Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
FEBS Lett ; 579(5): 1227-34, 2005 Feb 14.
Article in English | MEDLINE | ID: mdl-15710418

ABSTRACT

Vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) are key angiogenic stimulators during normal development and wound healing, as well as in a variety of pathological conditions. Recent studies have demonstrated a synergistic effect of VEGF and PlGF in pathological angiogenesis and suggest a role for PlGF in amplifying VEGF action in endothelial cells. We show here in the mouse model of oxygen-induced retinopathy that VEGF is significantly increased (P<0.01) in the retina at both the mRNA and protein levels. In this mouse model, PlGF was significantly upregulated in the retina at the protein level (P<0.01) without a corresponding change in mRNA levels. In cultured human retinal and umbilical vein endothelial cells, VEGF induced the production of PlGF protein by over 10-fold (P<0.01) in a dose-dependent manner through a post-transcriptional mechanism. The increased PlGF expression upon VEGF treatment was significantly reduced by inhibition of the protein kinase C (PKC) and MEK signaling pathways, as well as by treatment with the calcium ionophore A23187. Taken together, our findings demonstrate that VEGF can amplify its effects on endothelial cells by inducing the production of PlGF via a post-transcriptional mechanism in a PKC-dependent manner, and provide a potential link between PKC inhibition and amelioration of vascular complications in the development of angiogenic diseases.


Subject(s)
Pregnancy Proteins/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Oxygen/pharmacology , Phorbol Esters/pharmacology , Placenta Growth Factor , Pregnancy Proteins/biosynthesis , Pregnancy Proteins/genetics , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Am J Ophthalmol ; 137(4): 668-74, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15059706

ABSTRACT

PURPOSE: Pigment epithelium-derived factor (PEDF) has been demonstrated to suppress ocular angiogenesis in several animal models. In this study, we sought to measure the levels of PEDF and vascular endothelial growth factor (VEGF) in the vitreous of patients with and without ocular neovascular disorders. DESIGN: Case-control study of patients undergoing intraocular surgery for a variety of neovascular and nonneovascular conditions. METHODS: Vitreous samples were collected from 65 eyes of 65 patients with no neovascular disorder (n = 24), choroidal neovascularization (n = 9), active proliferative diabetic retinopathy (n = 16), and inactive proliferative diabetic retinopathy (n = 16). The levels of VEGF and PEDF in these vitreous samples were determined by enzyme-linked immunosorbent assay. RESULTS: The VEGF levels were at or below the level of detectability in the reference and choroidal neovascularization groups. The VEGF levels were significantly elevated in both the active and inactive PDR groups, and significantly higher in the active PDR group as compared with the inactive PDR group. The PEDF levels, which were present at relatively high concentrations in all groups, were higher in patients with active PDR compared with the control and choroidal neovascularization groups. CONCLUSIONS: High levels of immunoreactive PEDF are present in the vitreous of individuals with or without ocular neovascularization, but PEDF levels are significantly higher in patients with active PDR compared with patients with choroidal neovascularization or nonneovascular retinal diseases. Although these results do not preclude the possibility that endogenous PEDF helps to modulate ocular neovascularization, they do not support ischemia-induced downregulation of PEDF as a mechanism for such modulation.


Subject(s)
Choroidal Neovascularization/metabolism , Nerve Growth Factors , Proteins/metabolism , Serpins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vitreous Body/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Case-Control Studies , Choroidal Neovascularization/diagnosis , Diabetic Retinopathy/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Female , Fluorescein Angiography , Humans , Male , Middle Aged
3.
Hum Gene Ther ; 14(2): 129-41, 2003 Jan 20.
Article in English | MEDLINE | ID: mdl-12614564

ABSTRACT

Vascular endothelial growth factor (VEGF) is a critical stimulus for both retinal and choroidal neovascularization, and for diabetic macular edema. We used mouse models for these diseases to explore the potential of gene transfer of soluble VEGF receptor-1 (sFlt-1) as a treatment. Intravitreous or periocular injection of an adenoviral vector encoding sFlt-1 (AdsFlt-1.10) markedly suppressed choroidal neovascularization at rupture sites in Bruch's membrane. Periocular injection of AdsFlt-1.10 also caused significant reduction in VEGF-induced breakdown of the blood-retinal barrier, but failed to significantly inhibit ischemia-induced retinal neovascularization. Periocular delivery of an adenoviral vector encoding pigment epithelium-derived factor (PEDF), another secreted protein, resulted in high levels of PEDF in the retinal pigmented epithelium and choroid, but not in the retina. This may explain why periocular injection of AdsFlt-1.10 inhibited choroidal, but not retinal neovascularization. Periocular delivery offers potential advantages over other routes of delivery and the demonstration that sFlt-1 enters the eye from the periocular space in sufficient levels to achieve efficacy in treating choroidal neovascularization and retinal vascular permeability is a novel finding that has important clinical implications. These data suggest that periocular gene transfer of sFlt-1 should be considered for treatment of choroidal neovascularization and diabetic macular edema.


Subject(s)
Blood-Retinal Barrier/physiology , Endothelial Growth Factors/metabolism , Extracellular Matrix Proteins/genetics , Eye/blood supply , Gene Transfer Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Neovascularization, Pathologic/prevention & control , Adenoviridae , Animals , Extracellular Matrix Proteins/biosynthesis , Eye/metabolism , Genes, Reporter , Genetic Vectors , Mice , Myosin Heavy Chains , Nonmuscle Myosin Type IIB , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Invest Ophthalmol Vis Sci ; 43(3): 821-9, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11867604

ABSTRACT

PURPOSE: To determine the effect of pigment epithelium-derived factor (PEDF) in a mouse model of ischemia-induced retinal neovascularization and on vascular endothelial growth factor (VEGF)--induced migration and growth of cultured microvascular endothelial cells. METHODS: Human recombinant PEDF was expressed in the human embryonic kidney 293 cell line and purified by ammonium sulfate precipitation and cation exchange chromatography. C57BL/6 mice were exposed to 75% oxygen from postnatal day (P)7 to P12 and then returned to room air. Mice received intravitreal injections of 2 microg PEDF in one eye and vehicle in the contralateral eye on P12 and P14. At P17, mice were killed and eyes enucleated for quantitation of retinal neovascularization. The mitogenic and motogeneic effects of VEGF on cultured bovine retinal and adrenal capillary endothelial cells were examined in the presence or absence of PEDF, using cell counts and migration assays. RESULTS: Two species of human recombinant PEDF, denoted A and B, were purified to apparent homogeneity. PEDF B appeared to comigrate on SDS-PAGE with PEDF from human vitreous samples. Changes in electrophoretic mobility after peptide-N-glycosidase F (PNGase F) digestion suggest that both PEDF forms contain N-linked carbohydrate. Analyses of the intact proteins by liquid chromatography-electrospray mass spectrometry (LC-ESMS) revealed the major molecular weight species for PEDF A (47,705 +/- 4) and B (46,757 +/- 5). LC-ESMS analysis of tryptic peptides indicated that PEDF A and B exhibit differences in glycopeptides containing N-acetylneuraminic acid (NeuAc) and N-acetylhexosamine (HexNAc). Intravitreal administration of either species of PEDF significantly inhibited retinal neovascularization (83% for PEDF A and 55% for PEDF B; P = 0.024 and 0.0026, respectively). PEDF A and B (20 nM) suppressed VEGF-induced retinal microvascular endothelial cell proliferation by 48.8% and 41.4%, respectively, after 5 days (P < 0.001) and VEGF-induced migration by 86.5% +/- 16.7% and 78.1% +/- 22.3%, respectively, after 4 hours (P = 0.004 and P = 0.008, respectively). CONCLUSIONS: These data indicate that elevated concentrations of PEDF inhibit VEGF-induced retinal endothelial cell growth and migration and retinal neovascularization. These findings suggest that localized administration of PEDF may be an effective approach for the treatment of ischemia-induced retinal neovascular disorders.


Subject(s)
Cell Division/drug effects , Cell Movement/drug effects , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/pathology , Eye Proteins , Lymphokines/pharmacology , Nerve Growth Factors , Proteins/therapeutic use , Reperfusion Injury/drug therapy , Retinal Neovascularization/drug therapy , Serpins/therapeutic use , Animals , Blotting, Western , Cell Count , Cells, Cultured , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Proteins/genetics , Proteins/isolation & purification , Recombinant Proteins , Reperfusion Injury/complications , Reperfusion Injury/pathology , Retinal Neovascularization/etiology , Retinal Neovascularization/pathology , Retinal Vessels/drug effects , Retinal Vessels/pathology , Serpins/genetics , Serpins/isolation & purification , Spectrometry, Mass, Electrospray Ionization , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...