Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lab Invest ; 90(7): 1078-90, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20458281

RESUMO

Castration experiments in rodents show that the stromal vasculature is critical to the androgen-mediated prostate growth regulation. However, the role of angiogenesis inhibitors, such as thrombospondin-1 (TSP-1), in this process is unclear. TSP-1 is a multifunctional glycoprotein that can function as a potent angiogenesis inhibitor and an in vivo activator of latent transforming growth factor-beta (TGF-beta) in some tissues. On the basis of these observations, we hypothesized that TSP-1 regulated androgen withdrawal-induced prostate regression and that this process was mediated not only through antiangiogenic activity but also through TGF-beta activation. To test this, we evaluated angiogenic activity in human prostate epithelial and stromal cells treated with androgens and hypoxia in vitro. TSP-1 knockout mice were characterized to investigate the in vivo functions of TSP-1. In vitro, we found that androgens and hypoxia differentially regulated TSP-1 and angiogenic activity. Androgens stimulated normal epithelial cell, but inhibited normal stromal cell, angiogenic activity. Conversely, hypoxia stimulated stromal while inhibiting epithelial activity. Thus, in vivo, net angiogenic activity must reflect cellular interactions. And, we found that media conditioned by epithelial cells grown under normoxic conditions stimulated stromal cell angiogenic activity, and if epithelial cells were grown under hypoxic conditions, stromal activity was further increased. TSP-1 levels, however, were unchanged. In vivo, TSP-1 loss in a mouse model led to prostate epithelial hyperplasia by 3 months of age with only a modest stromal effect. Androgens suppressed TSP-1 as expression increased after castration both in normal mouse prostate and in human prostate cancer tissues. In addition, TSP-1 expression corresponded to increased TGF-beta activation in mouse tissues, specifically in the stromal compartment. These data show a critical role for TSP-1 in prostate epithelial and stromal growth regulation through angiogenic inhibition and activation of latent TGF-beta. Therefore, loss of TSP-1 during tumorigenesis would eliminate two barriers to cancer progression.


Assuntos
Androgênios/deficiência , Neovascularização Fisiológica , Próstata/fisiologia , Trombospondina 1/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Carcinoma/fisiopatologia , Linhagem Celular , Di-Hidrotestosterona , Células Epiteliais/metabolismo , Humanos , Hiperplasia , Hipóxia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orquiectomia , Fenótipo , Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Células Estromais/fisiologia
2.
Adv Exp Med Biol ; 617: 591-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18497086

RESUMO

Pigment epithelium-derived factor (PEDF) is an endogenous antiangiogenic protein that also possesses antitumor activity. The mechanisms by which PEDF exerts its actions remains poorly understood. We sought to understand the role of PEDF in hepatocellular carcinoma (HCC), a hypervascular malignancy that has been shown to upregulate enzymes involved in fatty acid synthesis. PEDF expression occurs in two HCC cell lines and is oxygen dependent. Migration studies confirm PEDF's role as an endogenous inhibitor of angiogenesis in HCC cells. Loss of PEDF in an animal model leads to hepatocyte lipid accumulation, proliferation, and cellular atypia. To investigate potential interactions with transcription factors that are involved in fatty acid metabolism and cellular proliferation, we examined PEDF's interaction with PPARalpha in vitro and its functional activity through transactivation assays. We show that PEDF binds to PPARalpha but minimally to PPARgamma. In the presence of the ligand, ciprofibrate, PEDF binding to PPARalpha decreases whereas the presence of troglitazone does not alter PEDF interactions with PPARgamma. Transfection of the PEDF gene in the presence of the PPARalpha/RXR heterodimer demonstrates transcriptional activation of PPARalpha by PEDF. These data show that PEDF regulates lipid metabolism through activation of the transcription factor PPARalpha.


Assuntos
Carcinoma Hepatocelular/irrigação sanguínea , Proteínas do Olho/metabolismo , Metabolismo dos Lipídeos , Neoplasias Hepáticas/irrigação sanguínea , Neovascularização Patológica/patologia , Fatores de Crescimento Neural/metabolismo , PPAR alfa/metabolismo , Serpinas/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Hipóxia Celular , Movimento Celular/fisiologia , Cromanos/farmacologia , Ácido Clofíbrico/análogos & derivados , Ácido Clofíbrico/farmacologia , Endotélio Vascular/citologia , Ácidos Fíbricos , Humanos , Hipoglicemiantes/farmacologia , Hipolipemiantes/farmacologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Troglitazona , Células Tumorais Cultivadas
3.
Nat Med ; 9(6): 774-80, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12740569

RESUMO

Angiogenesis sustains tumor growth and metastasis, and recent studies indicate that the vascular endothelium regulates tissue mass. In the prostate, androgens drive angiogenic inducers to stimulate growth, whereas androgen withdrawal leads to decreased vascular endothelial growth factor, vascular regression and epithelial cell apoptosis. Here, we identify the angiogenesis inhibitor pigment epithelium-derived factor (PEDF) as a key inhibitor of stromal vasculature and epithelial tissue growth in mouse prostate and pancreas. In PEDF-deficient mice, stromal vessels were increased and associated with epithelial cell hyperplasia. Androgens inhibited prostatic PEDF expression in cultured cells. In vivo, androgen ablation increased PEDF in normal rat prostates and in human cancer biopsies. Exogenous PEDF induced tumor epithelial apoptosis in vitro and limited in vivo tumor xenograft growth, triggering endothelial apoptosis. Thus, PEDF regulates normal pancreas and prostate mass. Its androgen sensitivity makes PEDF a likely contributor to the anticancer effects of androgen ablation.


Assuntos
Inibidores da Angiogênese/metabolismo , Proteínas do Olho , Fatores de Crescimento Neural , Pâncreas/anatomia & histologia , Pâncreas/irrigação sanguínea , Próstata/anatomia & histologia , Próstata/irrigação sanguínea , Proteínas/metabolismo , Serpinas/metabolismo , Adolescente , Adulto , Idoso , Androgênios/metabolismo , Animais , Vasos Sanguíneos/anatomia & histologia , Vasos Sanguíneos/metabolismo , Castração , Cobalto/metabolismo , Humanos , Hiperplasia , Hipóxia , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Transplante de Neoplasias , Neovascularização Fisiológica , Próstata/patologia , Neoplasias da Próstata/metabolismo , Proteínas/genética , Ratos , Serpinas/genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...