Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Anesth Analg ; 111(1): 164-70, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20519416

RESUMO

BACKGROUND: S-ketamine is frequently used for analgosedation, especially during sepsis and cardiovascular instability. Because S-ketamine blocks voltage-gated sodium (Na+) channels in neurons and skeletal muscle, it is conceivable that S-ketamine also blocks alveolar epithelial Na+ channels that are crucial for alveolar fluid clearance (AFC). We studied the effects of alveolar and IV S-ketamine on transalveolar Na+ transport and AFC, and investigated whether IV S-ketamine enters the alveolar space in response to endotoxemia-induced pulmonary inflammation. METHODS: Cultured rat alveolar type II (ATII) cells were exposed to S-ketamine and/or the Na+ channel blocker amiloride (100 microM) and transepithelial transport indicated by short circuit current (ISC) was measured in Ussing chambers. AFC was measured in fluid-instilled lungs of anesthetized rats with or without amiloride added to the instillate. S-ketamine was either added to the instillate or injected IV. To induce mild lung injury that might favor the appearance of IV S-ketamine at the alveolar surface, endotoxemia was induced by IV lipopolysaccharide (7.5 mg/kg). RESULTS: In ATII cells, S-ketamine (25 microg/mL) caused a decrease of ISC regardless of apical (-18.9%+/- 1.4%; P < 0.001) or basolateral (-20.4% +/- 3.7%; P < 0.001) application. In ATII cells pretreated with amiloride, addition of apical or basolateral S-ketamine did not decrease ISC. AFC was approximately 8% per 30 minutes in control rats. S-ketamine (5 microg/mL) in the instillate reduced AFC to 1.1% +/- 1.5% (P = 0.04) by decreasing amiloride-sensitive transepithelial Na+ transport. Intravenous S-ketamine (20 mg/kg) did not affect AFC (P = 0.31). In the presence of lipopolysaccharide-induced inflammation, the concentration of IV-injected S-ketamine in bronchoalveolar lavage fluid remained below the concentration that inhibited AFC. CONCLUSIONS: Although exposure of the rat alveolar epithelium to S-ketamine decreases amiloride-sensitive transalveolar Na+ transport and AFC, IV S-ketamine at clinically relevant bolus concentrations does not affect AFC, even in the presence of mild lung injury.


Assuntos
Anestésicos Dissociativos/farmacologia , Líquidos Corporais/metabolismo , Ketamina/farmacologia , Pulmão/metabolismo , Alvéolos Pulmonares/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Anestésicos Dissociativos/administração & dosagem , Animais , Câmaras de Exposição Atmosférica , Líquido da Lavagem Broncoalveolar/citologia , Separação Celular , Células Cultivadas , Endotoxemia/sangue , Injeções Intravenosas , Ketamina/administração & dosagem , Ketamina/análogos & derivados , Ketamina/sangue , Lipopolissacarídeos/toxicidade , Pulmão/efeitos dos fármacos , Masculino , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos
11.
Immunology ; 128(1 Suppl): e766-76, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19740338

RESUMO

Upon infection with Mycobacterium (M.) tuberculosis, the development of a strong T helper 1 (Th1)-mediated adaptive immune response is considered as being most important for containment of the infection. Osteopontin (OPN) is a phosphorylated glycoprotein that is chemotactic for inflammatory cells and has been implicated in the induction of Th1 responses and granulomatous disease. We tested the hypothesis that OPN facilitates protective immunity during M. tuberculosis infection using wild-type (WT) and OPN knockout (KO) mice in a model of pulmonary tuberculosis. OPN expression was up-regulated in alveolar macrophages and lymphoid cells during M. tuberculosis infection. There were no significant differences in bacterial outgrowth, inflammation or recruitment of lymphocytes, macrophages and polymorphonuclear cells in the lungs after 2 and 5 weeks of infection. However, the numbers of CD4(+) and CD8(+) T cells were reduced in the absence of OPN 5 weeks after infection. Similar concentrations of cytokine were observed in lungs from both WT mice and OPN KO mice; however, there was a trend towards decreased levels of interferon-gamma (IFN-gamma) in OPN KO mice 5 weeks after infection. Despite an unaltered immune response in the early phase of tuberculosis, OPN KO mice had a modest survival advantage. Of note, both pulmonary bacterial loads and lung inflammation were reduced in these mice 31 weeks after infection. These data suggest that OPN is not crucial for protective immunity upon M. tuberculosis infection and during the late phase of tuberculosis may even be detrimental for the host.


Assuntos
Macrófagos Alveolares/imunologia , Mycobacterium tuberculosis , Osteopontina/metabolismo , Tuberculose Pulmonar/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/microbiologia , Hipersensibilidade Tardia/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteopontina/genética , Tuberculina/imunologia , Tuberculose Pulmonar/patologia
12.
Cell Microbiol ; 10(1): 81-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17645551

RESUMO

Burkholderia pseudomallei, which causes melioidosis, a severe, mainly pulmonary disease endemic in South-East Asia, is considered to be the most pathogenic of the Burkholderia genus. B. thailandensis, however, is considered avirulent. We determined differences in patterns of inflammation of B. pseudomallei 1026b (clinical virulent isolate), B. pseudomallei AJ1D8 (an in vitro invasion-deficient mutant generated from strain 1026b by Tn5-OT182 mutagenesis) and B. thailandensis by intranasally inoculating C57BL/6 mice with each strain. Mice infected with B. thailandensis showed a markedly decreased bacterial outgrowth from lungs, spleen and blood 24 h after inoculation, compared with infection with B. pseudomallei and the invasion mutant AJ1D8. Forty-eight hours after inoculation, B. thailandensis was no longer detectable. This was consistent with elevated pulmonary cytokine and chemokine concentrations after infection with B. pseudomallei 1026b and AJ1D8, and the absence of these mediators 48 h, but not 24 h, after inoculation with B. thailandensis. Histological examination, however, did show marked pulmonary inflammation in the mice infected with B. thailandensis, corresponding with substantial granulocyte influx and raised myeloperoxidase levels. Survival experiments showed that infection with 1 x 10(3) cfu B. thailandensis was not lethal, whereas inoculation with 1 x 10(6) cfu B. thailandensis was equally lethal as 1 x 10(3) cfu B. pseudomallei 1026b or AJ1D8. These data show that B. pseudomallei AJ1D8 is just as lethal as wild-type B. pseudomallei in an in vivo mouse model, and B. thailandensis is perhaps more virulent than is often recognized.


Assuntos
Burkholderia/imunologia , Burkholderia/patogenicidade , Inflamação/patologia , Melioidose/microbiologia , Melioidose/patologia , Animais , Sangue/microbiologia , Burkholderia/genética , Contagem de Colônia Microbiana , Citocinas/análise , Elementos de DNA Transponíveis/genética , Granulócitos/imunologia , Pulmão/química , Pulmão/microbiologia , Pulmão/patologia , Melioidose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , Peroxidase/análise , Baço/microbiologia , Análise de Sobrevida
13.
Immunology ; 125(2): 272-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18393969

RESUMO

Toll-like receptors play an essential role in the innate recognition of micro-organisms by the host. CD14 is one of the extracellular adaptor proteins required for recognition of Gram-negative bacteria and possibly also Mycobacterium tuberculosis. Therefore, we intranasally infected wild-type (WT) and CD14 knock-out (KO) mice with virulent M. tuberculosis H37Rv. We found no differences in bacterial load in the main target organ lung up to 32 weeks after infection. From 20 weeks onward 57% of WT mice succumbed, whereas all CD14 KO mice survived. The improved outcome of CD14 KO mice was accompanied by reduced pulmonary inflammation; lung cell counts and percentage of inflamed lung tissue were reduced in CD14 WT mice. These data suggest that during chronic infection CD14 KO mice are protected from lethality caused by lung tuberculosis because of a reduction of the inflammatory response.


Assuntos
Receptores de Lipopolissacarídeos/imunologia , Mycobacterium tuberculosis/patogenicidade , Pneumonia/imunologia , Tuberculose Pulmonar/imunologia , Animais , Quimiocinas/metabolismo , Quimiotaxia/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Receptores de Lipopolissacarídeos/genética , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/crescimento & desenvolvimento , Pneumonia/microbiologia , Pneumonia/patologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia , Virulência
14.
PLoS Med ; 4(7): e248, 2007 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-17676990

RESUMO

BACKGROUND: Toll-like receptors (TLRs) are essential in host defense against pathogens by virtue of their capacity to detect microbes and initiate the immune response. TLR2 is seen as the most important receptor for gram-positive bacteria, while TLR4 is regarded as the gram-negative TLR. Melioidosis is a severe infection caused by the gram-negative bacterium, Burkholderia pseudomallei, that is endemic in Southeast Asia. We aimed to characterize the expression and function of TLRs in septic melioidosis. METHODS AND FINDINGS: Patient studies: 34 patients with melioidosis demonstrated increased expression of CD14, TLR1, TLR2, and TLR4 on the cell surfaces of monocytes and granulocytes, and increased CD14, TLR1, TLR2, TLR4, LY96 (also known as MD-2), TLR5, and TLR10 mRNA levels in purified monocytes and granulocytes when compared with healthy controls. In vitro experiments: Whole-blood and alveolar macrophages obtained from TLR2 and TLR4 knockout (KO) mice were less responsive to B. pseudomallei in vitro, whereas in the reverse experiment, transfection of HEK293 cells with either TLR2 or TLR4 rendered these cells responsive to this bacterium. In addition, the lipopolysaccharide (LPS) of B. pseudomallei signals through TLR2 and not through TLR4. Mouse studies: Surprisingly, TLR4 KO mice were indistinguishable from wild-type mice with respect to bacterial outgrowth and survival in experimentally induced melioidosis. In contrast, TLR2 KO mice displayed a markedly improved host defenses as reflected by a strong survival advantage together with decreased bacterial loads, reduced lung inflammation, and less distant-organ injury. CONCLUSIONS: Patients with melioidosis displayed an up-regulation of multiple TLRs in peripheral blood monocytes and granulocytes. Although both TLR2 and TLR4 contribute to cellular responsiveness to B. pseudomallei in vitro, TLR2 detects the LPS of B. pseudomallei, and only TLR2 impacts on the immune response of the intact host in vivo. Inhibition of TLR2 may be a novel treatment strategy in melioidosis.


Assuntos
Bacteriemia/metabolismo , Burkholderia pseudomallei , Melioidose/metabolismo , Receptor 2 Toll-Like/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Bacteriemia/imunologia , Linhagem Celular , Células Cultivadas , Citocinas/sangue , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Granulócitos/imunologia , Granulócitos/metabolismo , Granulócitos/microbiologia , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , Masculino , Melioidose/imunologia , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
15.
Microbes Infect ; 9(2): 134-41, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17224292

RESUMO

Mycobacterium tuberculosis and the associated disease tuberculosis are health risks causing many deaths worldwide each year in humans. M. tuberculosis targets dendritic cell (DC)-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN) to induce immunosuppression, since interaction of DC-SIGN with mycobacterial mannose-capped lipoarabinomannan (ManLAM) induces interleukin (IL)-10 and prevents DC maturation. We investigated the role of a murine homolog of DC-SIGN, SIGN Related 1 (SIGNR1), in a model of M. tuberculosis infection using SIGNR1 deficient (KO) mice. Although SIGNR1 is expressed by macrophages and not by DCs, it also interacts with M. tuberculosis similar to DC-SIGN. Peritoneal macrophages from SIGNR1 KO mice produce less IL-10 upon stimulation with ManLAM than those from wild-type mice, suggesting that the interaction of ManLAM with SIGNR1 can result in immunosuppression similar to its human homolog. Indeed, early in infection, we observed increased T cell activity in SIGNR1 KO mice and increased IFNgamma production by splenocytes in SIGNR1 KO mice. However, we did not detect any differences between WT and KO mice in mycobacterial loads in the lungs or distant organs after M. tuberculosis infection resulting in similar survival rates. Moreover, we found that SIGNR1 is not present on alveolar macrophages of uninfected mice nor is it induced on lung macrophages throughout infection. Therefore, our data suggest that although SIGNR1 has a similar binding specificity as DC-SIGN, its role is limited during murine M. tuberculosis infection.


Assuntos
Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/fisiologia , Lectinas Tipo C/deficiência , Lectinas Tipo C/fisiologia , Mycobacterium tuberculosis/imunologia , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/fisiologia , Linfócitos T/imunologia , Tuberculose/imunologia , Animais , Moléculas de Adesão Celular/genética , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Histocitoquímica , Humanos , Interferon gama/biossíntese , Interleucina-10/biossíntese , Lectinas Tipo C/genética , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Fígado/microbiologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia , Receptores de Superfície Celular/genética , Baço/imunologia , Baço/microbiologia , Sobrevida
17.
Microbes Infect ; 8(9-10): 2409-13, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16797208

RESUMO

Mycobacterium kansasii is an emerging pathogen that is able to induce pulmonary disease resembling tuberculosis. To determine the role of interleukin (IL-)1 in lung infection caused by this atypical mycobacterium, IL-1 receptor type 1 knockout (IL-1R(1) KO) and normal wild type mice were intranasally infected with M. kansasii. IL-1R(1) KO mice demonstrated a reduced antibacterial response in the lungs and an increased dissemination to the liver, which was accompanied by an enhanced pulmonary inflammatory response. These data identify IL-1 as an important component of the innate immune response to lung infection by M. kansasii.


Assuntos
Interleucina-1/imunologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium kansasii/imunologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Receptores Tipo I de Interleucina-1/imunologia , Animais , Humanos , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Mycobacterium não Tuberculosas/microbiologia , Receptores Tipo I de Interleucina-1/deficiência , Receptores Tipo I de Interleucina-1/genética
18.
Shock ; 25(4): 414-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16670645

RESUMO

Leptin is a pleiotrophic protein mainly produced by adipocytes that has been implicated as a link between nutritional status and immune function. Severe bacterial infection is associated with elevated plasma levels of leptin. To determine the role of leptin in the host response to bacterial pneumonia leptin deficient ob/ob mice and normal wild-type (WT) mice were intranasally infected with different doses of the Gram-positive pathogen Streptococcus (S.) pneumoniae or the Gram-negative bacterium Klebsiella (K.) pneumoniae. After infection with lower doses of either pathogen ob/ob mice displayed lower pulmonary levels of proinflammatory cytokines, in particular tumor necrosis factor-alpha and chemokines. However, after infection with a higher dose of S. pneumoniae or K. pneumoniae the lung concentrations of these inflammatory mediators did not differ between ob/ob and WT mice. In addition, the extent and severity of lung inflammation, as assessed by semi-quantitative histopathology scores, were similar in both mouse strains. Finally, leptin deficiency did not impact on the bacterial outgrowth in the lungs during either Gram-positive or Gram-negative pneumonia irrespective of the infective dose. These data suggest that although leptin may play a modest role in the regulation of inflammation during bacterial pneumonia, it does not contribute to host defense mechanisms that act to limit the outgrowth of S. pneumoniae or K. pneumoniae in the lower airways.


Assuntos
Infecções por Klebsiella/imunologia , Klebsiella pneumoniae/imunologia , Leptina/fisiologia , Pneumonia Pneumocócica/imunologia , Animais , Imunidade/fisiologia , Inflamação/metabolismo , Infecções por Klebsiella/metabolismo , Leptina/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/patologia
19.
Shock ; 22(6): 543-7, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15545826

RESUMO

Platelet-activating factor (PAF), a glycerophospholipid with proinflammatory properties, exerts its biological effects by interacting with the PAF receptor (PAFR) expressed on many different cell types. The PAFR specifically binds phosphorylcholine, the biologically active component of PAF. However, phosphorylcholine is also a component of the cell wall of nontypeable Haemophilus influenzae (NTHi). In recently published in vitro experiments, the invasion of respiratory epithelial cells by NTHi was mediated by the PAFR. To determine the role of the PAFR in host defense against pneumonia induced by NTHi, PAFR-deficient (PAFR-/-) and normal wild-type mice were intranasally inoculated with NTHi. The absence of a functional PAFR was associated with a normal innate immune response as indicated by similar bacterial counts, myeloperoxidase activity, and inflammation within the pulmonary compartment of PAFR-/- and wild-type mice. These data indicate that the PAFR does not interfere with the clearance of NTHi from the respiratory tract.


Assuntos
Infecções por Haemophilus/microbiologia , Haemophilus influenzae/imunologia , Glicoproteínas da Membrana de Plaquetas/deficiência , Receptores Acoplados a Proteínas G/deficiência , Sistema Respiratório/microbiologia , Animais , Quimiocina CXCL2 , Quimiocinas/análise , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Haemophilus influenzae/crescimento & desenvolvimento , Haemophilus influenzae/isolamento & purificação , Pulmão/enzimologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peroxidase/metabolismo , Glicoproteínas da Membrana de Plaquetas/genética , Receptores Acoplados a Proteínas G/genética , Sistema Respiratório/enzimologia
20.
J Microbiol Methods ; 49(1): 55-62, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11777582

RESUMO

Polymorphonuclear neutrophils (PMN) and mononuclear phagocytes represent an important first line and effector function in the control of Candida infections. Their relative contribution to host defence is frequently assessed by means of microbiological assays. However, reported results are divergent and might well be associated with study design-related issues. In the present study, we compared frequently used microbiological candidacidal assays, with the purpose of determining the most adequate method for assessment of phagocytosis and intracellular killing. We concluded that microbiological assays using yeast-phagocyte suspensions are inappropriate for the assessment of intracellular killing of Candida blastoconidia by murine macrophages, due to adherence or clumping of cells. In contrast, an adherent monolayer of phagocytes can be applied as a single microbiological assay to independently study the process of phagocytosis and intracellular killing, by exudate peritoneal macrophages as well as exudate peritoneal PMN.


Assuntos
Candida albicans/fisiologia , Macrófagos Peritoneais/microbiologia , Neutrófilos/microbiologia , Fagocitose/imunologia , Animais , Feminino , Humanos , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Micologia/métodos , Neutrófilos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA