Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 192(8): 3837-46, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24623132

RESUMO

Macrophages play a fundamental role in innate immunity and the pathogenesis of silicosis. Phagocytosis of silica particles is associated with the generation of reactive oxygen species (ROS), secretion of cytokines, such as TNF, and cell death that contribute to silica-induced lung disease. In macrophages, ROS production is executed primarily by activation of the NADPH oxidase (Phox) and by generation of mitochondrial ROS (mtROS); however, the relative contribution is unclear, and the effects on macrophage function and fate are unknown. In this study, we used primary human and mouse macrophages (C57BL/6, BALB/c, and p47(phox-/-)) and macrophage cell lines (RAW 264.7 and IC21) to investigate the contribution of Phox and mtROS to silica-induced lung injury. We demonstrate that reduced p47(phox) expression in IC21 macrophages is linked to enhanced mtROS generation, cardiolipin oxidation, and accumulation of cardiolipin hydrolysis products, culminating in cell death. mtROS production is also observed in p47(phox-/-) macrophages, and p47(phox-/-) mice exhibit increased inflammation and fibrosis in the lung following silica exposure. Silica induces interaction between TNFR1 and Phox in RAW 264.7 macrophages. Moreover, TNFR1 expression in mitochondria decreased mtROS production and increased RAW 264.7 macrophage survival to silica. These results identify TNFR1/Phox interaction as a key event in the pathogenesis of silicosis that prevents mtROS formation and reduces macrophage apoptosis.


Assuntos
Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Silicose/metabolismo , Animais , Morte Celular , Linhagem Celular , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Lesão Pulmonar/etiologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , NADPH Oxidases/genética , Ligação Proteica , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Dióxido de Silício/efeitos adversos , Dióxido de Silício/metabolismo , Silicose/genética
2.
Am J Pathol ; 182(4): 1286-96, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23490254

RESUMO

Influenza infection is widespread in the United States and the world. Despite low mortality rates due to infection, morbidity is common and little is known about the molecular events involved in recovery. Influenza infection results in persistent distal lung remodeling, and the mechanism(s) involved are poorly understood. Recently IL-22 has been found to mediate epithelial repair. We propose that IL-22 is critical for recovery of normal lung function and architecture after influenza infection. Wild-type and IL-22(-/-) mice were infected with influenza A PR8/34 H1N1 and were followed up for up to 21 days post infection. IL-22 receptor was localized to the airway epithelium in naive mice but was expressed at the sites of parenchymal lung remodeling induced by influenza infection. IL-22(-/-) mice displayed exacerbated lung injury compared with wild-type mice, which correlated with decreased lung function 21 days post infection. Epithelial metaplasia was observed in wild-type mice but was not evident in IL-22(-/-) animals that were characterized with an increased fibrotic phenotype. Gene expression analysis revealed aberrant expression of epithelial genes involved in repair processes, among changes in several other biological processes. These data indicate that IL-22 is required for normal lung repair after influenza infection. IL-22 represents a novel pathway involved in interstitial lung disease.


Assuntos
Epitélio/patologia , Epitélio/virologia , Interleucinas/metabolismo , Pulmão/patologia , Pulmão/virologia , Infecções por Orthomyxoviridae/patologia , Cicatrização , Animais , Membrana Basal/metabolismo , Membrana Basal/patologia , Colágeno/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Epitélio/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Interleucinas/deficiência , Pulmão/fisiopatologia , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/fisiopatologia , Infecções por Orthomyxoviridae/virologia , Receptores de Interleucina/metabolismo , Testes de Função Respiratória , Transdução de Sinais/genética , Interleucina 22
3.
Am J Physiol Lung Cell Mol Physiol ; 304(5): L350-60, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23275622

RESUMO

Hypozincemia, with hepatic zinc accumulation at the expense of other organs, occurs in infection, inflammation, and aseptic lung injury. Mechanisms underlying zinc partitioning or its impact on extrahepatic organs are unclear. Here we show that the major zinc-binding protein, metallothionein (MT), is critical for zinc transmigration from lung to liver during hyperoxia and preservation of intrapulmonary zinc during hyperoxia is associated with an injury-resistant phenotype in MT-null mice. Particularly, lung-to-liver zinc ratios decreased in wild-type (WT) and increased significantly in MT-null mice breathing 95% oxygen for 72 h. Compared with female adult WT mice, MT-null mice were significantly protected against hyperoxic lung injury indicated by reduced inflammation and interstitial edema, fewer necrotic changes to distal airway epithelium, and sustained lung function at 72 h hyperoxia. Lungs of MT-null mice showed decreased levels of immunoreactive LC3, an autophagy marker, compared with WT mice. Analysis of superoxide dismutase (SOD) activity in the lungs revealed similar levels of manganese-SOD activity between strains under normoxia and hyperoxia. Lung extracellular SOD activity decreased significantly in both strains at 72 h of hyperoxia, although there was no difference between strains. Copper-zinc-SOD activity was ~4× higher under normoxic conditions in MT-null compared with WT mice but was not affected in either group by hyperoxia. Collectively the data suggest that genetic deletion of MT-I/II in mice is associated with compensatory increase in copper-zinc-SOD activity, prevention of hyperoxia-induced zinc transmigration from lung to liver, and hyperoxia-resistant phenotype strongly associated with differences in zinc homeostasis during hyperoxic acute lung injury.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Metalotioneína/metabolismo , Superóxido Dismutase/metabolismo , Zinco/metabolismo , Animais , Feminino , Hiperóxia , Inflamação/imunologia , Metalotioneína/genética , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/análise , Mucosa Respiratória/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L664-71, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20729388

RESUMO

Accumulating evidence suggests that gender can have a profound effect on incidence and severity of a variety of pulmonary diseases. To address the influence of gender on the development of silica-induced pulmonary fibrosis, we instilled 0.2 g/kg silica into male and female C57BL/6 mice and examined the fibrotic and inflammatory response at 14 days postexposure. Both silica-exposed male and female mice had significant increases in total lung hydroxyproline compared with saline controls. However, silica-exposed female mice had significantly less total lung hydroxyproline than silica-exposed male mice. This observation was confirmed by color thresholding image analysis. Interestingly, silica-exposed female mice had significantly more inflammatory cells, the majority of which were macrophages, as well as higher levels of the macrophage-specific chemokines MCP-1 and CCL9 in whole lung lavage compared with silica-exposed male mice. We also show that at baseline, estrogen receptor α (ERα) mRNA expression is lower in female mice than in males and that ERα mRNA expression is decreased by silica exposure. Finally, we show that the response of ovariectomized female mice to silica instillation is similar to that of male mice. These observations together show that gender influences the lung response to silica.


Assuntos
Fibrose Pulmonar/induzido quimicamente , Dióxido de Silício/efeitos adversos , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Hidroxiprolina/análise , Pulmão/citologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Fatores Sexuais
5.
Antioxid Redox Signal ; 10(2): 379-85, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18031201

RESUMO

Apoptosis plays an important role in both normal lung homeostasis and lung remodeling associated with fibrotic lung disease. Whether apoptosis promotes or inhibits the pathogenesis of pulmonary fibrosis depends upon the cell type involved and the microenvironment of the affected lung. Undue cell loss in the alveolar epithelium may be important early in idiopathic pulmonary fibrosis (IPF) progression, while reduced fibroblast and myofibroblast apoptosis has been associated with the formation of fibrotic lesions. As such, novel therapies based on the stimulation or inhibition of apoptosis may prove beneficial to the treatment of patients with IPF.


Assuntos
Apoptose , Estresse Oxidativo , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Humanos , Mitocôndrias/fisiologia , Modelos Biológicos , Transdução de Sinais
6.
Free Radic Biol Med ; 40(4): 601-7, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16458190

RESUMO

Asbestosis is a chronic form of interstitial lung disease characterized by inflammation and fibrosis that results from the inhalation of asbestos fibers. Although the pathogenesis of asbestosis is poorly understood, reactive oxygen species may mediate the progression of this disease. The antioxidant enzyme extracellular superoxide dismutase (EC-SOD) can protect the lung against a variety of insults; however, its role in asbestosis is unknown. To determine if EC-SOD plays a direct role in protecting the lung from asbestos-induced injury, intratracheal injections of crocidolite were given to wild-type and ec-sod-null mice. Bronchoalveolar lavage fluid (BALF) from asbestos-treated ec-sod-null mice at 24 h, 14 days, or 28 days posttreatment showed increased inflammation and total BALF protein content compared to that of wild-type mice. In addition, lungs from ec-sod-null mice showed increased hydroxyproline content compared to those of wild-type mice, indicating a greater fibrotic response. Finally, lungs from ec-sod-null mice showed greater oxidative damage, as assessed by nitrotyrosine content compared to those of their wild-type counterparts. These results indicate that depletion of EC-SOD from the lung increases oxidative stress and injury in response to asbestos.


Assuntos
Asbesto Crocidolita/toxicidade , Pneumopatias/induzido quimicamente , Pulmão , Estresse Oxidativo , Animais , Líquido da Lavagem Broncoalveolar/química , Hidroxiprolina/metabolismo , Inflamação , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pneumopatias/enzimologia , Lesão Pulmonar , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Superóxido Dismutase/genética , Superóxido Dismutase/fisiologia , Tirosina/análogos & derivados , Tirosina/metabolismo
7.
Environ Health Perspect ; 124(8): 1199-207, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26955063

RESUMO

BACKGROUND: Fibrotic lung diseases occur predominantly in males, and reports describe better survival in affected females. Male mice are more sensitive to silica-induced lung fibrosis than silica-treated female mice. Secreted phosphoprotein 1 (SPP1, also known as osteopontin) increases in pulmonary fibrosis, and Spp1 transcription may be regulated by estrogen or estrogen receptor-related receptors. OBJECTIVE: We determined whether differences in silica-induced SPP1 levels contribute to sex differences in lung fibrosis. METHODS: Male and female mice were treated with 0.2 g/kg intratracheal silica, and lung injury was assessed 1, 3, or 14 days post-exposure. Gene-targeted (Spp1-/-) mice, control Spp1+/+ (C57BL/6J) mice, ovariectomized (OVX) female mice, and estrogen-treated male mice were treated with silica, and lung injury was assessed. RESULTS: Silica-induced SPP1 in lung tissue, bronchoalveolar lavage, and serum increased more in male than in female mice. Following silica treatment, bronchoalveolar lavage cell infiltrates decreased in female Spp1-/- mice compared with female Spp1+/+ mice, and lung hydroxyproline decreased in male Spp1-/- mice compared with male Spp1+/+ mice. OVX female mice had increased lung SPP1 expression in response to silica compared with silica-treated sham female mice. Silica-induced lung collagen and hydroxyproline (markers of fibrosis), and SPP1 levels decreased in estrogen-treated males compared with untreated males. CONCLUSION: These findings suggest that sex-specific differences in SPP1 levels contribute to the differential sensitivity of male and female mice to the development of silica-induced fibrosis. CITATION: Latoche JD, Ufelle AC, Fazzi F, Ganguly K, Leikauf GD, Fattman CL. 2016. Secreted phosphoprotein 1 and sex-specific differences in silica-induced pulmonary fibrosis in mice. Environ Health Perspect 124:1199-1207; http://dx.doi.org/10.1289/ehp.1510335.


Assuntos
Poluentes Atmosféricos/toxicidade , Exposição por Inalação/análise , Osteopontina/metabolismo , Dióxido de Silício/toxicidade , Animais , Líquido da Lavagem Broncoalveolar , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores Sexuais
8.
Free Radic Biol Med ; 35(3): 236-56, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12885586

RESUMO

Accumulated evidence has shown that reactive oxygen species (ROS) are important mediators of cell signaling events such as inflammatory reactions (superoxide) and the maintenance of vascular tone (nitric oxide). However, overproduction of ROS such as superoxide has been associated with the pathogenesis of a variety of diseases including cardiovascular diseases, neurological disorders, and pulmonary diseases. Antioxidant enzymes are, in part, responsible for maintaining low levels of these oxygen metabolites in tissues and may play key roles in controlling or preventing these conditions. One key antioxidant enzyme implicated in the regulation of ROS-mediated tissue damage is extracellular superoxide dismutase (EC-SOD). EC-SOD is found in the extracellular matrix of tissues and is ideally situated to prevent cell and tissue damage initiated by extracellularly produced ROS. In addition, EC-SOD is likely to play an important role in mediating nitric oxide-induced signaling events, since the reaction of superoxide and nitric oxide can interfere with nitric oxide signaling. This review will discuss the regulation of EC-SOD and its role in a variety of oxidant-mediated diseases.


Assuntos
Superóxido Dismutase/fisiologia , Animais , Antioxidantes/metabolismo , Doenças Cardiovasculares/patologia , Radicais Livres , Glicosilação , Humanos , Inflamação , Pneumopatias/patologia , Modelos Biológicos , Modelos Genéticos , Doenças do Sistema Nervoso/patologia , Óxido Nítrico/metabolismo , Estresse Oxidativo , Estrutura Terciária de Proteína , Espécies Reativas de Oxigênio , Traumatismo por Reperfusão , Transdução de Sinais , Superóxido Dismutase/metabolismo
9.
Free Radic Biol Med ; 35(7): 763-71, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-14583340

RESUMO

Extracellular superoxide dismutase (EC-SOD) is highly expressed in the extracellular matrix of lung and vascular tissue. Localization of EC-SOD to the matrix of the lung may protect against oxidative tissue damage that leads to pulmonary fibrosis. This study directly examines the protective role of EC-SOD in a bleomycin model of pulmonary fibrosis and the effect of this enzyme on oxidative protein fragmentation. Mice null for ec-sod display a marked increase in lung inflammation at 14 d post-bleomycin treatment as compared to their wild-type counterparts. Hydroxyproline analysis determined that both wild-type and ec-sod null mice display a marked increase in interstitial fibrosis at 14 d post-treatment, and the severity of fibrosis is significantly increased in ec-sod null mice compared to wild-type mice. To determine if the lack of EC-SOD promotes bleomycin-induced oxidative protein modification, 2-pyrrolidone content (as a measure of oxidative protein fragmentation at proline residues) was assessed in lung tissue from treated mice. 2-Pyrrolidone levels in the lung hydrolysates from ec-sod null mice were increased at both 7 and 14 d post-bleomycin treatment as compared to wild-type mice, indicating EC-SOD can inhibit oxidative fragmentation of proteins in this specific model of oxidative stress.


Assuntos
Bleomicina/farmacologia , Espaço Extracelular/enzimologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Superóxido Dismutase/deficiência , Animais , Líquido da Lavagem Broncoalveolar/química , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Hidroxiprolina/análise , Pulmão/química , Pulmão/enzimologia , Pneumopatias/induzido quimicamente , Pneumopatias/enzimologia , Pneumopatias/patologia , Camundongos , Camundongos Transgênicos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/patologia , Pirrolidinonas/análise , Superóxido Dismutase/metabolismo
10.
J Appl Physiol (1985) ; 97(5): 2006-13, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15298984

RESUMO

Inhalation of asbestos fibers leads to interstitial lung disease (asbestosis) characterized by inflammation and fibrosis. The pathogenesis of asbestosis is not fully understood, but reactive oxygen species are thought to play a central role. Extracellular superoxide dismutase (EC-SOD) is an antioxidant enzyme that protects the lung in a bleomycin-induced pulmonary fibrosis model, but its role has not been studied in asbestos-mediated disease. EC-SOD is found in high levels in the extracellular matrix of lung alveoli because of its positively charged heparin-binding domain. Proteolytic removal of this domain results in clearance of EC-SOD from the matrix of tissues. We treated wild-type C57BL/6 mice with 0.1 mg of crocidolite asbestos by intratracheal instillation and euthanized them 24 h later. Compared with saline- or titanium dioxide-treated control mice, bronchoalveolar lavage fluid (BALF) from asbestos-treated mice contained significantly higher total protein levels and increased numbers of inflammatory cells, predominantly neutrophils, indicating acute lung injury in response to asbestos. Decreased EC-SOD protein and activity were found in the lungs of asbestos-treated mice, whereas more EC-SOD was found in the BALF of these mice. The EC-SOD in the BALF was predominantly in the proteolyzed form, which lacks the heparin-binding domain. This redistribution of EC-SOD correlated with development of fibrosis 14 days after asbestos exposure. These data suggest that asbestos injury leads to enhanced proteolysis and clearance of EC-SOD from lung parenchyma into the air spaces. The depletion of EC-SOD from the extracellular matrix may increase susceptibility of the lung to oxidative stress during asbestos-mediated lung injury.


Assuntos
Amianto/farmacologia , Pulmão/enzimologia , Superóxido Dismutase/metabolismo , Animais , Amianto/administração & dosagem , Instilação de Medicamentos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeo Hidrolases/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/patologia , Distribuição Tecidual/efeitos dos fármacos , Traqueia
11.
PLoS One ; 7(7): e40789, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22815821

RESUMO

Acute exacerbations of pulmonary fibrosis are characterized by rapid decrements in lung function. Environmental factors that may contribute to acute exacerbations remain poorly understood. We have previously demonstrated that exposure to inhaled lipopolysaccharide (LPS) induces expression of genes associated with fibrosis. To address whether exposure to LPS could exacerbate fibrosis, we exposed male C57BL/6 mice to crystalline silica, or vehicle, followed 28 days later by LPS or saline inhalation. We observed that mice receiving both silica and LPS had significantly more total inflammatory cells, more whole lung lavage MCP-1, MIP-2, KC and IL-1ß, more evidence of oxidative stress and more total lung hydroxyproline than mice receiving either LPS alone, or silica alone. Blocking oxidative stress with N-acetylcysteine attenuated whole lung inflammation but had no effect on total lung hydroxyproline. These observations suggest that exposure to innate immune stimuli, such as LPS in the environment, may exacerbate stable pulmonary fibrosis via mechanisms that are independent of inflammation and oxidative stress.


Assuntos
Imunidade Inata/efeitos dos fármacos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , Acetilcisteína/farmacologia , Administração por Inalação , Animais , Lavagem Broncoalveolar , Citocinas/metabolismo , Água Potável , Hidroxiprolina/metabolismo , Inflamação/patologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Carbonilação Proteica/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Dióxido de Silício
12.
Free Radic Biol Med ; 50(10): 1288-96, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21362472

RESUMO

Previous work by others suggests that there is a strain-dependent variation in the susceptibility to inflammatory lung injury in mice. Specifically, the 129/J mice appear to be more resistant to asbestos-induced pulmonary fibrosis than the C57BL/6 strain. A separate line of evidence suggests that extracellular superoxide dismutase (ecSOD) may play an important role in protecting the lung from such injuries. We have recently reported that the 129/J strain of mice has an ecSOD genotype and phenotype distinctly different from those of the C57BL/6 mice. In order to identify ecSOD as a potential "asbestos-injury resistance" gene, we bred congenic mice, on the C57BL/6 background, carrying the wild type (sod3wt) or the 129/J (sod3129) allele for ecSOD. This allowed us to examine the role of ecSOD polymorphism in susceptibility to lung injury in an otherwise identical genetic background. Interestingly, asbestos treatment induces a significant (~40%) increase in plasma ecSOD activity in the sod3129 mice, but not in the sod3wt mice. Asbestos administration results in a loss of ecSOD activity and protein from lung tissue of both congenic strains, but the lung ecSOD activity remains significantly higher in sod3129 mice. As expected, asbestos treatment results in a significant recovery of ecSOD protein in bronchoalveolar lavage fluid (BALF). The BALF of sod3129 mice also have significantly lower levels of proteins and inflammatory cells, especially neutrophils, accompanied by a significantly lower extent of lung injury, as measured by a pathology index score or hydroxyproline content. Immunohistochemistry reveals a significant loss of ecSOD from the tips of the respiratory epithelial cells in response to asbestos treatment and that the loss of immunodetectable ecSOD is compensated for by enzyme expression by infiltrating cells, especially in the sod3wt mice. Our studies thus identify ecSOD as an important anti-inflammatory gene, responsible for most, if not all of the resistance to asbestos-induced lung injury reported for the 129/J strain of mice. The data further suggest allele-specific differences in the regulation of ecSOD expression. These congenic mice therefore represent a very useful model to study the role of this enzyme in all inflammatory diseases. Polymorphisms in human ecSOD have also been reported and it appears logical to assume that such variations may have a profound effect on disease susceptibility.


Assuntos
Espaço Extracelular/enzimologia , Fibrose Pulmonar/metabolismo , Superóxido Dismutase/genética , Alelos , Animais , Amianto , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Superóxido Dismutase/sangue , Superóxido Dismutase/metabolismo
14.
PLoS One ; 5(3): e9604, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20333255

RESUMO

BACKGROUND: The role of the receptor for advanced glycation end-products (RAGE) has been shown to differ in two different mouse models of asbestos and bleomycin induced pulmonary fibrosis. RAGE knockout (KO) mice get worse fibrosis when challenged with asbestos, whereas in the bleomycin model they are largely protected against fibrosis. In the current study the role of RAGE in a mouse model of silica induced pulmonary fibrosis was investigated. METHODOLOGY/PRINCIPAL FINDINGS: Wild type (WT) and RAGE KO mice received a single intratracheal (i.t.) instillation of silica in saline or saline alone as vehicle control. Fourteen days after treatment mice were subjected to a lung mechanistic study and the lungs were lavaged and inflammatory cells, protein and TGF-beta levels in lavage fluid determined. Lungs were subsequently either fixed for histology or excised for biochemical assessment of fibrosis and determination of RAGE protein- and mRNA levels. There was no difference in the inflammatory response or degree of fibrosis (hydroxyproline levels) in the lungs between WT and RAGE KO mice after silica injury. However, histologically the fibrotic lesions in the RAGE KO mice had a more diffuse alveolar septal fibrosis compared to the nodular fibrosis in WT mice. Furthermore, RAGE KO mice had a significantly higher histologic score, a measure of affected areas of the lung, compared to WT silica treated mice. A lung mechanistic study revealed a significant decrease in lung function after silica compared to control, but no difference between WT and RAGE KO. While a dose response study showed similar degrees of fibrosis after silica treatment in the two strains, the RAGE KO mice had some differences in the inflammatory response compared to WT mice. CONCLUSIONS/SIGNIFICANCE: Aside from the difference in the fibrotic pattern, these studies showed no indicators of RAGE having an effect on the severity of pulmonary fibrosis following silica injury.


Assuntos
Regulação da Expressão Gênica , Produtos Finais de Glicação Avançada/metabolismo , Receptores Imunológicos/genética , Silicose/metabolismo , Animais , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Hidroxiprolina/metabolismo , Inflamação , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibrose Pulmonar/genética , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/fisiologia , Fator de Crescimento Transformador beta/metabolismo
15.
J Environ Pathol Toxicol Oncol ; 28(2): 153-62, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19817702

RESUMO

We tested the postulate that asbestos exposure alters iron homeostasis in the mouse lung. Crocidolite asbestos (100 microg intratracheally) was instilled into C57BL/6 mice. TiO2 served as a control exposure. Using iron staining and immunohistochemistry, concentrations of this metal and expression of several iron transport and storage proteins were evaluated at one day and one month following asbestos exposure. Iron was not stainable one day following asbestos instillation but was increased one month later. There was an elevated expression of duodenal cytochrome b (Dcytb), divalent metal transporter 1 (DMT1), and ferritin at both one day and one month after crocidolite exposure. While ferroportin (FPN1) expression was increased one day after asbestos exposure, levels of this metal exporter had returned to baseline at one month. TiO2 did not affect changes in either the iron concentration or the expression of these iron-related proteins at one day and one month. We conclude that asbestos exposure alters lung iron homeostasis with an accumulation of the metal resulting. Elevations in available iron affect changes in the expression of Dcytb, DMT1, ferritin, and FPN1, which further modify metal homeostasis in the lung.


Assuntos
Asbesto Crocidolita/toxicidade , Proteínas de Transporte de Cátions/metabolismo , Citocromos b/metabolismo , Ferro/metabolismo , Animais , Duodeno/enzimologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL
16.
Am J Physiol Lung Cell Mol Physiol ; 294(3): L572-81, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18178676

RESUMO

Matrix metalloproteinases (MMPs) are mediators of lung injury, and their activity has been associated with the development of pulmonary fibrosis. To understand how MMPs regulate the development of pulmonary fibrosis, we examined MMP expression in two strains of mice with differing sensitivities to the fibrosis-inducing drug bleomycin. After a single intratracheal injection of the drug, bleomycin-sensitive C57BL/6 mice showed increased expression for MMPs (-2, -7, -9, -13) at both 7 and 14 days posttreatment compared with the bleomycin-resistant BALB/c strain. In addition, TIMP-1, an endogenous inhibitor of MMPs, was upregulated in the lungs of C57BL/6 mice but not BALB/c mice. We designed two strategies to decrease MMP expression to potentially decrease sensitivity of C57BL/6 mice: 1) we engineered C57BL/6 mice that overexpressed TIMP-1 in their lungs via surfactant protein C (SP-C) promoter; and 2) we inhibited expression of MMPs independent of TIMP-1 by knocking out metallothionein (MT), a critical zinc binding protein. SP-C-TIMP-1 mice reduced MMP expression in response to bleomycin. However, they were equally sensitive to bleomycin as their wild-type counterparts, displaying similar levels of hydroxyproline in the lung tissue. MT null mice displayed decreased lung activity of MMPs with no change in TIMP-1. Nonetheless, there was no difference between the MT null and wild-type control littermates with regards to any of the lung injury parameters measured. We conclude that although TIMP-1 expression is differentially regulated in fibrosis-sensitive and fibrosis-resistant strains, epithelial overexpression of TIMP-1 does not appear to substantially alter fibrotic lung disease in mice.


Assuntos
Fibrose Pulmonar/induzido quimicamente , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Animais , Bleomicina , Ativação Enzimática , Epitélio/metabolismo , Feminino , Metaloproteinases da Matriz/metabolismo , Metalotioneína/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regulação para Cima
17.
Am J Pathol ; 172(3): 583-91, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18245812

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a severely debilitating disease associated with a dismal prognosis. There are currently no effective therapies for IPF, thus the identification of novel therapeutic targets is greatly needed. The receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin superfamily of cell surface receptors whose activation has been linked to various pathologies. In healthy adult animals, RAGE is expressed at the highest levels in the lung compared to other tissues. To investigate the hypothesis that RAGE is involved in IPF pathogenesis, we have examined its expression in two mouse models of pulmonary fibrosis and in human tissue from IPF patients. In each instance we observed a depletion of membrane RAGE and its soluble (decoy) isoform, sRAGE, in fibrotic lungs. In contrast to other diseases in which RAGE signaling promotes pathology, immunohistochemical and hydroxyproline quantification studies on aged RAGE-null mice indicate that these mice spontaneously develop pulmonary fibrosis-like alterations. Furthermore, when subjected to a model of pulmonary fibrosis, RAGE-null mice developed more severe fibrosis, as measured by hydroxyproline assay and histological scoring, than wild-type controls. Combined with data from other studies on mouse models of pulmonary fibrosis and human IPF tissues indicate that loss of RAGE contributes to IPF pathogenesis.


Assuntos
Fibrose Pulmonar/genética , Receptores Imunológicos/fisiologia , Fatores Etários , Animais , Amianto/toxicidade , Asbestose/genética , Bovinos , Regulação para Baixo , Humanos , Pulmão/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
18.
Am J Respir Cell Mol Biol ; 34(2): 226-32, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16224105

RESUMO

Extracellular superoxide dismutase (EC-SOD) is an antioxidant abundant in the lung. Previous studies demonstrated depletion of lung parenchymal EC-SOD in mouse models of interstitial lung disease coinciding with an accumulation of EC-SOD in airspaces. EC-SOD sticks to the matrix by a proteolytically sensitive heparin-binding domain; therefore, we hypothesized that interstitial inflammation and matrix remodeling contribute to proteolytic redistribution of EC-SOD from lung parenchyma into the airspaces. To determine if inflammation limited to airspaces leads to EC-SOD redistribution, we examined a bacterial pneumonia model. This model led to increases in airspace polymorphonuclear leukocytes staining strongly for EC-SOD. EC-SOD accumulated in airspaces at 24 h without depletion of EC-SOD from lung parenchyma. This led us to hypothesize that airspace EC-SOD was released from inflammatory cells and was not a redistribution of matrix EC-SOD. To test this hypothesis, transgenic mice with lung-specific expression of human EC-SOD were treated with asbestos or bleomycin to initiate an interstitial lung injury. In these studies, EC-SOD accumulating in airspaces was entirely the mouse isoform, demonstrating an extrapulmonary source (inflammatory cells) for this EC-SOD. We also demonstrate that EC-SOD knockout mice possess greater lung inflammation in response to bleomycin and bacteria when compared with wild types. We conclude that the source of accumulating EC-SOD in airspaces in interstitial lung disease is inflammatory cells and not the lung and that interstitial processes such as those found in pulmonary fibrosis are required to remove EC-SOD from lung matrix.


Assuntos
Pulmão/enzimologia , Pneumonia/enzimologia , Pneumonia/patologia , Superóxido Dismutase/metabolismo , Animais , Bleomicina/toxicidade , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Infecções por Escherichia coli/enzimologia , Espaço Extracelular , Humanos , Pulmão/patologia , Doenças Pulmonares Intersticiais/enzimologia , Doenças Pulmonares Intersticiais/patologia , Lesão Pulmonar , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neutrófilos/metabolismo , Neutrófilos/patologia , Superóxido Dismutase/genética
19.
Am J Respir Cell Mol Biol ; 35(3): 289-97, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16574944

RESUMO

Inhalation of asbestos fibers causes pulmonary inflammation and eventual pulmonary fibrosis (asbestosis). Although the underlying molecular events are poorly understood, protease/antiprotease and oxidant/antioxidant imbalances are believed to contribute to the disease. Implicated in other forms of pulmonary fibrosis, the matrix metalloproteinases (MMPs) have not been examined in asbestosis. We therefore hypothesized that MMPs play a pathogenic role in asbestosis development. Wild-type C57BL/6 mice were intratracheally instilled with 0.1 mg crocidolite asbestos, causing an inflammatory response at 1 d and a developing fibrotic response at 7, 14, and 28 d. Gelatin zymography demonstrated an increase in MMP-9 (gelatinase B) during the inflammatory phase, while MMP-2 (gelatinase A) was profoundly increased in the fibrotic phase. Immunohistochemistry revealed MMP-9 in and around bronchiolar and airspace neutrophils that were often associated with visible asbestos fibers. MMP-2 was found in fibrotic regions at 7, 14, and 28 d. No increases in RNA levels of MMP-2, MMP-9, or MMP-8 were found, but levels of MMP-7, MMP-12, and MMP-13 RNA did increase at 14 d. The MMP inhibitors, TIMP-1 and TIMP-2, were also increased at 7-28 d after asbestos exposure. To confirm the importance of MMP activity in disease progression, mice exposed to asbestos were given daily injections of the MMP inhibitor, GM6001. MMP inhibition reduced inflammation and fibrosis in asbestos-treated mice. Collectively, these data suggest that MMPs contribute to the pathogenesis of asbestosis through effects on inflammation and fibrosis development.


Assuntos
Asbesto Crocidolita/toxicidade , Metaloproteinases da Matriz/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/enzimologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/enzimologia , Animais , Dipeptídeos/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Inibidores de Metaloproteinases de Matriz , Metaloproteinases da Matriz/análise , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/patologia , Inibidores de Proteases/farmacologia , Fibrose Pulmonar/patologia
20.
Am J Respir Crit Care Med ; 172(4): 417-22, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15894605

RESUMO

Idiopathic ulmonary fibrosis (histopathology of usual interstitial pneumonia) is a progressive lung disease of unknown etiology. No treatment has been shown to improve the prognosis of the patients with this disease. Recent evidence, including the observations that the patients with idiopathic pulmonary fibrosis have higher levels of oxidant stress than control patients, and a recent multicenter European study examining the effect of the antioxidant N-acetylcysteine on the progression of idiopathic pulmonary fibrosis suggest that the cellular redox state may play a significant role in the progression of this disease. These complex mechanisms include activation of growth factors as well as regulation of matrix metalloproteinases and protease inhibitors. Potential future approaches for the therapy of interstitial pulmonary fibrosis may involve synthetic agents able to modulate cellular redox state. Investigation into therapeutic approaches to inhibit oxidant-mediated reactions in the initiation and progression of pulmonary fibrosis may provide hope for the future treatment of this disease.


Assuntos
Estresse Oxidativo , Fibrose Pulmonar/metabolismo , Animais , Antioxidantes/metabolismo , Modelos Animais de Doenças , Radicais Livres , Humanos , Pulmão/metabolismo , Oxirredução , Fibrose Pulmonar/terapia , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA