Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS One ; 13(11): e0207867, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30458050

RESUMO

Automated IHC double staining using diaminobenzidine and HRP Magenta is illustrated utilizing a new acidic block with sulfuric acid to prevent cross-reactivity. Residual cross-reactivity in double staining is determined to arise from chromogenic-bound antibodies and amplification system during the first part of the double staining.


Assuntos
Peroxidase do Rábano Silvestre/metabolismo , Imuno-Histoquímica/métodos , Coloração e Rotulagem/métodos , Automação , Benzidinas/metabolismo , Compostos de Diazônio/metabolismo
2.
PLoS One ; 9(3): e88259, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24642901

RESUMO

Elucidating the sites and mechanisms of sRAGE action in the healthy state is vital to better understand the biological importance of the receptor for advanced glycation end products (RAGE). Previous studies in animal models of disease have demonstrated that exogenous sRAGE has an anti-inflammatory effect, which has been reasoned to arise from sequestration of pro-inflammatory ligands away from membrane-bound RAGE isoforms. We show here that sRAGE exhibits in vitro binding with high affinity and reversibly to extracellular matrix components collagen I, collagen IV, and laminin. Soluble RAGE administered intratracheally, intravenously, or intraperitoneally, does not distribute in a specific fashion to any healthy mouse tissue, suggesting against the existence of accessible sRAGE sinks and receptors in the healthy mouse. Intratracheal administration is the only effective means of delivering exogenous sRAGE to the lung, the organ in which RAGE is most highly expressed; clearance of sRAGE from lung does not differ appreciably from that of albumin.


Assuntos
Colágeno Tipo IV/metabolismo , Colágeno Tipo I/metabolismo , Laminina/metabolismo , Receptores Imunológicos/metabolismo , Administração por Inalação , Animais , Disponibilidade Biológica , Fibronectinas/metabolismo , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Cinética , Pulmão/química , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/administração & dosagem , Receptores Imunológicos/isolamento & purificação , Solubilidade
3.
PLoS One ; 6(5): e20132, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21629785

RESUMO

BACKGROUND: The receptor for advanced glycation end-products (RAGE) has been suggested to modulate lung injury in models of acute pulmonary inflammation. To study this further, model systems utilizing wild type and RAGE knockout (KO) mice were used to determine the role of RAGE signaling in lipopolysaccharide (LPS) and E. coli induced acute pulmonary inflammation. The effect of intraperitoneal (i.p.) and intratracheal (i.t.) administration of mouse soluble RAGE on E. coli injury was also investigated. METHODOLOGY/PRINCIPAL FINDINGS: C57BL/6 wild type and RAGE KO mice received an i.t. instillation of LPS, E. coli, or vehicle control. Some groups also received i.p. or i.t. administration of mouse soluble RAGE. After 24 hours, the role of RAGE expression on inflammation was assessed by comparing responses in wild type and RAGE KO. RAGE protein levels decreased in wild type lung homogenates after treatment with either LPS or bacteria. In addition, soluble RAGE and HMGB1 increased in the BALF after E. coli instillation. RAGE KO mice challenged with LPS had the same degree of inflammation as wild type mice. However, when challenged with E. coli, RAGE KO mice had significantly less inflammation when compared to wild type mice. Most cytokine levels were lower in the BALF of RAGE KO mice compared to wild type mice after E. coli injury, while only monocyte chemotactic protein-1, MCP-1, was lower after LPS challenge. Neither i.p. nor i.t. administration of mouse soluble RAGE attenuated the severity of E. coli injury in wild type mice. CONCLUSIONS/SIGNIFICANCE: Lack of RAGE in the lung does not protect against LPS induced acute pulmonary inflammation, but attenuates injury following live E. coli challenge. These findings suggest that RAGE mediates responses to E. coli-associated pathogen-associated molecular pattern molecules other than LPS or other bacterial specific signaling responses. Soluble RAGE treatment had no effect on inflammation.


Assuntos
Escherichia coli/patogenicidade , Pulmão/metabolismo , Pulmão/microbiologia , Pneumonia/metabolismo , Pneumonia/microbiologia , Receptores Imunológicos/metabolismo , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL3/metabolismo , Técnicas In Vitro , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peroxidase/genética , Peroxidase/metabolismo , Pneumonia/genética , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Fator de Necrose Tumoral alfa/metabolismo
4.
Int J Clin Exp Pathol ; 4(3): 241-54, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21487520

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive disease with poor survival. The identification of therapeutic targets is essential to improving outcomes. Previous studies found that expression of the receptor for advanced glycation end products (RAGE) in the lung is significantly decreased in human IPF lungs and in two animal models of pulmonary fibrosis. In addition, RAGE-null mice spontaneously develop pulmonary fibrosis with age and more severe fibrosis when challenged with asbestos. In contrast to the findings that the lack of RAGE enhanced pulmonary fibrosis, He et al. found that RAGE null mice were protected from bleomycin-induced fibrosis and suggested the effect was due to a lack of HMGB1 induced RAGE signaling. The current study further tests this hypothesis by blocking RAGE signaling via administration of soluble RAGE, a decoy receptor, to determine if this will also protect against pulmonary fibrosis. Wild-type, RAGE(+/-), and RAGE(-/-) mice were treated with bleomycin and assessed for fibrosis. Wild-type mice were also treated with exogenous soluble RAGE or vehicle control. In addition, in vitro studies with primary alveolar epithelial cells from wild-type and RAGE null mice were used to investigate the effect of RAGE on cell viability and migration in response to injury. A lack of RAGE was found to be protective against bleomycin injury in both in vivo and in vitro studies. However, soluble RAGE administration was unable to ameliorate fibrosis. This study confirms paradoxical responses to two different models of pulmonary fibrosis and suggests a further role for RAGE in cellular migration.


Assuntos
Fibrose Pulmonar/metabolismo , Receptores Imunológicos/metabolismo , Análise de Variância , Animais , Bleomicina , Western Blotting , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibrose Pulmonar/induzido quimicamente , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Transdução de Sinais
5.
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
6.
Protein Expr Purif ; 61(1): 99-101, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18558495

RESUMO

The receptor for advanced glycation end-products (RAGE) has been implicated in numerous disease processes including: atherosclerosis, diabetic nephropathy, impaired wound healing and neuropathy to name a few. Treatment of animals with a soluble isoform of the receptor (sRAGE) has been shown to prevent and even reverse many disease processes. Isolating large quantities of pure sRAGE for in vitro and in vivo studies has hindered its development as a therapeutic strategy in other RAGE mediated diseases that require long-term therapy. This article provides an improvement in both yield and detail of a previously published method to obtain 10mg of pure, endotoxin free sRAGE from 65 g of lung tissue.


Assuntos
Pulmão/química , Receptor para Produtos Finais de Glicação Avançada/isolamento & purificação , Animais , Cromatografia Líquida , Ensaio de Imunoadsorção Enzimática , Humanos , Solubilidade , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
7.
J Biol Chem ; 283(22): 15031-6, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18385137

RESUMO

Human extracellular superoxide dismutase (EC-SOD) is a tetrameric glycoprotein responsible for the removal of superoxide generated in the extracellular space. Two different folding variants of EC-SOD exist based on the disulfide bridge connectivity, resulting in enzymatically active (aEC-SOD) and inactive (iEC-SOD) subunits. As a consequence of this, the assembly of the EC-SOD tetramers produces molecules with variable activity and may represent a way to regulate the antioxidant level in the extracellular space. To determine whether the formation of these two folding variants is an intra- or extracellular event, we analyzed the biosynthesis in human embryonic kidney 293 cells expressing wild-type EC-SOD. These analyses revealed that both folding variants were present in the intra- and extracellular spaces, suggesting that the formation is an intracellular event. To further analyze the biosynthesis, we constructed mutants with the capacity to generate only aEC-SOD (C195S) or iEC-SOD (C45S). The expression of these suggested that the cellular biosynthetic machinery supported the secretion of aEC-SOD but not iEC-SOD. The coexpression of these two mutants did not affect the expression pattern. This study shows that generation of the EC-SOD folding variants is an intracellular event that depends on a free cysteine residue not involved in disulfide bonding.


Assuntos
Espaço Extracelular/enzimologia , Dobramento de Proteína , Superóxido Dismutase/metabolismo , Substituição de Aminoácidos , Sequência de Bases , Linhagem Celular , Dissulfetos , Ativação Enzimática/fisiologia , Humanos , Dados de Sequência Molecular , Biossíntese de Proteínas/fisiologia , Superóxido Dismutase/genética , Superóxidos/metabolismo
8.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA