Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Shock ; 42(1): 60-4, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24667617

RESUMO

Innate immunity pathways are found to play an important role in ventilator-induced lung injury. We analyzed pulmonary expression of Toll-like receptor 2 (TLR2) in humans and mice and determined the role of TLR2 in the pathogenesis of ventilator-induced lung injury in mice. Toll-like receptor 2 gene expression was analyzed in human bronchoalveolar lavage fluid (BALF) cells and murine lung tissue after 5 h of ventilation. In addition, wild-type (WT) and TLR2 knockout (KO) mice were ventilated with either lower tidal volumes (VT) of 7 mL/kg with positive end-expiratory pressure (PEEP) or higher VT of 15 mL/kg without PEEP for 5 h. Spontaneously breathing mice served as controls. Total protein and immunoglobulin M levels in BALF, neutrophil influx into the alveolar compartment, and interleukin 6 (IL-6), IL-1ß, and keratinocyte-derived chemokine concentrations in lung tissue homogenates were measured. We observed enhanced TLR2 gene expression in BALF cells of ventilated patients and in lung tissue of ventilated mice. In WT mice, ventilation with higher VT without PEEP resulted in lung injury and inflammation with higher immunoglobulin M levels, neutrophil influx, and levels of inflammatory mediators compared with controls. In TLR2 KO mice, neutrophil influx and IL-6, IL-1ß, and keratinocyte-derived chemokine were enhanced by this ventilation strategy. Ventilation with lower VT with PEEP only increased neutrophil influx and was similar in WT and TLR2 KO mice. In summary, injurious ventilation enhances TLR2 expression in lungs. Toll-like receptor 2 deficiency does not protect lungs from ventilator-induced lung injury. In contrast, ventilation with higher VT without PEEP aggravates inflammation in TLR2 KO mice.


Assuntos
Respiração Artificial/métodos , Receptor 2 Toll-Like/deficiência , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/química , Dióxido de Carbono/sangue , Regulação da Expressão Gênica , Humanos , Pulmão/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/sangue , Pressão Parcial , Respiração com Pressão Positiva/efeitos adversos , RNA Mensageiro/genética , Respiração Artificial/efeitos adversos , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/fisiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/genética
2.
Intensive Care Med Exp ; 2(1): 22, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26215707

RESUMO

BACKGROUND: Mechanical ventilation (MV) can cause ventilator-induced lung injury (VILI). The innate immune response mediates this iatrogenic inflammatory condition. The receptor for advanced glycation end products (RAGE) is a multiligand receptor that can amplify immune and inflammatory responses. We hypothesized that RAGE signaling contributes to the pro-inflammatory state induced by MV. METHODS: RAGE expression was analyzed in lung brush and lavage cells obtained from ventilated patients and lung tissue of ventilated mice. Healthy wild-type (WT) and RAGE knockout (KO) mice were ventilated with relatively low (approximately 7.5 ml/kg) or high (approximately 15 ml/kg) tidal volume. Positive end-expiratory pressure was set at 2 cm H2O during both MV strategies. Also, WT and RAGE KO mice with lipopolysaccharide (LPS)-induced lung injury were ventilated with the above described ventilation strategies. In separate experiments, the contribution of soluble RAGE, a RAGE isoform that may function as a decoy receptor, in ventilated RAGE KO mice was investigated. Lung wet-to-dry ratio, cell and neutrophil influx, cytokine and chemokine concentrations, total protein levels, soluble RAGE, and high-mobility group box 1 (HMGB1) presence in lung lavage fluid were analyzed. RESULTS: MV was associated with increased RAGE mRNA levels in both human lung brush samples and lung tissue of healthy mice. In healthy high tidal volume-ventilated mice, RAGE deficiency limited inflammatory cell influx. Other VILI parameters were not affected. In our second set of experiments where we compared RAGE KO and WT mice in a 2-hit model, we observed higher pulmonary cytokine and chemokine levels in RAGE KO mice undergoing LPS/high tidal volume MV as compared to WT mice. Third, in WT mice undergoing the LPS/high tidal volume MV, we observed HMGB1 presence in lung lavage fluid. Moreover, MV increased levels of soluble RAGE in lung lavage fluid, with the highest levels found in LPS/high tidal volume-ventilated mice. Administration of soluble RAGE to LPS/high tidal volume-ventilated RAGE KO mice attenuated the production of inflammatory mediators. CONCLUSIONS: RAGE was not a crucial contributor to the pro-inflammatory state induced by MV. However, the presence of sRAGE limited the production of pro-inflammatory mediators in our 2-hit model of LPS and high tidal volume MV.

3.
PLoS One ; 8(7): e68694, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874727

RESUMO

BACKGROUND: Bacterial products add to mechanical ventilation in enhancing lung injury. The role of endogenous triggers of innate immunity herein is less well understood. S100A8/A9 proteins are released by phagocytes during inflammation. The present study investigates the role of S100A8/A9 proteins in ventilator-induced lung injury. METHODS: Pulmonary S100A8/A9 levels were measured in samples obtained from patients with and without lung injury. Furthermore, wild-type and S100A9 knock-out mice, naive and with lipopolysaccharide-induced injured lungs, were randomized to 5 hours of spontaneously breathing or mechanical ventilation with low or high tidal volume (VT). In addition, healthy spontaneously breathing and high VT ventilated mice received S100A8/A9, S100A8 or vehicle intratracheal. Furthermore, the role of Toll-like receptor 4 herein was investigated. RESULTS: S100A8/A9 protein levels were elevated in patients and mice with lung injury. S100A8/A9 levels synergistically increased upon the lipopolysaccharide/high VT MV double hit. Markers of alveolar barrier dysfunction, cytokine and chemokine levels, and histology scores were attenuated in S100A9 knockout mice undergoing the double-hit. Exogenous S100A8/A9 and S100A8 induced neutrophil influx in spontaneously breathing mice. In ventilated mice, these proteins clearly amplified inflammation: neutrophil influx, cytokine, and chemokine levels were increased compared to ventilated vehicle-treated mice. In contrast, administration of S100A8/A9 to ventilated Toll-like receptor 4 mutant mice did not augment inflammation. CONCLUSION: S100A8/A9 proteins increase during lung injury and contribute to inflammation induced by HVT MV combined with lipopolysaccharide. In the absence of lipopolysaccharide, high levels of extracellular S100A8/A9 still amplify ventilator-induced lung injury via Toll-like receptor 4.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia , Animais , Líquido da Lavagem Broncoalveolar/química , Calgranulina B/efeitos adversos , Calgranulina B/genética , Humanos , Camundongos , Camundongos Knockout , Estatísticas não Paramétricas , Receptor 4 Toll-Like/genética
4.
Thromb Haemost ; 109(4): 726-37, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23348224

RESUMO

The protein C (PC) system is an important regulator of both coagulation and inflammation. Activated PC (APC), together with its receptor the endothelial protein C receptor (EPCR), has anticoagulant and anti-inflammatory properties. During tuberculosis (TB), a devastating chronic pulmonary disease caused by Mycobacterium (M.) tuberculosis, both a local inflammatory reaction characterised by the recruitment of mainly mononuclear cells and the formation of pulmonary granulomas as well as activation of coagulation occurs as part of the host immune response. We investigated the role of EPCR and APC in a mouse model of TBusing mice overexpressing EPCR (Tie2-EPCR), mice deficient for EPCR (EPCR-/-), mice treated with APC-inhibiting antibodies and mice overexpressing APC (APChigh) and compared them with wild-type (WT) mice. Blood and organs were harvested to quantify bacterial loads, cellular influxes, cytokines, histopathology and coagulation parameters. Additionally observation studies were performed. Lung EPCR expression was upregulated during experimental TB. No significant differences in bacterial growth were seen between WT and Tie2-EPCR mice. However, Tie2-EPCR mice had decreased pulmonary coagulation activation, displayed an increased influx of macrophages 2 and 6 weeks after infection, but no increase in other proinflammatory markers. On the other hand, in EPCR-/--mice coagulation activation was decreased 6 weeks post-infection, with little impact on other inflammation markers. APC-overexpression or treatment with anti-(A)PC antibodies displayed minimal effects during experimental TB. In conclusion, EPCR and APC play a limited role in the host response during experimental pulmonary TB.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Pulmão/metabolismo , Proteína C/metabolismo , Receptores de Superfície Celular/metabolismo , Tuberculose Pulmonar/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Coagulação Sanguínea , Citocinas/metabolismo , Modelos Animais de Doenças , Receptor de Proteína C Endotelial , Feminino , Glicoproteínas/deficiência , Glicoproteínas/genética , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/imunologia , Pulmão/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Mycobacterium tuberculosis/patogenicidade , Regiões Promotoras Genéticas , Proteína C/genética , Receptor TIE-2/genética , Fatores de Tempo , Tuberculose Pulmonar/sangue , Tuberculose Pulmonar/genética , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Regulação para Cima
5.
J Biol Chem ; 288(10): 7047-52, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23322781

RESUMO

Adenosine monophosphate-activated protein (AMP)-activated kinase (AMPK) is a highly conserved kinase that plays a key role in energy homeostasis. Activation of AMPK was shown to reduce inflammation in response to lipolysaccharide in vitro and in vivo. 5-Aminoimidazole-4-carbox-amide-1-ß-D-ribofuranoside (AICAR) is intracellularly converted to the AMP analog ZMP, which activates AMPK. Lipoteichoic acid (LTA) is a major component of the cell wall of Gram-positive bacteria that can trigger inflammatory responses. In contrast to lipopolysaccharide, little is known on the effects of AMPK activation in LTA-triggered innate immune responses. Here, we studied the potency of AMPK activation to reduce LTA-induced inflammation in vitro and in lungs in vivo. Activation of AMPK in vitro reduced cytokine production in the alveolar macrophage cell line MH-S. In vivo, AMPK activation reduced LTA-induced neutrophil influx, as well as protein leak and cytokine/chemokine levels in the bronchoalveolar space. In conclusion, AMPK activation inhibits LTA-induced lung inflammation in mice.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Pneumonia/prevenção & controle , Ribonucleotídeos/farmacologia , Acetil-CoA Carboxilase/metabolismo , Aminoimidazol Carboxamida/farmacologia , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Linhagem Celular , Quimiocinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Hipoglicemiantes/farmacologia , Interleucina-6/metabolismo , Lipopolissacarídeos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Fosforilação/efeitos dos fármacos , Pneumonia/induzido quimicamente , Pneumonia/enzimologia , Ácidos Teicoicos , Fator de Necrose Tumoral alfa/metabolismo
6.
PLoS One ; 7(11): e50559, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226314

RESUMO

INTRODUCTION: Uric acid released from injured tissue is considered a major endogenous danger signal and local instillation of uric acid crystals induces acute lung inflammation via activation of the NLRP3 inflammasome. Ventilator-induced lung injury (VILI) is mediated by the NLRP3 inflammasome and increased uric acid levels in lung lavage fluid are reported. We studied levels in human lung injury and the contribution of uric acid in experimental VILI. METHODS: Uric acid levels in lung lavage fluid of patients with acute lung injury (ALI) were determined. In a different cohort of cardiac surgery patients, uric acid levels were correlated with pulmonary leakage index. In a mouse model of VILI the effect of allopurinol (inhibits uric acid synthesis) and uricase (degrades uric acid) pre-treatment on neutrophil influx, up-regulation of adhesion molecules, pulmonary and systemic cytokine levels, lung pathology, and regulation of receptors involved in the recognition of uric acid was studied. In addition, total protein and immunoglobulin M in lung lavage fluid and pulmonary wet/dry ratios were measured as markers of alveolar barrier dysfunction. RESULTS: Uric acid levels increased in ALI patients. In cardiac surgery patients, elevated levels correlated significantly with the pulmonary leakage index. Allopurinol or uricase treatment did not reduce ventilator-induced inflammation, IκB-α degradation, or up-regulation of NLRP3, Toll-like receptor 2, and Toll-like receptor 4 gene expression in mice. Alveolar barrier dysfunction was attenuated which was most pronounced in mice pre-treated with allopurinol: both treatment strategies reduced wet/dry ratio, allopurinol also lowered total protein and immunoglobulin M levels. CONCLUSIONS: Local uric acid levels increase in patients with ALI. In mice, allopurinol and uricase attenuate ventilator-induced alveolar barrier dysfunction.


Assuntos
Alopurinol/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/fisiopatologia , Urato Oxidase/farmacologia , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia , Lesão Pulmonar Aguda/metabolismo , Adulto , Alopurinol/uso terapêutico , Animais , Líquido da Lavagem Broncoalveolar , Permeabilidade Capilar/efeitos dos fármacos , Proteínas de Transporte/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas I-kappa B/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Inibidor de NF-kappaB alfa , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Urato Oxidase/uso terapêutico , Ácido Úrico/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo
7.
J Transl Med ; 10: 201, 2012 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-23006534

RESUMO

BACKGROUND: Helium inhalation protects myocardium, brain and endothelium against ischemia/reperfusion injury in animals and humans, when applied according to specific "conditioning" protocols. Before widespread use of this "conditioning" agent in clinical practice, negative side effects have to be ruled out. We investigated the effect of prolonged helium inhalation on the responsiveness of the human immune response in whole blood ex vivo. METHODS: Male healthy volunteers inhaled 30 minutes heliox (79%He/21%O(2)) or air in a cross over design, with two weeks between measurements. Blood was withdrawn at T0 (baseline), T1 (25 min inhalation) and T2-T5 (1, 2, 6, 24 h after inhalation) and incubated with lipopolysaccharide (LPS), lipoteichoic acid (LTA), T-cell stimuli anti-CD3/ anti-CD28 (TCS) or RPMI (as control) for 2, 4 and 24 hours or not incubated (0 h). An additional group of six volunteers inhaled 60 minutes of heliox or air, followed by blood incubation with LPS and RPMI. Tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), interleukin-8 (IL-8), interferon-γ (IFN-γ) and interleukin-2 (IL-2) was analyzed by cytometric bead array. Statistical analysis was performed by the Wilcoxon test for matched samples. RESULTS: Incubation with LPS, LTA or TCS significantly increased TNF-α, IL-1ß, IL-6, IL-8, IFN-γ and IL-2 in comparison to incubation with RPMI alone. Thirty min of helium inhalation did not influence the amounts of TNF-α, IL-1ß, IL-6, IL-8, IFN-γ and IL-2 in comparison to air. Sixty min of helium inhalation did not affect cytokine production after LPS stimulation. CONCLUSIONS: We conclude that 79% helium inhalation does not affect the responsiveness of the human immune system in healthy volunteers. TRIAL REGISTRATION: Dutch Trial Register: http://www.trialregister.nl/ NTR2152.


Assuntos
Ar , Hélio/administração & dosagem , Imunidade Inata , Citocinas/metabolismo , Humanos , Exposição por Inalação , Masculino , Valores de Referência
8.
J Infect Dis ; 206(9): 1415-23, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22904341

RESUMO

BACKGROUND: Pneumonia is frequently caused by gram-negative pathogens, among which Klebsiella pneumoniae prominently features. Recognition of pathogen-associated molecular patterns by Toll-like receptors (TLRs) is important for an appropriate immune response during infection. TLR signaling can proceed via 2 distinct routes that are dependent on the adaptor proteins Myeloid differentiation primary response gene (88) (MyD88) and TIR-domain-containing adaptor-inducing interferon-ß (TRIF). The aim of the study was to determine the relative contribution of MyD88 and TRIF signaling in resident and hematopoietic cells to host defense during pneumonia. METHODS: Bone marrow chimeras of MyD88 deficient/wild type and TRIF mutant/wild type mice were created and infected with K. pneumoniae via the airways. RESULTS: MyD88 in both resident and hematopoietic cells contributed to survival and antibacterial defense in late-stage infection, whereas only TRIF in hematopoietic cells was protective. On the other hand, resident MyD88 and hematopoietic TRIF contributed to distant cellular injury. Resident MyD88 was pivotal for early chemokine release and neutrophil recruitment in the bronchoalveolar space. CONCLUSIONS: MyD88- and TRIF-dependent signaling has a differential contribution to host defense in different cell types that changes from early- to late-stage gram-negative pneumonia.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Infecções por Klebsiella/imunologia , Klebsiella pneumoniae/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Pneumonia Bacteriana/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Feminino , Infecções por Klebsiella/patologia , Klebsiella pneumoniae/patogenicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Pneumonia Bacteriana/patologia , Análise de Sobrevida
9.
Shock ; 38(4): 375-80, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22777120

RESUMO

Mechanical ventilation (MV) has the potential to induce lung damage in healthy lungs or aggravate existing lung injury. Polymorphonuclear neutrophil (PMN) recruitment plays an important role in driving the inflammatory response in ventilator-induced lung injury (VILI). The cyclin-dependent kinase inhibitor r-roscovitine has been shown to induce apoptosis in PMNs. In this study, we investigated the potential of r-roscovitine treatment in reducing lung damage in a mouse model of VILI. Mice were tracheotomized and subjected to lung-protective MV with lower (∼7.5 mL/kg) or lung-injurious MV with higher (∼15 mL/kg) tidal volume (VT). R-roscovitine treatment enhanced apoptosis in PMNs in vitro. Ventilator-induced lung injury was associated with pulmonary PMN influx in low and high VT MV. During lung-injurious MV, r-roscovitine treatment reduced the number of PMNs and lowered levels of the lung damage markers RAGE (receptor for advanced glycation end products) and total immunoglobulin M in bronchoalveolar lavage fluid. R-roscovitine did not affect cytokine or chemokine levels in the bronchoalveolar space, neither during lung-protective nor lung-injurious MV. Thus, r-roscovitine treatment reduces lung damage in VILI, possibly dependent on increased apoptosis of PMNs.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Pulmão/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Células Cultivadas , Quinases Ciclina-Dependentes/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Pulmão/patologia , Masculino , Camundongos , Neutrófilos/enzimologia , Neutrófilos/patologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Roscovitina , Lesão Pulmonar Induzida por Ventilação Mecânica/enzimologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Ventiladores Mecânicos/efeitos adversos
10.
Mol Med ; 18: 1086-95, 2012 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-22692577

RESUMO

Bacterial pneumonia remains associated with high morbidity and mortality. The gram-positive pathogen Streptococcus pneumoniae is the most common cause of community-acquired pneumonia. Lipoteichoic acid (LTA) is an important proinflammatory component of the gram-positive bacterial cell wall. R-roscovitine, a purine analog, is a potent cyclin-dependent kinase (CDK)-1, -2, -5 and -7 inhibitor that has the ability to inhibit the cell cycle and to induce polymorphonuclear cell (PMN) apoptosis. We sought to investigate the effect of R-roscovitine on LTA-induced activation of cell lines with relevance for lung inflammation in vitro and on lung inflammation elicited by either LTA or viable S. pneumoniae in vivo. In vitro R-roscovitine enhanced apoptosis in PMNs and reduced tumor necrosis factor (TNF)-α and keratinocyte chemoattractant (KC) production in MH-S (alveolar macrophage) and MLE-12/MLE-15 (respiratory epithelial) cell lines. In vivo R-roscovitine treatment reduced PMN numbers in bronchoalveolar lavage fluid during LTA-induced lung inflammation; this effect was reversed by inhibiting apoptosis. Postponed treatment with R-roscovitine (24 and 72 h) diminished PMN numbers in lung tissue during gram-positive pneumonia; this step was associated with a transient increase in pulmonary bacterial loads. R-roscovitine inhibits proinflammatory responses induced by the gram-positive stimuli LTA and S. pneumoniae. R-roscovitine reduces PMN numbers in lungs upon LTA administration by enhancing apoptosis. The reduction in PMN numbers caused by R-roscovitine during S. pneumoniae pneumonia may hamper antibacterial defense.


Assuntos
Pneumonia/tratamento farmacológico , Purinas/uso terapêutico , Streptococcus pneumoniae/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/citologia , Caspase 3/metabolismo , Linhagem Celular , Quimiocinas/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Contagem de Leucócitos , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Neutrófilos/patologia , Pneumonia/sangue , Pneumonia/induzido quimicamente , Pneumonia/microbiologia , Purinas/farmacologia , Roscovitina , Streptococcus pneumoniae/efeitos dos fármacos , Ácidos Teicoicos
11.
Mol Immunol ; 52(3-4): 183-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22698798

RESUMO

The development of active tuberculosis after infection with Mycobacterium tuberculosis is almost invariably associated with a persistent or transient state of relative immunodeficiency. The receptor for advanced glycation end products (RAGE) is a promiscuous receptor that is involved in pulmonary inflammation and infection. To investigate the role of RAGE in tuberculosis, we intranasally infected wild-type (Wt) and RAGE deficient (RAGE(-/-)) mice with live virulent M. tuberculosis. While lungs of uninfected Wt mice expressed RAGE, in particular on endothelium, M. tuberculosis pneumonia was associated with an enhanced pulmonary expression of RAGE. Lung inflammation was increased in RAGE(-/-) mice, as indicated by histopathology, percentage of inflamed area, lung weight and cytokine and chemokine levels. In addition, lung lymphocyte and neutrophil numbers were increased in the RAGE(-/-) mice. RAGE(-/-) mice had modestly higher mycobacterial loads in the lungs after 3 weeks but not after 6 weeks of infection. Moreover, RAGE(-/-) mice displayed more body weight loss and enhanced mortality. In summary, pulmonary RAGE expression is increased during tuberculosis. In addition, these data suggest that RAGE plays a beneficial role in the host response to pulmonary tuberculosis.


Assuntos
Receptores Imunológicos/fisiologia , Tuberculose Pulmonar/imunologia , Animais , Carga Bacteriana , Quimiocinas/biossíntese , Citocinas/biossíntese , Contagem de Leucócitos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/patogenicidade , Neutrófilos/imunologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/biossíntese , Receptores Imunológicos/genética , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
13.
Anesthesiology ; 116(5): 1104-15, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22531249

RESUMO

BACKGROUND: The innate immune response is important in ventilator-induced lung injury (VILI) but the exact pathways involved are not elucidated. The authors studied the role of the intracellular danger sensor NLRP3 inflammasome. METHODS: NLRP3 inflammasome gene expression was analyzed in respiratory epithelial cells and alveolar macrophages obtained from ventilated patients (n = 40). In addition, wild-type and NLRP3 inflammasome deficient mice were randomized to low tidal volume (approximately 7.5 ml/kg) and high tidal volume (approximately 15 ml/kg) ventilation. The presence of uric acid in lung lavage, activation of caspase-1, and NLRP3 inflammasome gene expression in lung tissue were investigated. Moreover, mice were pretreated with interleukin-1 receptor antagonist, glibenclamide, or vehicle before start of mechanical ventilation. VILI endpoints were relative lung weights, total protein in lavage fluid, neutrophil influx, and pulmonary and systemic cytokine and chemokine concentrations. Data represent mean ± SD. RESULTS: Mechanical ventilation up-regulated messenger RNA expression levels of NLRP3 in alveolar macrophages (1.0 ± 0 vs. 1.70 ± 1.65, P less than 0.05). In mice, mechanical ventilation increased both NLRP3 and apoptosis-associated speck-like protein messenger RNA levels, respectively (1.08 ± 0.55 vs. 3.98 ± 2.89; P less than 0.001 and 0.95 ± 0.53 vs. 6.0 ± 3.55; P less than 0.001), activated caspase-1, and increased uric acid levels (6.36 ± 1.85 vs. 41.9 ± 32.0, P less than 0.001). NLRP3 inflammasome deficient mice displayed less VILI due to high tidal volume mechanical ventilation compared with wild-type mice. Furthermore, treatment with interleukin-1 receptor antagonist or glibenclamide reduced VILI. CONCLUSIONS: Mechanical ventilation induced a NLRP3 inflammasome dependent pulmonary inflammatory response. NLRP3 inflammasome deficiency partially protected mice from VILI.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Inflamassomos/genética , Lesão Pulmonar Induzida por Ventilação Mecânica/genética , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Animais , Líquido da Lavagem Broncoalveolar/química , Caspase 1/metabolismo , Citocinas/sangue , Citocinas/metabolismo , Ativação Enzimática/fisiologia , Células Epiteliais/metabolismo , Glibureto/farmacologia , Humanos , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Infiltração de Neutrófilos , Tamanho do Órgão/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Interleucina-1/antagonistas & inibidores , Respiração Artificial , Volume de Ventilação Pulmonar/fisiologia , Regulação para Cima/fisiologia , Ácido Úrico/metabolismo
14.
Microbes Infect ; 14(9): 748-55, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22484384

RESUMO

Tuberculosis, caused by Mycobacterium (M.) tuberculosis, is a devastating infectious disease causing many deaths worldwide. Non-specific host defense mechanisms such as the coagulation and fibrinolytic system may give insight in possible new therapeutic targets. Plasminogen activator inhibitor type-1 (PAI-1), an important regulator of inflammation and fibrinolysis, might be of interest as tuberculosis patients have elevated plasma levels of PAI-1. In this study we set out to investigate the role of PAI-1 during tuberculosis in vivo. Wildtype (WT) and PAI-1 deficient (PAI-1⁻/⁻) mice were intranasally infected with M. tuberculosis H37rv and sacrificed after 2, 5 and 29 weeks. Five weeks post-infection, bacterial loads in lungs of PAI-1⁻/⁻ mice were significantly higher compared to WT mice, while no differences were seen 2 and 29 weeks post-infection. At two weeks post-infection increased influx of macrophages and lymphocytes was observed. PAI-1 deficiency was associated with a reduced cytokine response in the lungs; however, upon stimulation with tuberculin purified protein derivative (PPD), PAI-1⁻/⁻ splenocytes released increased levels of IFN-γ compared to WT. No clear differences were found between PAI-1⁻/⁻ and WT mice at 29 weeks after infection. In conclusion, these data suggest that PAI-1 contributes to transient, non-specific changes in immunity during the early phase of murine tuberculosis.


Assuntos
Mycobacterium tuberculosis/imunologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Tuberculose/imunologia , Animais , Carga Bacteriana , Citocinas/metabolismo , Modelos Animais de Doenças , Pulmão/microbiologia , Pulmão/patologia , Linfócitos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor 1 de Ativador de Plasminogênio/deficiência
15.
Infect Immun ; 80(5): 1853-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22331429

RESUMO

Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is an important cause of community-acquired sepsis in Southeast Asia and northern Australia. An important controller of the immune system is the pleiotropic cytokine transforming growth factor ß (TGF-ß), of which Smad2 and Smad3 are the major signal transducers. In this study, we aimed to characterize TGF-ß expression and function in experimental melioidosis. TGF-ß expression was determined in 33 patients with culture-proven infection with B. pseudomallei and 30 healthy controls. We found that plasma TGF-ß concentrations were strongly elevated during melioidosis. In line with this finding, TGF-ß expression in C57BL/6 mice intranasally inoculated with B. pseudomallei was enhanced as well. To assess the role of TGF-ß, we inhibited TGF-ß using a selective murine TGF-ß antibody. Treatment of mice with anti-TGF-ß antibody resulted in decreased lung Smad2 phosphorylation. TGF-ß blockade appeared to be protective: mice treated with anti-TGF-ß antibody and subsequently infected with B. pseudomallei showed diminished bacterial loads. Moreover, less distant organ injury was observed in anti-TGF-ß treated mice as shown by reduced blood urea nitrogen (BUN) and aspartate transaminase (AST) values. However, anti-TGF-ß treatment did not have an effect on survival. In conclusion, TGF-ß is upregulated during B. pseudomallei infection and plays a limited but proinflammatory role during experimental melioidosis.


Assuntos
Regulação da Expressão Gênica/fisiologia , Melioidose/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Inflamação/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Sepse , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/genética
16.
PLoS One ; 6(4): e18519, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21483672

RESUMO

BACKGROUND: Pneumonia represents a major health burden. Previous work demonstrated that although the induction of inflammation is important for adequate host defense against pneumonia, an inability to regulate the host's inflammatory response within the lung later during infection can be detrimental. Intracellular signaling pathways commonly rely on activation of kinases, and kinases play an essential role in the regulation of the inflammatory response of immune cells. METHODOLOGY/PRINCIPAL FINDINGS: Pneumonia was induced in mice via intranasal instillation of Streptococcus (S.) pneumoniae. Kinomics peptide arrays, exhibiting 1024 specific consensus sequences for protein kinases, were used to produce a systems biology analysis of cellular kinase activity during the course of pneumonia. Several differences in kinase activity revealed by the arrays were validated in lung homogenates of individual mice using western blot. We identified cascades of activated kinases showing that chemotoxic stress and a T helper 1 response were induced during the course of pneumococcal pneumonia. In addition, our data point to a reduction in WNT activity in lungs of S. pneumoniae infected mice. Moreover, this study demonstrated a reduction in overall CDK activity implying alterations in cell cycle biology. CONCLUSIONS/SIGNIFICANCE: This study utilizes systems biology to provide insight into the signaling events occurring during lung infection with the common cause of community acquired pneumonia, and may assist in identifying novel therapeutic targets in the treatment of bacterial pneumonia.


Assuntos
Fosfotransferases/metabolismo , Pneumonia Pneumocócica/enzimologia , Análise Serial de Proteínas/métodos , Streptococcus pneumoniae/patogenicidade , Animais , Western Blotting , Feminino , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
17.
Mol Med ; 17(7-8): 741-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21424109

RESUMO

Pneumonia is a severe disease with high morbidity and mortality. A major causative pathogen is the Gram-negative bacterium Klebsiella (K.) pneumoniae. Kinases play an integral role in the transduction of intracellular signaling cascades and regulate a diverse array of biological processes essential to immune cells. The current study explored signal transduction events during murine Gram-negative pneumonia using a systems biology approach. Kinase activity arrays enable the analysis of 1,024 consensus sequences of protein kinase substrates. Using a kinase activity array on whole lung lysates, cellular kinase activities were determined in a mouse model of K. pneumoniae pneumonia. Notable kinase activities also were validated with phospho-specific Western blots. On the basis of the profiling data, mitogen-activated protein kinase (MAPK) signaling via p42 mitogen-activated protein kinase (p42) and p38 mitogen-activated protein kinase (p38) and transforming growth factor ß (TGFß) activity were reduced during infection, whereas v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (avian) (SRC) activity generally was enhanced. AKT signaling was represented in both metabolic and inflammatory (mitogen-activated protein kinase kinase 2 [MKK], apoptosis signal-regulating kinase/mitogen-activated protein kinase kinase kinase 5 [ASK] and v-raf murine sarcoma viral oncogene homolog B1 [b-RAF]) context. This study reaffirms the importance of classic inflammation pathways, such as MAPK and TGFß signaling and reveals less known involvement of glycogen synthase kinase 3ß (GSK-3ß), AKT and SRC signaling cassettes in pneumonia.


Assuntos
Infecções por Klebsiella/enzimologia , Fosfotransferases/metabolismo , Pneumonia Bacteriana/enzimologia , Proteômica/métodos , Animais , Western Blotting , Quimiocinas/metabolismo , Análise por Conglomerados , Citocinas/metabolismo , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Interações Hospedeiro-Patógeno , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/fisiologia , Pulmão/enzimologia , Pulmão/metabolismo , Pulmão/microbiologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosforilação , Fosfotransferases/classificação , Pneumonia Bacteriana/microbiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/metabolismo
18.
Am J Respir Crit Care Med ; 183(7): 932-40, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20959556

RESUMO

RATIONALE: After surviving the initial hyperinflammatory phase, patients with sepsis display features consistent with immunosuppression, which renders the host susceptible to nosocomial infections, in particular bacterial pneumonia. Suppression of tumorigenicity 2 (ST2) is a negative regulator of Toll-like receptor signaling implicated in endotoxin tolerance. OBJECTIVES: The present study sought to determine the role of ST2 in modulating host defense in the lung during sepsis, using a murine model of cecal ligation and puncture (CLP)-induced sepsis followed by a secondary infection with Pseudomonas aeruginosa via the airways. METHODS: CLP or sham surgery was performed on BALB/c wild-type (WT) and ST2 knockout (KO) mice, and 24 hours later animals were challenged with 10(8) live P. aeruginosa. MEASUREMENTS AND MAIN RESULTS: CLP mice demonstrated impaired clearance of Pseudomonas from their lungs and reduced pulmonary levels of tumor necrosis factor-α and IL-6 compared with sham mice. After CLP, ST2KO mice with secondary pneumonia displayed a strongly improved survival and a better bacterial clearance compared with WT mice, which was accompanied by enhanced lung inflammation. CLP did not influence the responsiveness of alveolar macrophages toward P. aeruginosa ex vivo irrespective of the st2 genotype. In contrast, CLP resulted in a reduced capacity of WT CD4(+) and CD8(+) T cells to produce IFN-γ and tumor necrosis factor-α, an immune suppressive effect that was not seen in ST2KO mice. CONCLUSIONS: These findings indicate that gene products of ST2 contribute to the immune-compromised state during sepsis and the ensuing disturbed homeostasis of lung host defense.


Assuntos
Pneumonia Bacteriana/genética , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/imunologia , Receptores de Interleucina/metabolismo , Sepse/genética , Sepse/imunologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade Inata/fisiologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Estimativa de Kaplan-Meier , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia Bacteriana/mortalidade , Infecções por Pseudomonas/mortalidade , Distribuição Aleatória , Sepse/mortalidade , Estatísticas não Paramétricas , Taxa de Sobrevida , Fatores de Necrose Tumoral/metabolismo
19.
Crit Care ; 15(6): 235, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22216838

RESUMO

Mechanical ventilation (MV) has the potential to worsen pre-existing lung injury or even to initiate lung injury. Moreover, it is thought that injurious MV contributes to the overwhelming inflammatory response seen in patients with acute lung injury or acute respiratory distress syndrome. Ventilator-induced lung injury (VILI) is characterized by increased endothelial and epithelial permeability and pulmonary inflammation, in which the innate immune system plays a key role. A growing body of evidence indicates that endogenous danger molecules, also termed damage-associated molecular patterns (DAMPs), are released upon tissue injury and modulate the inflammatory response. DAMPs activate pattern recognition receptors, may induce the release of proinflammatory cytokines and chemokines, and have been shown to initiate or propagate inflammation in non-infectious conditions. Experimental and clinical studies demonstrate the presence of DAMPs in bronchoalveolar lavage fluid in patients with VILI and the upregulation of pattern recognition receptors in lung tissue by MV. The objective of the present article is to review research in the area of DAMPs, their recognition by the innate immune system, their role in VILI, and the potential utility of blocking DAMP signaling pathways to reduce VILI in the critically ill.


Assuntos
Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Animais , Proteínas de Transporte/fisiologia , Proteínas de Choque Térmico/fisiologia , Humanos , Ácido Hialurônico/fisiologia , Imunidade Inata/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/fisiologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/fisiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia
20.
PLoS Negl Trop Dis ; 4(8)2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20824216

RESUMO

BACKGROUND: Melioidosis, caused by infection with Burkholderia (B.) pseudomallei, is a severe illness that is endemic in Southeast Asia. Osteopontin (OPN) is a phosphorylated glycoprotein that is involved in several immune responses including induction of T-helper 1 cytokines and recruitment of inflammatory cells. METHODOLOGY AND PRINCIPAL FINDINGS: OPN levels were determined in plasma from 33 melioidosis patients and 31 healthy controls, and in wild-type (WT) mice intranasally infected with B. pseudomallei. OPN function was studied in experimental murine melioidosis using WT and OPN knockout (KO) mice. Plasma OPN levels were elevated in patients with severe melioidosis, even more so in patients who went on to die. In patients who recovered plasma OPN concentrations had decreased after treatment. In experimental melioidosis in mice plasma and pulmonary OPN levels were also increased. Whereas WT and OPN KO mice were indistinguishable during the first 24 hours after infection, after 72 hours OPN KO mice demonstrated reduced bacterial numbers in their lungs, diminished pulmonary tissue injury, especially due to less necrosis, and decreased neutrophil infiltration. Moreover, OPN KO mice displayed a delayed mortality as compared to WT mice. OPN deficiency did not influence the induction of proinflammatory cytokines. CONCLUSIONS: These data suggest that sustained production of OPN impairs host defense during established septic melioidosis.


Assuntos
Burkholderia pseudomallei/imunologia , Burkholderia pseudomallei/patogenicidade , Melioidose/imunologia , Melioidose/patologia , Osteopontina/sangue , Osteopontina/imunologia , Animais , Burkholderia pseudomallei/isolamento & purificação , Contagem de Colônia Microbiana , Humanos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Melioidose/microbiologia , Melioidose/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/patologia , Neutrófilos/imunologia , Osteopontina/análise , Osteopontina/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA