Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 35(12): 2496-507, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26515415

RESUMO

Atherosclerosis is mediated by local and systematic inflammation. The multiligand receptor for advanced glycation end products (RAGE) has been studied in animals and humans and is an important mediator of inflammation and atherosclerosis. This review focuses on S100/calgranulin proteins (S100A8, S100A9, and S100A12) and their receptor RAGE in mediating vascular inflammation. Mice lack the gene for S100A12, which in humans is located on chromosome 3 between S100A8 and S100A9. Transgenic mice with smooth muscle cell-targeted expression of S100A12 demonstrate increased coronary and aortic calcification, as well as increased plaque vulnerability. Serum S100A12 has recently been shown to predict future cardiovascular events in a longitudinal population study, underscoring a role for S100A12 as a potential biomarker for coronary artery disease. Genetic ablation of S100A9 or RAGE in atherosclerosis-susceptible apolipoprotein E null mice results in reduced atherosclerosis. Importantly, S100A12 and the RAGE axis can be modified pharmacologically. For example, soluble RAGE reduces murine atherosclerosis and vascular inflammation. Additionally, a class of compounds currently in phase III clinical trials for multiple sclerosis and rheumatologic conditions, the quinoline-3-carboxamides, reduce atherosclerotic plaque burden and complexity in transgenic S100A12 apolipoprotein E null mice, but have not been tested with regards to human atherosclerosis. The RAGE axis is an important mediator for inflammation-induced atherosclerosis, and S100A12 has emerged as biomarker for human atherosclerosis. Decreasing inflammation by inhibiting S100/calgranulin-mediated activation of RAGE attenuates murine atherosclerosis, and future studies in patients with coronary artery disease are warranted to confirm S100/RAGE as therapeutic target for atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Proteína S100A12/metabolismo , Animais , Anti-Inflamatórios/uso terapêutico , Aterosclerose/diagnóstico , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Biomarcadores/metabolismo , Modelos Animais de Doenças , Humanos , Inflamação/diagnóstico , Inflamação/tratamento farmacológico , Inflamação/genética , Mediadores da Inflamação/antagonistas & inibidores , Camundongos Knockout , Terapia de Alvo Molecular , Valor Preditivo dos Testes , Prognóstico , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Proteína S100A12/antagonistas & inibidores , Proteína S100A12/genética , Transdução de Sinais
2.
Artigo em Inglês | MEDLINE | ID: mdl-26082935

RESUMO

BACKGROUND: Cardiovascular disease including left ventricular hypertrophy, diastolic dysfunction and ectopic valvular calcification are common in patients with chronic kidney disease (CKD). Both S100A12 and fibroblast growth factor 23 (FGF23) have been identified as biomarkers of cardiovascular morbidity and mortality in patients with CKD. We tested the hypothesis that human S100/calgranulin would accelerate cardiovascular disease in mice subjected to CKD. METHODS: This review paper focuses on S100 proteins and their receptor for advanced glycation end products (RAGE) and summarizes recent findings obtained in novel developed transgenic hBAC-S100 mice that express S100A12 and S100A8/9 proteins. A bacterial artificial chromosome of the human S100/calgranulin gene cluster containing the genes and regulatory elements for S100A8, S100A9 and S100A12 was expressed in C57BL/6J mice (hBAC-S100). CKD was induced by ureteral ligation, and hBAC-S100 mice and WT mice were studied after 10 weeks of chronic uremia. RESULTS: hBAC-S100 mice with CKD showed increased FGF23 in the heart, left ventricular hypertrophy (LVH), diastolic dysfunction, focal cartilaginous metaplasia and calcification of the mitral and aortic valve annulus together with aortic valve sclerosis. This phenotype was not observed in WT mice with CKD or in hBAC-S100 mice lacking RAGE with CKD, suggesting that the inflammatory milieu mediated by S100/RAGE promotes pathological cardiac hypertrophy in CKD. In vitro, inflammatory stimuli including IL-6, TNFα, LPS, or serum from hBAC-S100 mice up regulated FGF23 mRNA and protein in primary murine neonatal and adult cardiac fibroblasts. CONCLUSIONS: Taken together, our study shows that myeloid-derived human S100/calgranulin is associated with the development of cardiac hypertrophy and ectopic cardiac calcification in a RAGE dependent manner in a mouse model of CKD. We speculate that FGF23 produced by cardiac fibroblasts in response to cytokines may act in a paracrine manner to accelerate LVH and diastolic dysfunction in hBAC-S100 mice with CKD. We suggest that S100/RAGE-mediated chronic sustained systemic inflammation is linked to pathological cardiac remodeling via direct up regulation of FGF23 in cardiac fibroblasts, thereby providing a new mechanistic understanding for the common association between CKD, diabetes, metabolic syndrome, or hypertension with left ventricular hypertrophy with diastolic dysfunction.

3.
J Allergy Ther ; 3(Suppl 1)2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25984393

RESUMO

Airway inflammation, lung remodeling, and Airway Hyperresponsiveness (AHR) are major features of asthma and Chronic Obstructive Pulmonary Disease (COPD). The inflammatory response to allergens, air pollutants, and other insults is likely to play a key role in promoting structural changes in the lung including the overabundance of Airway Smooth Muscle (ASM) seen in asthmatics. These alterations or remodeling could, in turn, impact the immunmodulatory actions of the ASM, the ASM's contractile properties, and the development of AHR. New evidences suggest that airway inflammation and AHR are not tightly related to each other and that the structural component of the airway, mainly the ASM, is a chief driver of AHR. Members of the S100/calgranulins family have been implicated in the regulation of inflammation and cell apoptosis in various systems. S100A12 is highly expressed in neutrophils and is one of the most abundant proteins in the lungs of patients with asthma or COPD. Studies with genetic engineered mice with smooth muscle cell-targeted expression of human S100A12 revealed that S100A12 reduces airway smooth muscle amounts and dampens airway inflammation and airway hyperreactivity in a model of allergic lung inflammation. Thus, targeting airway smooth muscle for instance through delivery of pro-apoptotic S100A12 could represent an attractive means to promote ASM apoptosis and to reduce ASM abundance in asthmatics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA