages and other myeloid cells, remains largely unexplored. Here, we used a library of CLR-Fc fusion proteins to search for CLRs that can bind the bacterium and identified the specific binding of CLEC12A to L. pneumophila. Subsequent infection experiments in human and murine macrophages, however, did not provide evidence for a substantial role of CLEC12A in controlling innate immune responses to the bacterium. Consistently, antibacterial and inflammatory responses to Legionella lung infection were not significantly influenced by CLEC12A deficiency. Collectively, CLEC12A is able to bind to L. pneumophila-derived ligands but does not appear to play a major role in the innate defense against L. pneumophila.


Assuntos
Interações Hospedeiro-Patógeno , Imunidade Inata , Lectinas Tipo C , Legionella pneumophila , Doença dos Legionários , Receptores Mitogênicos , Animais , Humanos , Camundongos , Lectinas Tipo C/metabolismo , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Doença dos Legionários/microbiologia , Macrófagos/metabolismo , Macrófagos Alveolares/metabolismo , Receptores Mitogênicos/imunologia
2.
Int J Med Microbiol ; 308(1): 161-167, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29097162

RESUMO

Legionella pneumophila is a facultative intracellular bacterium which can cause a severe pneumonia called Legionnaires' disease after inhalation of contaminated water droplets and replication in alveolar macrophages. The innate immune system is generally able to sense and -in most cases- control L. pneumophila infection. Comorbidities and genetic risk factors, however, can compromise the immune system and high infection doses might overwhelm its capacity, thereby enabling L. pneumophila to grow and disseminate inside the lung. The innate immune system mediates sensing of L. pneumophila by employing e.g. NOD-like receptors (NLRs), Toll-like receptors (TLRs), as well as the cGAS/STING pathway to stimulate death of infected macrophages as well as production of proinflammatory cytokines and interferons (IFNs). Control of pulmonary L. pneumophila infection is largely mediated by inflammasome-, TNFα- and IFN-dependent macrophage-intrinsic resistance mechanisms. This article summarizes the current knowledge of innate immune responses to L. pneumophila infection in general, and of macrophage-intrinsic defense mechanisms in particular.


Assuntos
Imunidade Inata , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Macrófagos Alveolares/imunologia , Transdução de Sinais/imunologia , Citocinas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamassomos/metabolismo , Doença dos Legionários/microbiologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia
3.
Sci Rep ; 6: 30943, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27476670

RESUMO

Bacterial pneumonia is a major cause of acute lung injury and acute respiratory distress syndrome, characterized by alveolar barrier disruption. NLRP3 is best known for its ability to form inflammasomes and to regulate IL-1ß and IL-18 production in myeloid cells. Here we show that NLRP3 protects the integrity of the alveolar barrier in a mouse model of Streptococcus pneumoniae-induced pneumonia, and ex vivo upon treatment of isolated perfused and ventilated lungs with the purified bacterial toxin, pneumolysin. We reveal that the preserving effect of NLRP3 on the lung barrier is independent of inflammasomes, IL-1ß and IL-18. NLRP3 improves the integrity of alveolar epithelial cell monolayers by enhancing cellular adherence. Collectively, our study uncovers a novel function of NLRP3 by demonstrating that it protects epithelial barrier function independently of inflammasomes.


Assuntos
Células Epiteliais/imunologia , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Pneumonia Pneumocócica/prevenção & controle , Alvéolos Pulmonares/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Animais , Proteínas de Bactérias/metabolismo , Adesão Celular , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Feminino , Interleucina-18/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia , Transdução de Sinais , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/microbiologia
4.
Infect Immun ; 83(9): 3624-37, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26123804

RESUMO

Intracellular pathogens are differentially sensed by the compartmentalized host immune system. Nevertheless, gene expression studies of infected cells commonly average the immune responses, neglecting the precise pathogen localization. To overcome this limitation, we dissected the transcriptional immune response to Shigella flexneri across different infection stages in bulk and single cells. This identified six distinct transcriptional profiles characterizing the dynamic, multilayered host response in both bystander and infected cells. These profiles were regulated by external and internal danger signals, as well as whether bacteria were membrane bound or cytosolic. We found that bacterial internalization triggers a complex, effector-independent response in bystander cells, possibly to compensate for the undermined host gene expression in infected cells caused by bacterial effectors, particularly OspF. Single-cell analysis revealed an important bacterial strategy to subvert host responses in infected cells, demonstrating that OspF disrupts concomitant gene expression of proinflammatory, apoptosis, and stress pathways within cells. This study points to novel mechanisms through which bacterial internalization, localization, and injected effectors orchestrate immune response transcriptional signatures.


Assuntos
Disenteria Bacilar/imunologia , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Células HeLa , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Shigella flexneri/imunologia , Transfecção
5.
PLoS Pathog ; 8(2): e1002507, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22319448

RESUMO

Survival within macrophages is a central feature of Mycobacterium tuberculosis pathogenesis. Despite significant advances in identifying new immunological parameters associated with mycobacterial disease, some basic questions on the intracellular fate of the causative agent of human tuberculosis in antigen-presenting cells are still under debate. To get novel insights into this matter, we used a single-cell fluorescence resonance energy transfer (FRET)-based method to investigate the potential cytosolic access of M. tuberculosis and the resulting cellular consequences in an unbiased, quantitative way. Analysis of thousands of THP-1 macrophages infected with selected wild-type or mutant strains of the M. tuberculosis complex unambiguously showed that M. tuberculosis induced a change in the FRET signal after 3 to 4 days of infection, indicating phagolysosomal rupture and cytosolic access. These effects were not seen for the strains M. tuberculosisΔRD1 or BCG, both lacking the ESX-1 secreted protein ESAT-6, which reportedly shows membrane-lysing properties. Complementation of these strains with the ESX-1 secretion system of M. tuberculosis restored the ability to cause phagolysosomal rupture. In addition, control experiments with the fish pathogen Mycobacterium marinum showed phagolysosomal translocation only for ESX-1 intact strains, further validating our experimental approach. Most importantly, for M. tuberculosis as well as for M. marinum we observed that phagolysosomal rupture was followed by necrotic cell death of the infected macrophages, whereas ESX-1 deletion- or truncation-mutants that remained enclosed within phagolysosomal compartments did not induce such cytotoxicity. Hence, we provide a novel mechanism how ESX-1 competent, virulent M. tuberculosis and M. marinum strains induce host cell death and thereby escape innate host defenses and favor their spread to new cells. In this respect, our results also open new research directions in relation with the extracellular localization of M. tuberculosis inside necrotic lesions that can now be tackled from a completely new perspective.


Assuntos
Macrófagos/metabolismo , Infecções por Mycobacterium/patologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Fagossomos/patologia , Morte Celular , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas de Homeodomínio/metabolismo , Humanos , Evasão da Resposta Imune , Macrófagos/microbiologia , Infecções por Mycobacterium/imunologia , Infecções por Mycobacterium/metabolismo , Mycobacterium marinum/imunologia , Mycobacterium marinum/metabolismo , Mycobacterium marinum/patogenicidade , Mycobacterium tuberculosis/metabolismo , Fagossomos/metabolismo , Fagossomos/microbiologia , Salmonella typhimurium/patogenicidade , Shigella flexneri/patogenicidade
6.
Cell Microbiol ; 13(11): 1668-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21790939

RESUMO

Defence mechanisms against intracellular bacterial pathogens are incompletely understood. Our study characterizes a type I IFN-dependent cell-autonomous defence pathway directed against Legionella pneumophila, an intracellular model organism and frequent cause of pneumonia. We show that macrophages infected with L. pneumophila produced IFNß in a STING- and IRF3- dependent manner. Paracrine type I IFNs stimulated upregulation of IFN-stimulated genes and a cell-autonomous defence pathway acting on replicating and non-replicating Legionella within their specialized vacuole. Our infection experiments in mice lacking receptors for type I and/or II IFNs show that type I IFNs contribute to expression of IFN-stimulated genes and to bacterial clearance as well as resistance in L. pneumophila pneumonia in addition to type II IFN. Overall, our study shows that paracrine type I IFNs mediate defence against L. pneumophila, and demonstrates a protective role of type I IFNs in in vivo infections with intracellular bacteria.


Assuntos
Interferon Tipo I/imunologia , Legionella pneumophila/imunologia , Legionella pneumophila/patogenicidade , Doença dos Legionários/imunologia , Doença dos Legionários/patologia , Transdução de Sinais , Animais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Fator Regulador 3 de Interferon/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Camundongos , Vacúolos/imunologia , Vacúolos/microbiologia
7.
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
8.
J Virol ; 84(2): 1034-46, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19906927

RESUMO

Herpes simplex virus type 1 (HSV-1) is one of the most frequent and successful human pathogens. It targets immature dendritic cells (iDCs) to interfere with the antiviral immune response. The mechanisms underlying apoptosis of HSV-1-infected iDCs are not fully understood. Previously, we have shown that HSV-1-induced apoptosis of iDCs is associated with downregulation of the cellular FLICE-inhibitory protein (c-FLIP), a potent inhibitor of caspase-8-mediated apoptosis. In this study, we prove that HSV-1 induces degradation of c-FLIP in a proteasome-independent manner. In addition, by using c-FLIP-specific small interfering RNA (siRNA) we show for the first time that downregulation of c-FLIP expression is sufficient to drive uninfected iDCs into apoptosis, underlining the importance of this molecule for iDC survival. Surprisingly, we also observed virus-induced c-FLIP downregulation in epithelial cells and many other cell types that do not undergo apoptosis after HSV-1 infection. Microarray analyses revealed that HSV-1-encoded latency-associated transcript (LAT) sequences, which can substitute for c-FLIP as an inhibitor of caspase-8-mediated apoptosis, are much less abundant in iDCs as compared to epithelial cells. Finally, iDCs infected with an HSV-1 LAT knockout mutant showed increased apoptosis when compared to iDCs infected with the corresponding wild-type HSV-1. Taken together, our results demonstrate that apoptosis of HSV-1-infected iDCs requires both c-FLIP downregulation and diminished expression of viral LAT.


Assuntos
Apoptose/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Células Dendríticas , Regulação para Baixo , Herpesvirus Humano 1/patogenicidade , MicroRNAs/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Linhagem Celular , Células Dendríticas/citologia , Células Dendríticas/fisiologia , Células Dendríticas/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Humanos , Latência Viral
9.
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
10.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA