Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L446-53, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20034963

RESUMO

Pulmonary expression of granulocyte/macrophage colony-stimulating factor (GM-CSF) is critically important for normal functional maturation of alveolar macrophages. We found previously that lung GM-CSF is dramatically suppressed in mice exposed to hyperoxia. Alveolar epithelial cells (AEC) are a major source of GM-CSF in the peripheral lung, and in vivo hyperoxia resulted in greatly reduced expression of GM-CSF protein by AEC ex vivo. We now explore the mechanisms responsible for this effect, using primary cultures of murine AEC exposed to hyperoxia in vitro. Exposure of AEC to 80% oxygen/5% CO(2) for 48 h did not induce overt toxicity, but resulted in significantly decreased GM-CSF protein and mRNA expression compared with cells in normoxia. Similar effects were seen when AEC were stressed with serum deprivation, an alternative inducer of oxidative stress. The effects in AEC were opposite those in a murine lung epithelial cell line (MLE-12 cells), in which hyperoxia induced GM-CSF expression. Both hyperoxia and serum deprivation resulted in increased intracellular reactive oxygen species (ROS) in AEC. Hyperoxia and serum deprivation induced significantly accelerated turnover of GM-CSF mRNA. Treatment of AEC with catalase during oxidative stress preserved GM-CSF protein and mRNA and was associated with stabilization of GM-CSF mRNA. We conclude that hyperoxia-induced suppression of AEC GM-CSF expression is a function of ROS-induced destabilization of GM-CSF mRNA. We speculate that AEC oxidative stress results in significantly impaired pulmonary innate immune defense due to effects on local GM-CSF expression in the lung.


Assuntos
Células Epiteliais Alveolares/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Hiperóxia/metabolismo , Hiperóxia/patologia , Estresse Oxidativo , Células Epiteliais Alveolares/efeitos dos fármacos , Animais , Catalase/farmacologia , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Meios de Cultura Livres de Soro , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Estresse Oxidativo/efeitos dos fármacos , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
Infect Immun ; 77(3): 1053-60, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19124601

RESUMO

Pneumocystis infections increase host susceptibility to additional insults that would be tolerated in the absence of infection, such as hyperoxia. In an in vivo model using CD4-depleted mice, we previously demonstrated that Pneumocystis murina pneumonia causes significant mortality following an otherwise nonlethal hyperoxic insult. Infected mice demonstrated increased pulmonary inflammation and alveolar epithelial cell apoptosis compared to controls. To test the mechanisms underlying these observations, we examined expression of components of the Fas-Fas ligand pathway in P. murina-infected mice exposed to hyperoxia. Hyperoxia alone increased expression of Fas on the surface of type II alveolar epithelial cells; conversely, infection with P. murina led to increased lung expression of Fas ligand. We hypothesized that inhibition of inflammatory responses or direct inhibition of alveolar epithelial cell apoptosis would improve survival in P. murina-infected mice exposed to hyperoxia. Mice were depleted of CD4(+) T cells and infected with P. murina and then were exposed to >95% oxygen for 4 days, followed by return to normoxia. Experimental groups received vehicle, dexamethasone, or granulocyte-macrophage colony-stimulating factor (GM-CSF). Compared with the vehicle-treated group, treatment with dexamethasone reduced Fas ligand expression and significantly improved survival. Similarly, treatment with GM-CSF, an agent we have shown protects alveolar epithelial cells against apoptosis, decreased Fas ligand expression and also improved survival. Our results suggest that the dual stresses of P. murina infection and hyperoxia induce lung injury via activation of the Fas-Fas ligand pathway and that corticosteroids and GM-CSF reduce mortality in P. murina-infected mice exposed to hyperoxic stress by inhibition of inflammation and apoptosis.


Assuntos
Lesão Pulmonar Aguda/imunologia , Apoptose/imunologia , Hiperóxia/imunologia , Inflamação/imunologia , Pneumonia por Pneumocystis/imunologia , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Dexametasona/farmacologia , Proteína Ligante Fas/efeitos dos fármacos , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Hiperóxia/complicações , Hiperóxia/patologia , Imuno-Histoquímica , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/complicações , Pneumonia/imunologia , Pneumonia/patologia , Pneumonia por Pneumocystis/complicações , Pneumonia por Pneumocystis/patologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Transdução de Sinais/imunologia , Receptor fas/efeitos dos fármacos , Receptor fas/imunologia , Receptor fas/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 291(6): L1246-55, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16891399

RESUMO

We have previously demonstrated that mice exposed to sublethal hyperoxia (an atmosphere of >95% oxygen for 4 days, followed by return to room air) have significantly impaired pulmonary innate immune response. Alveolar macrophages (AM) from hyperoxia-exposed mice exhibit significantly diminished antimicrobial activity and markedly reduced production of inflammatory cytokines in response to stimulation with LPS compared with AM from control mice in normoxia. As a consequence of these defects, mice exposed to sublethal hyperoxia are more susceptible to lethal pneumonia with Klebsiella pneumoniae than control mice. Granulocyte/macrophage colony-stimulating factor (GM-CSF) is a growth factor produced by normal pulmonary alveolar epithelial cells that is critically involved in maintenance of normal AM function. We now report that sublethal hyperoxia in vivo leads to greatly reduced alveolar epithelial cell GM-CSF expression. Systemic treatment of mice with recombinant murine GM-CSF during hyperoxia exposure preserved AM function, as indicated by cell surface Toll-like receptor 4 expression and by inflammatory cytokine secretion following stimulation with LPS ex vivo. Treatment of hyperoxic mice with GM-CSF significantly reduced lung bacterial burden following intratracheal inoculation with K. pneumoniae, returning lung bacterial colony-forming units to the level of normoxic controls. These data point to a critical role for continuous GM-CSF activity in the lung in maintenance of normal AM function and demonstrate that lung injury due to hyperoxic stress results in significant impairment in pulmonary innate immunity through suppression of alveolar epithelial cell GM-CSF expression.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Hiperóxia/fisiopatologia , Pulmão/fisiopatologia , Animais , Primers do DNA , Imunidade Inata , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Biol Chem ; 279(41): 42605-11, 2004 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-15294901

RESUMO

Lung surfactant is the surface-active agent comprised of phospholipids and proteins that lines pulmonary alveoli. Surfactant stabilizes the alveolar volume by reducing surface tension. Previously, we identified a lysosomal phospholipase A2, termed LPLA2, with specificity toward phosphatidylcholine and phosphatidylethanolamine. The phospholipase is localized to lysosomes, is calcium-independent, has an acidic pH optimum, and transacylates ceramide. Here, we demonstrate that LPLA2 is selectively expressed in alveolar macrophages but not in peritoneal macrophages, peripheral blood monocytes, or other tissues. Other macrophage-associated phospholipase A2s do not show a comparable distribution. LPLA2 is of high specific activity and recognizes disaturated phosphatidylcholine as a substrate. The lysosomal phospholipase A2 activity is six times lower in alveolar macrophages from mice with a targeted deletion of the granulocyte macrophage colony-stimulating factor (GM-CSF), a model of impaired surfactant catabolism, compared with those from wild-type mice. However, LPLA2 activity and protein levels are measured in GM-CSF null mice in which GM-CSF is expressed as a transgene under the control of the surfactant protein C promoter. Thus LPLA2 may be a major enzyme of pulmonary surfactant phospholipid degradation by alveolar macrophages and may be deficient in disorders of surfactant metabolism.


Assuntos
Lisossomos/enzimologia , Macrófagos Alveolares/metabolismo , Fosfolipases A/química , 1,2-Dipalmitoilfosfatidilcolina/química , Animais , Células COS , Primers do DNA/química , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Concentração de Íons de Hidrogênio , Immunoblotting , Leucócitos Mononucleares/metabolismo , Macrófagos/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Monócitos/metabolismo , Peptídeos/genética , Fosfolipases A2 , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , RNA/química , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Distribuição Tecidual , Transgenes
5.
Am J Pathol ; 163(6): 2397-406, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14633611

RESUMO

Granulocyte macrophage-colony stimulating factor (GM-CSF) plays an important role in pulmonary homeostasis, with effects on both alveolar macrophages and alveolar epithelial cells. We hypothesized that overexpression of GM-CSF in the lung would protect mice from hyperoxic lung injury by limiting alveolar epithelial cell injury. Wild-type C57BL/6 mice and mutant mice in which GM-CSF was overexpressed in the lung under control of the SP-C promoter (SP-C-GM mice) were placed in >95% oxygen. Within 6 days, 100% of the wild-type mice had died, while 70% of the SP-C-GM mice remained alive after 10 days in hyperoxia. Histological assessment of the lungs at day 4 revealed less disruption of the alveolar wall in SP-C-GM mice compared to wild-type mice. The concentration of albumin in bronchoalveolar lavage fluid after 4 days in hyperoxia was significantly lower in SP-C-GM mice than in wild-type mice, indicating preservation of alveolar epithelial barrier properties in the SP-C-GM mice. Alveolar fluid clearance was preserved in SP-C-GM mice in hyperoxia, but decreased significantly in hyperoxia-exposed wild-type mice. Staining of lung tissue for caspase 3 demonstrated increased apoptosis in alveolar wall cells in wild-type mice in hyperoxia compared to mice in room air. In contrast, SP-C-GM mice exposed to hyperoxia demonstrated only modest increase in alveolar wall apoptosis compared to room air. Systemic treatment with GM-CSF (9 micro g/kg/day) during 4 days of hyperoxic exposure resulted in decreased apoptosis in the lungs compared to placebo. In studies using isolated murine type II alveolar epithelial cells, treatment with GM-CSF greatly reduced apoptosis in response to suspension culture. In conclusion, overexpression of GM-CSF enhances survival of mice in hyperoxia; this effect may be explained by preservation of alveolar epithelial barrier function and fluid clearance, at least in part because of reduction in hyperoxia-induced apoptosis of cells in the alveolar wall.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Hiperóxia/prevenção & controle , Pneumopatias/prevenção & controle , Pulmão/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Hiperóxia/metabolismo , Hiperóxia/patologia , Hiperóxia/fisiopatologia , Pulmão/patologia , Pneumopatias/metabolismo , Pneumopatias/patologia , Pneumopatias/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/fisiopatologia , Proteínas Recombinantes/farmacologia , Albumina Sérica/metabolismo , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Infect Immun ; 71(10): 5970-8, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14500517

RESUMO

Patients with Pneumocystis pneumonia often develop respiratory failure after entry into medical care, and one mechanism for this deterioration may be increased alveolar epithelial cell injury. In vitro, we previously demonstrated that Pneumocystis is not cytotoxic for alveolar epithelial cells. In vivo, however, infection with Pneumocystis could increase susceptibility to injury by stressors that, alone, would be sublethal. We examined transient exposure to hyperoxia as a prototypical stress that does cause mortality in normal mice. Mice were depleted of CD4+ T cells and inoculated intratracheally with Pneumocystis. Control mice were depleted of CD4+ T cells but did not receive Pneumocystis. After 4 weeks, mice were maintained in normoxia, were exposed to hyperoxia for 4 days, or were exposed to hyperoxia for 4 days followed by return to normoxia. CD4-depleted mice with Pneumocystis pneumonia demonstrated significant mortality after transient exposure to hyperoxia, while all uninfected control mice survived this stress. We determined that organism burdens were not different. However, infected mice exposed to hyperoxia and then returned to normoxia demonstrated significant increases in inflammatory cell accumulation and lung cell apoptosis. We conclude that Pneumocystis pneumonia leads to increased mortality following a normally sublethal hyperoxic insult, accompanied by alveolar epithelial cell injury and increased pulmonary inflammation.


Assuntos
Hiperóxia/complicações , Pneumonia por Pneumocystis/complicações , Animais , Apoptose , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Humanos , Hiperóxia/imunologia , Hiperóxia/patologia , Inflamação/patologia , Lesão Pulmonar , Depleção Linfocítica , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fagocitose , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/patologia
7.
J Immunol ; 171(2): 955-63, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12847267

RESUMO

Supplemental oxygen is often required in the treatment of critically ill patients. The impact of hyperoxia on pulmonary host defense is not well-established. We hypothesized that hyperoxia directly impairs pulmonary host defense, beyond effects on alveolar wall barrier function. C57BL/6 mice were kept in an atmosphere of >95% O(2) for 4 days followed by return to room air. This exposure does not lead to mortality in mice subsequently returned to room air. Mice kept in room air served as controls. Mice were intratracheally inoculated with Klebsiella pneumoniae and followed for survival. Alveolar macrophages (AM) were harvested by bronchoalveolar lavage after 4 days of in vivo hyperoxia for ex vivo experiments. Mortality from pneumonia increased significantly in mice exposed to hyperoxia compared with infected mice in room air. Burden of organisms in the lung and dissemination of infection were increased in the hyperoxia group whereas accumulation of inflammatory cells in the lung was impaired. Hyperoxia alone had no impact on AM numbers, viability, or ability to phagocytize latex microbeads. However, following in vivo hyperoxia, AM phagocytosis and killing of Gram-negative bacteria and production of TNF-alpha and IL-6 in response to LPS were significantly reduced. AM surface expression of Toll-like receptor-4 was significantly decreased following in vivo hyperoxia. Thus sublethal hyperoxia increases Gram-negative bacterial pneumonia mortality and has a significant adverse effect on AM host defense function. Impaired AM function due to high concentrations of supplemental oxygen may contribute to the high rate of ventilator-associated pneumonia seen in critically ill patients.


Assuntos
Hiperóxia/imunologia , Hiperóxia/mortalidade , Pulmão/imunologia , Pulmão/fisiopatologia , Animais , Contagem de Células , Sobrevivência Celular/imunologia , Quimiocinas/biossíntese , Imunidade Inata , Mediadores da Inflamação/metabolismo , Interleucina-10/biossíntese , Interleucina-6/biossíntese , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/microbiologia , Infecções por Klebsiella/mortalidade , Infecções por Klebsiella/fisiopatologia , Klebsiella pneumoniae/crescimento & desenvolvimento , Klebsiella pneumoniae/imunologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Fagocitose/imunologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/mortalidade , Pneumonia Bacteriana/fisiopatologia , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/biossíntese , Receptores Toll-Like , Fator de Crescimento Transformador beta/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
8.
Ecotoxicol Environ Saf ; 54(1): 105-17, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12547641

RESUMO

In the US Environmental Protection Agency methods for sediment toxicity testing, the light regimen is specified as a 16:8 light dark cycle with 500-1000 lx. The potential for photoinduced toxic effects from this requirement is evaluated. Hyalella azteca were exposed to fluoranthene in both water only and sediment to examine the impact of light spectra on the toxicity of fluoranthene. The light sources included gold fluorescent light (lambda > 500 nm), cool white fluorescent light, and UV-enhanced fluorescent light. Toxicity was determined as mortality after 10 days of exposure. The extent of mortality was determined both as LC(50) and LR(50) (median lethal body residue). In water-only exposures, the toxicity of fluoranthene was greatest under the UV-enhanced spectra, followed by fluorescent light, and least toxic under the gold light. Both the LC(50) and LR(50) values exhibited the same pattern. The toxicity under gold light gave an LR(50) of 0.81 mmol kg(-1) (0.82-0.79, 95% CI) similar to values expected for the acute toxicity of nonpolar narcotic (anesthetic) compounds. The LR(50) values under the other two light sources were substantially lower, 4 and 58 times lower for the fluorescent and UV-enhanced exposures, respectively. In sediment, toxicity was not significantly affected by the light source. Toxicity occurred only when the body residue concentration approached that of the LR(50) under gold light from the water-only exposures. Thus, H. azteca were significantly protected from the light by burrowing into the sediment.


Assuntos
Anfípodes , Inibidores Enzimáticos/toxicidade , Fluorenos/toxicidade , Luz , Animais , Sedimentos Geológicos/química , Dose Letal Mediana , Fotoquímica
9.
Am J Physiol Lung Cell Mol Physiol ; 283(1): L180-7, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12060575

RESUMO

We postulate that intercellular adhesion molecule-1 (ICAM-1) on type I alveolar epithelial cells (AEC) facilitates phagocytic activity of alveolar macrophages (AM) in the alveolus. When wild-type and ICAM-1-deficient mice were inoculated intratracheally with FITC-labeled microspheres, AM phagocytosis of beads (after 1 and 4 h) was significantly reduced in ICAM-1-/- mice compared with controls. To focus on ICAM-1-mediated interactions specifically involving AM and AEC, rat AM were placed in culture with rat AEC treated with neutralizing anti-ICAM-1 F(ab')(2) fragments. Blocking ICAM-1 significantly decreased the AM phagocytosis of beads. Planar chemotaxis of AM over the surface of AEC was also significantly impaired by neutralization of AEC ICAM-1. ICAM-1 in rat AEC is associated with the actin cytoskeleton. Planar chemotaxis of AM was also significantly reduced by pretreatment of the AEC monolayer with cytochalasin B to disrupt the actin cytoskeleton. These studies indicate that ICAM-1 on the AEC surface promotes mobility of AM in the alveolus and is critically important for the efficient phagocytosis of particulates by AM.


Assuntos
Quimiotaxia/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Macrófagos Alveolares/imunologia , Fagocitose/imunologia , Mucosa Respiratória/citologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD18/imunologia , Reagentes de Ligações Cruzadas/farmacologia , Citoesqueleto/fisiologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Técnicas In Vitro , Molécula 1 de Adesão Intercelular/genética , Látex , Macrófagos Alveolares/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microesferas , Pneumonia/imunologia , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/imunologia , Mucosa Respiratória/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA