Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Pathol ; 168(1): 331-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16400034

ABSTRACT

The Eph receptor/ephrin system is a recently discovered regulator of vascular development during embryogenesis. Activation of EphA2, one of the Eph receptors, reportedly suppresses cell proliferation and adhesion in a wide range of cell types, including vascular endothelial cells. Vascular endothelial growth factor (VEGF) plays a primary role in both pathological angiogenesis and abnormal vascular leakage in diabetic retinopathy. In the study described herein, we demonstrated that EphA2 stimulation by ephrinA1 in cultured bovine retinal endothelial cells inhibits VEGF-induced VEGFR2 receptor phosphorylation and its downstream signaling cascades, including PKC (protein kinase C)-ERK (extracellular signal-regulated kinase) 1/2 and Akt. This inhibition resulted in the reduction of VEGF-induced angiogenic cell activity, including migration, tube formation, and cellular proliferation. These inhibitory effects were further confirmed in animal models. Intraocular injection of ephrinA1 suppressed ischemic retinal neovascularization in a dose-dependent manner in a mouse model. At a dose of 125 ng/eye, the inhibition was 36.0 +/- 14.9% (P < 0.001). EphrinA1 also inhibited VEGF-induced retinal vascular permeability in a rat model by 46.0 +/- 10.0% (P < 0.05). These findings suggest a novel therapeutic potential for EphA2/ephrinA1 in the treatment of neovascularization and vasopermeability abnormalities in diabetic retinopathy.


Subject(s)
Blood-Retinal Barrier/metabolism , Ephrin-A1/metabolism , Retinal Neovascularization/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Retinal Barrier/pathology , Blotting, Northern , Blotting, Western , Cattle , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/metabolism , Ephrin-A2/metabolism , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Protein v-akt/metabolism , Permeability , Protein Kinase C/metabolism , Rats , Retinal Vessels/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
J Biol Chem ; 280(36): 31841-9, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16000309

ABSTRACT

Oxidative stress activates various signal transduction pathways, including Jun N-terminal kinase (JNK) and its substrates, that induce apoptosis. We reported here the role of angiopoietin-1 (Ang1), which is a prosurvival factor in endothelial cells, during endothelial cell damage induced by oxidative stress. Hydrogen peroxide (H2O2) increased apoptosis of endothelial cells through JNK activation, whereas Ang1 inhibited H2O2-induced apoptosis and concomitant JNK phosphorylation. The inhibition of H2O2-induced JNK phosphorylation was reversed by inhibitors of phosphatidylinositol (PI) 3-kinase and dominant-negative Akt, and constitutively active-Akt attenuated JNK phosphorylation without Ang1. These data suggested that Ang1-dependent Akt phosphorylation through PI 3-kinase leads to the inhibition of JNK phosphorylation. H2O2-induced phosphorylation of SAPK/Erk kinase (SEK1) at Thr261, which is an upstream regulator of JNK, was also attenuated by Ang1-dependent activation of the PI 3-kinase/Akt pathway. In addition, Ang1 induced SEK1 phosphorylation at Ser80, suggesting the existence of an additional signal transduction pathway through which Ang1 attenuates JNK phosphorylation. These results demonstrated that Ang1 attenuates H2O2-induced SEK1/JNK phosphorylation through the PI 3-kinase/Akt pathway and inhibits the apoptosis of endothelial cells to oxidative stress.


Subject(s)
Angiopoietin-1/physiology , Endothelium, Vascular/enzymology , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/physiology , Mitogen-Activated Protein Kinase 8/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt , Signal Transduction/physiology , Swine
3.
Diabetes ; 53(9): 2443-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331557

ABSTRACT

Diabetic retinopathy is the leading cause of new blindness in adults in developed countries. Leptin, an adipocyte-derived hormone, stimulates endothelial proliferation and angiogenesis. This study was designed to elucidate the pathophysiologic role of leptin in the progression of retinal neovascularization. Using the retinopathy of prematurity model, a mouse model of ischemia-induced retinal neovascularization, we have demonstrated more pronounced retinal neovascularization in 17-day-old transgenic mice overexpressing leptin than in age-matched wild-type littermates. Ischemia-induced retinal neovascularization was markedly suppressed in 17-day-old leptin-deficient ob/ob mice. Western blot analysis revealed that a biologically active leptin receptor isoform is expressed in mouse retinal endothelial cells. Leptin receptor expression was also detected in primary cultures of porcine retinal endothelial cells, where it upregulated vascular endothelial growth factor (VEGF) mRNA expression. This effect was thought to be mediated at least partly through the activation of signal transducers and activators of transcription (STAT)3, because adenoviral transfection of the dominant-negative form of STAT3 abolished the leptin-induced upregulation of VEGF mRNA expression in retinal endothelial cells. This study provides evidence that leptin stimulates the ischemia-induced retinal neovasucularization possibly through the upregulation of endothelial VEGF, thereby suggesting that leptin antagonism may offer a novel therapeutic strategy to prevent or treat diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/physiopathology , Ischemia/physiopathology , Leptin/genetics , Retinal Neovascularization/physiopathology , Vascular Endothelial Growth Factor A/genetics , Animals , DNA-Binding Proteins/metabolism , Female , Gene Expression , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen/pharmacology , Pregnancy , RNA, Messenger/analysis , Receptors, Cell Surface/metabolism , Receptors, Leptin , Retinal Vessels/drug effects , Retinal Vessels/physiology , STAT3 Transcription Factor , Trans-Activators/metabolism , Up-Regulation
4.
Am J Pathol ; 164(5): 1827-35, 2004 May.
Article in English | MEDLINE | ID: mdl-15111329

ABSTRACT

Transcription factor Ets-1 has been reported to regulate angiogenesis in vascular endothelial cells. Here, we investigated a mechanism that may regulate the expression of Ets-1 in vascular endothelial growth factor (VEGF)- and hypoxia-induced retinal neovascularization and that may have potential to inhibit ocular neovascular diseases. VEGF and hypoxia increased Ets-1 expression in cultured bovine retinal endothelial cells. The VEGF-induced mRNA increase of Ets-1 was suppressed by a tyrosine kinase inhibitor (genistein), by inhibitors of MEK (mitogen-activated protein and extracellular signal-regulated kinase kinase) (PD98059 and UO126), and by inhibitors of protein kinase C (GF109203X, staurosporine, and Gö6976). Dominant-negative Ets-1 inhibited VEGF-induced cell proliferation, tube formation, and the expression of neuropilin-1 and angiopoietin-2. In a mouse model of proliferative retinopathy, Ets-1 mRNA was up-regulated. Intravitreal injection of dominant-negative Ets-1 suppressed retinal angiogenesis in a mouse model of proliferative retinopathy. In conclusion, VEGF induces Ets-1 expression in bovine retinal endothelial cells and its expression is protein kinase C/ERK pathway-dependent. Ets-1 up-regulation is involved in the development of retinal neovascularization, and inhibition of Ets-1 may be beneficial in the treatment of ischemic ocular diseases.


Subject(s)
Neovascularization, Pathologic , Proto-Oncogene Proteins/physiology , Reperfusion Injury , Retina/pathology , Transcription Factors/physiology , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae/genetics , Angiopoietin-2/biosynthesis , Animals , Blotting, Northern , Blotting, Western , Butadienes/pharmacology , Carbazoles/pharmacology , Cattle , Cell Division , DNA/chemistry , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genes, Dominant , Humans , Hypoxia , Indoles/pharmacology , Maleimides/pharmacology , Mice , Models, Biological , Neuropilin-1/biosynthesis , Nitriles/pharmacology , Phosphorylation , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , RNA, Messenger/metabolism , Staurosporine/pharmacology , Time Factors , Up-Regulation
5.
Jpn J Ophthalmol ; 46(3): 270-8, 2002.
Article in English | MEDLINE | ID: mdl-12063036

ABSTRACT

PURPOSE: It has been reported that vitronectin receptor-type integrins mediate vascular cell proliferation and migration. In this study, we investigated the expression of vitronectin receptor-type integrins and osteopontin in ischemia-induced retinal neovascularization, and examined the role of osteopontin in angiogenesis as a ligand of vitronectin receptor-type integrins. METHODS: Retinal neovascularization was produced by exposing C57BL/6J mice to 75% oxygen from postnatal day (P) 7 to P12. Expression of vitronectin receptor-type integrins and osteopontin was assessed by Northern blot analysis, in situ hybridization, and immunofluorescence. The role of osteopontin in retinal angiogenesis was evaluated by tube formation assay using cultured bovine retinal microcapillary endothelial cells. RESULTS: In the murine model, integrin alpha(v) mRNA was increased from P14 with a 2.6-fold peak response observed on P19, when retinal neovascularization was remarkable. Indirect immunofluorescence for vitronectin receptor-type integrins revealed prominent expression of integrin alpha(v)beta3/beta5 in the neovascular endothelial cells. Osteopontin mRNA was increased from P14, with a 2.0-fold peak response observed on P19. In situ hybridization demonstrated localization of osteopontin mRNA in neovascular tufts. Vascular endothelial growth factor-induced tube formation (8.3 +/- 0.6 mm/field) was inhibited significantly by treatment with anti-osteopontin antibody (4.8 +/- 0.7 mm/field, P <.001). CONCLUSIONS: These data suggest that increased expression of both vitronectin receptor-type integrins and osteopontin in ischemic retina contribute to vascular endothelial cell proliferation and to retinal vascular formation by promoting interaction between endothelial cells and extracellular matrix, which leads to retinal neovascularization.


Subject(s)
Ischemia/complications , Receptors, Vitronectin/metabolism , Retinal Neovascularization/metabolism , Sialoglycoproteins/metabolism , Animals , Animals, Newborn , Blotting, Northern , Cells, Cultured , Disease Models, Animal , Fluorescent Antibody Technique , In Situ Hybridization , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Osteopontin , RNA, Messenger/metabolism , Receptors, Vitronectin/genetics , Retinal Neovascularization/etiology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Sialoglycoproteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...