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










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L569-L582, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351444

RESUMO

In pulmonary fibrosis (PF), fibroblasts and myofibroblasts proliferate and deposit excessive extracellular matrix in the interstitium, impairing normal lung function. Because most forms of PF have a poor prognosis and limited treatment options, PF represents an urgent unmet need for novel, effective therapeutics. Although the role of immune cells in lung fibrosis is unclear, recent studies suggest that T lymphocyte (T cell) activation may be impaired in PF patients. Furthermore, we have previously shown that activated T cells can produce prostaglandins with anti-scarring potential. Here, we test the hypothesis that activated T cells directly inhibit myofibroblast differentiation using a coculture system. Coculture with activated primary blood-derived T cells, from both healthy human donors and PF patients, inhibited transforming growth factor ß-induced myofibroblast differentiation in primary human lung fibroblasts isolated from either normal or PF lung tissue. Coculture supernatants contained anti-fibrotic prostaglandins D2 and E2, and the inhibitory effect of coculture on myofibroblast differentiation was largely reversed when prostaglandin production was abrogated either by resting the T cells before coculture or via specific pharmacological inhibitors. Moreover, coculture conditions induced COX-2 in HLFs but not in T cells, suggesting that T cells deliver an activating signal to HLFs, which in turn produce anti-fibrotic prostaglandins. We show for the first time that coculture with activated primary human T lymphocytes strongly inhibits myofibroblast differentiation, revealing a novel cell-to-cell communication network with therapeutic implications for fibrotic lung diseases.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dinoprostona/metabolismo , Fibroblastos/patologia , Miofibroblastos/patologia , Prostaglandina D2/metabolismo , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/farmacologia , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Linfócitos T/metabolismo , Linfócitos T/patologia
2.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1305-12, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26408551

RESUMO

Myofibroblasts are one of the primary cell types responsible for the accumulation of extracellular matrix in fibrosing diseases, and targeting myofibroblast differentiation is an important therapeutic strategy for the treatment of pulmonary fibrosis. Transforming growth factor-ß (TGF-ß) has been shown to be an important inducer of myofibroblast differentiation. We previously demonstrated that lactate dehydrogenase and its metabolic product lactic acid are important mediators of myofibroblast differentiation, via acid-induced activation of latent TGF-ß. Here we explore whether pharmacologic inhibition of LDH activity can prevent TGF-ß-induced myofibroblast differentiation. Primary human lung fibroblasts from healthy patients and those with pulmonary fibrosis were treated with TGF-ß and or gossypol, an LDH inhibitor. Protein and RNA were analyzed for markers of myofibroblast differentiation and extracellular matrix generation. Gossypol inhibited TGF-ß-induced expression of the myofibroblast marker α-smooth muscle actin (α-SMA) in a dose-dependent manner in both healthy and fibrotic human lung fibroblasts. Gossypol also inhibited expression of collagen 1, collagen 3, and fibronectin. Gossypol inhibited LDH activity, the generation of extracellular lactic acid, and the rate of extracellular acidification in a dose-dependent manner. Furthermore, gossypol inhibited TGF-ß bioactivity in a dose-dependent manner. Concurrent treatment with an LDH siRNA increased the ability of gossypol to inhibit TGF-ß-induced myofibroblast differentiation. Gossypol inhibits TGF-ß-induced myofibroblast differentiation through inhibition of LDH, inhibition of extracellular accumulation of lactic acid, and inhibition of TGF-ß bioactivity. These data support the hypothesis that pharmacologic inhibition of LDH may play an important role in the treatment of pulmonary fibrosis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Ácido Láctico/biossíntese , Miofibroblastos/citologia , Miofibroblastos/efeitos dos fármacos , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Gossipol/farmacologia , Humanos , L-Lactato Desidrogenase/antagonistas & inibidores , L-Lactato Desidrogenase/metabolismo , Pulmão/patologia , Vison , Miofibroblastos/metabolismo , Fibrose Pulmonar/patologia , Doadores de Tecidos , Fator de Crescimento Transformador beta/farmacologia
3.
PLoS One ; 10(8): e0135266, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26248335

RESUMO

INTRODUCTION: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disease with very few effective treatments. The key effector cells in fibrosis are believed to be fibroblasts, which differentiate to a contractile myofibroblast phenotype with enhanced capacity to proliferate and produce extracellular matrix. The role of the lung epithelium in fibrosis is unclear. While there is evidence that the epithelium is disrupted in IPF, it is not known whether this is a cause or a result of the fibroblast pathology. We hypothesized that healthy epithelial cells are required to maintain normal lung homeostasis and can inhibit the activation and differentiation of lung fibroblasts to the myofibroblast phenotype. To investigate this hypothesis, we employed a novel co-culture model with primary human lung epithelial cells and fibroblasts to investigate whether epithelial cells inhibit myofibroblast differentiation. MEASUREMENTS AND MAIN RESULTS: In the presence of transforming growth factor (TGF)-ß, fibroblasts co-cultured with epithelial cells expressed significantly less α-smooth muscle actin and collagen and showed marked reduction in cell migration, collagen gel contraction, and cell proliferation compared to fibroblasts grown without epithelial cells. Epithelial cells from non-matching tissue origins were capable of inhibiting TGF-ß induced myofibroblast differentiation in lung, keloid and Graves' orbital fibroblasts. TGF-ß promoted production of prostaglandin (PG) E2 in lung epithelial cells, and a PGE2 neutralizing antibody blocked the protective effect of epithelial cell co-culture. CONCLUSIONS: We provide the first direct experimental evidence that lung epithelial cells inhibit TGF-ß induced myofibroblast differentiation and pro-fibrotic phenotypes in fibroblasts. This effect is not restricted by tissue origin, and is mediated, at least in part, by PGE2. Our data support the hypothesis that the epithelium plays a crucial role in maintaining lung homeostasis, and that damaged and/ or dysfunctional epithelium contributes to the development of fibrosis.


Assuntos
Dinoprostona/metabolismo , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Actinas/genética , Actinas/metabolismo , Anticorpos Neutralizantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Técnicas de Cocultura , Dinoprostona/antagonistas & inibidores , Dinoprostona/farmacologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Cultura Primária de Células
4.
Am J Respir Cell Mol Biol ; 50(4): 737-47, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24175906

RESUMO

Idiopathic pulmonary fibrosis (IPF) is characterized by progressive fibrotic destruction of normal lung architecture. Due to a lack of effective treatment options, new treatment approaches are needed. We previously identified transglutaminase (TG)2, a multifunctional protein expressed by human lung fibroblasts (HLFs), as a positive driver of fibrosis. TG2 catalyzes crosslinking of extracellular matrix proteins, enhances cell binding to fibronectin and integrin, and promotes fibronectin expression. We investigated whether the small electrophilic molecules 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) and 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2) inhibit the expression and profibrotic functions of TG2. CDDO and 15d-PGJ2 reduced expression of TG2 mRNA and protein in primary HLFs from control donors and donors with IPF. CDDO and 15d-PGJ2 also decreased the in vitro profibrotic effector functions of HLFs including collagen gel contraction and cell migration. The decrease in TG2 expression did not occur through activation of the peroxisome proliferator activated receptor γ or generation of reactive oxidative species. CDDO and 15d-PGJ2 inhibited the extracellular signal-regulated kinase pathway, resulting in the suppression of TG2 expression. This is the first study to show that small electrophilic compounds inhibit the expression and profibrotic effector functions of TG2, a key promoter of fibrosis. These studies identify new and important antifibrotic activities of these two small molecules, which could lead to new treatments for fibrotic lung disease.


Assuntos
Inibidores Enzimáticos/farmacologia , Fibrose Pulmonar Idiopática/enzimologia , Pulmão/efeitos dos fármacos , Ácido Oleanólico/análogos & derivados , Prostaglandina D2/análogos & derivados , Transglutaminases/antagonistas & inibidores , Estudos de Casos e Controles , Movimento Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo , Inibidores Enzimáticos/química , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/patologia , Proteínas de Ligação ao GTP , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/enzimologia , Pulmão/patologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Terapia de Alvo Molecular , Ácido Oleanólico/química , Ácido Oleanólico/farmacologia , Fosforilação , Prostaglandina D2/química , Prostaglandina D2/farmacologia , Proteína 2 Glutamina gama-Glutamiltransferase , Inibidores de Proteínas Quinases/farmacologia , Transglutaminases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...