Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Diabetes Res Clin Pract ; 77 Suppl 1: S62-4, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17481772

ABSTRACT

Diabetic retinopathy is a leading cause of visual disturbance in adults. In proliferative diabetic retinopathy, ischemia-induced pathologic growth of new blood vessels often causes catastrophic loss of vision. Besides VEGF, the existence of another potent ischemia-induced angiogenic factor is postulated. Since ischemia-inducible erythropoietin (Epo) has recently been identified its angiogenic properties, we investigated its potential role during retinal angiogenesis in proliferative diabetic retinopathy (PDR). The vitreous Epo level in patients with PDR was significantly higher than that in nondiabetic patients. Multivariate logistic regression analyses indicated that Epo and VEGF were independently associated with PDR and that Epo was more strongly associated with PDR than VEGF. Blockade of Epo inhibits retinal neovascularization in vivo, and inhibits endothelial cell proliferation response to PDR vitreous in vitro. Our data provide strong evidence that erythropoietin is a potent retinal angiogenic factor independent of VEGF and is capable of stimulating ischemia-induced retinal angiogenesis in proliferative diabetic retinopathy. Inhibition of such molecular mechanisms in the retinal angiogenesis could be a new therapeutical strategy in halting or preventing pathologic angiogenesis in diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/physiopathology , Erythropoietin/physiology , Diabetic Retinopathy/pathology , Enzyme-Linked Immunosorbent Assay , Erythropoietin/genetics , Humans , Neovascularization, Pathologic/prevention & control , RNA, Messenger/genetics , Retinal Vessels/physiopathology , Vascular Diseases/physiopathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology
2.
Hypertension ; 49(2): 347-54, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17159082

ABSTRACT

Hypertension is known to exacerbate diabetic complications, such as retinopathy and nephropathy. Apoptosis of retinal vascular pericytes has been well established as the earliest conceivable change in diabetic retinopathy. In this study, we investigated the contribution of cyclic stretch, which mimics a hypertensive state to pericyte apoptosis. A 48-hour cyclic stretch induced DNA fragmentation in porcine retinal pericytes and increased the number of TUNEL+ cells at a pathophysiologically relevant extension level (10%/60 cycles per minute). Stretch also increased intracellular reactive oxygen species generation and increased c-Jun NH(2)-terminal kinase phosphorylation in a time- and magnitude-dependent manner, which were reduced by the nicotinamide-adenine dinucleotide phosphate oxidase inhibitor diphenylene iodonium or dominant-negative protein kinase C-delta. Stretch activated protein kinase C-delta and increased its association with p47phox. Stretch induced cleavage of caspase-9 and -3 and increased caspase-3 activity. Protein kinase C-delta or c-Jun NH(2)-terminal kinase inhibition normalized stretch-induced caspase-3 activity and prevented stretch-induced apoptosis. These data indicate that cyclic stretch induces apoptosis in porcine retinal pericytes by activation of the reactive oxygen species-c-Jun NH(2)-terminal kinase-caspase cascades, suggesting a novel molecular mechanism to explain the exacerbation of early diabetic retinopathy by concomitant hypertension.


Subject(s)
Apoptosis/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Pericytes/physiology , Reactive Oxygen Species/metabolism , Retina/physiology , Animals , Caspases/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , NADPH Oxidases/metabolism , Phosphorylation , Protein Kinase C-delta/metabolism , Retina/cytology , Stress, Mechanical , Swine , Time Factors
3.
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
4.
N Engl J Med ; 353(8): 782-92, 2005 Aug 25.
Article in English | MEDLINE | ID: mdl-16120858

ABSTRACT

BACKGROUND: Although vascular endothelial growth factor (VEGF) is a primary mediator of retinal angiogenesis, VEGF inhibition alone is insufficient to prevent retinal neovascularization. Hence, it is postulated that there are other potent ischemia-induced angiogenic factors. Erythropoietin possesses angiogenic activity, but its potential role in ocular angiogenesis is not established. METHODS: We measured both erythropoietin and VEGF levels in the vitreous fluid of 144 patients with the use of radioimmunoassay and enzyme-linked immunosorbent assay. Vitreous proliferative potential was measured according to the growth of retinal endothelial cells in vitro and with soluble erythropoietin receptor. In addition, a murine model of ischemia-induced retinal neovascularization was used to evaluate erythropoietin expression and regulation in vivo. RESULTS: The median vitreous erythropoietin level in 73 patients with proliferative diabetic retinopathy was significantly higher than that in 71 patients without diabetes (464.0 vs. 36.5 mIU per milliliter, P<0.001). The median VEGF level in patients with retinopathy was also significantly higher than that in patients without diabetes (345.0 vs. 3.9 pg per milliliter, P<0.001). Multivariate logistic-regression analyses indicated that erythropoietin and VEGF were independently associated with proliferative diabetic retinopathy and that erythropoietin was more strongly associated with the presence of proliferative diabetic retinopathy than was VEGF. Erythropoietin and VEGF gene-expression levels are up-regulated in the murine ischemic retina, and the blockade of erythropoietin inhibits retinal neovascularization in vivo and endothelial-cell proliferation in the vitreous of patients with diabetic retinopathy in vitro. CONCLUSIONS: Our data suggest that erythropoietin is a potent ischemia-induced angiogenic factor that acts independently of VEGF during retinal angiogenesis in proliferative diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/pathology , Erythropoietin/metabolism , Retina/cytology , Retinal Neovascularization/pathology , Vascular Endothelial Growth Factor A/metabolism , Vitreous Body/chemistry , Animals , Case-Control Studies , Cattle , Cell Proliferation , Cells, Cultured , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/physiopathology , Dose-Response Relationship, Drug , Erythropoietin/analysis , Erythropoietin/antagonists & inhibitors , Extracellular Matrix Proteins , Female , Humans , Logistic Models , Male , Mice , Middle Aged , Myosin Heavy Chains , Nonmuscle Myosin Type IIB , Proteins/pharmacology , RNA, Messenger/metabolism , Receptors, Erythropoietin/physiology , Retina/drug effects , Retina/metabolism , Retinal Neovascularization/physiopathology , Up-Regulation , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/antagonists & inhibitors
5.
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
6.
Am J Ophthalmol ; 139(3): 476-81, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15767056

ABSTRACT

PURPOSE: To investigate the levels of angiopoietin-2 (Ang2) and vascular endothelial growth factor (VEGF) in the vitreous fluids of patients with proliferative diabetic retinopathy (PDR) and to ascertain their involvement, if any, in angiogenesis of PDR. DESIGN: Retrospective case-control study. METHODS: Forty-one eyes of 41 patients with proliferative diabetic retinopathy and 18 eyes of 18 patients with nondiabetic ocular diseases (control group). Nondiabetic control eyes included 11 with idiopathic macular hole and 7 with idiopathic epiretinal membrane. Vitreous fluid samples were obtained at vitrectomy, and the levels of Ang2 and VEGF were measured by enzyme-linked immunosorbent assay. RESULTS: Vitreous level (mean +/- SD) of Ang2 was significantly higher in patients with PDR (1,753 +/- 3,213 pg/ml) than in control patients (112 +/- 113 pg/ml) (P < .0001). The vitreous concentration of VEGF was also significantly higher in patients with PDR (812 +/- 1,108 pg/ml) than in control patients (1.7 +/- 4.4 pg/ml) (P < .0001). Both Ang2 and VEGF levels in eyes with active PDR were significantly higher than in those with inactive PDR. The vitreous concentration of Ang2 correlated significantly with that of VEGF in eyes with proliferative diabetic retinopathy ([correlation coefficient] rho = 0.497, P = .001). CONCLUSIONS: These data demonstrate an increase of Ang2 in the vitreous fluid of patients with PDR and suggest an association of Ang2 and VEGF with angiogenic activity in PDR.


Subject(s)
Angiopoietin-2/metabolism , Diabetic Retinopathy/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vitreous Body/metabolism , Case-Control Studies , Diabetic Retinopathy/surgery , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Retinal Neovascularization/metabolism , Retrospective Studies , Vitrectomy
7.
J Neurosci ; 25(5): 1240-8, 2005 Feb 02.
Article in English | MEDLINE | ID: mdl-15689562

ABSTRACT

Insulin receptor substrates (Irs-proteins) integrate signals from the insulin and insulin-like growth factor-1 (IGF1) receptors with other processes to control cellular growth, function, and survival. Here, we show that Irs2 promoted the maturation and survival of photoreceptors in the murine retina immediately after birth. Irs2 was mainly localized to the outer plexiform layer as well as to photoreceptor inner segments. It was also seen in ganglion cells and inner plexiform layer but in smaller amounts. Compared with control littermates, Irs2 knock-out mice lose 10% of their photoreceptors 1 week after birth and up to 50% by 2 weeks of age as a result of increased apoptosis. The surviving photoreceptor cells developed short organized segments, which displayed proportionally diminished but otherwise normal electrical function. However, IGF1-stimulated Akt phosphorylation was barely detected, and cleaved/activated caspase-3 was significantly elevated in isolated retinas of Irs2-/- mice. When diabetes was prevented, which allowed the Irs2-/- mice to survive for 2 years, most photoreceptor cells were lost by 16 months of age. Because apoptosis is the final common pathway in photoreceptor degeneration, pharmacological strategies that increase Irs2 expression or function in photoreceptor cells could be a general treatment for blinding diseases such as retinitis pigmentosa.


Subject(s)
Eye Proteins/physiology , Phosphoproteins/physiology , Photoreceptor Cells/cytology , Age Factors , Animals , Animals, Newborn , Apoptosis , Cell Survival , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/physiopathology , Eye Proteins/genetics , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Hyperglycemia/genetics , Hyperinsulinism/genetics , Insulin Receptor Substrate Proteins , Insulin Resistance , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphorylation , Photic Stimulation , Photoreceptor Cells/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Retinal Ganglion Cells/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/physiology
8.
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
9.
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
10.
Circ Res ; 94(6): 785-93, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-14963002

ABSTRACT

Angiotensin II (Ang II) plays essential roles in vascular homeostasis, neointimal formation, and postinfarct remodeling. Although Ang II has been shown to regulate apoptosis in cardiomyocytes and vascular smooth muscle cells, its role in vascular endothelial cells (ECs) remains elusive. To address this issue, we first performed TUNEL and caspase-3 activity assays with porcine microvascular ECs challenged by serum deprivation. Ang II significantly reduced the ratio of apoptotic cells and caspase-3 activity. The Ang II type 1 receptor (AT1) was responsible for these effects. Among the signaling molecules downstream of AT1, we revealed that PI3-kinase/Akt pathway plays a predominant role in the antiapoptotic effect of Ang II. Interestingly, the expression of survivin, a central molecule of cell survival, increased after Ang II stimulation. Overexpression of a dominant-negative form of Akt abolished both Ang II-induced antiapoptosis and survivin protein expression. In a murine model of hyperoxygen-induced retinal vascular regression, AT1a knockout mice showed a significant increase in retinal avascular areas. Our data indicate that Ang II plays a critical antiapoptotic role in vascular ECs by a mechanism involving PI3-kinase/Akt activation, subsequent upregulation of survivin, and suppression of caspase-3 activity.


Subject(s)
Angiotensin II/physiology , Apoptosis/drug effects , Capillaries/cytology , Caspase Inhibitors , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Microtubule-Associated Proteins/biosynthesis , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Receptor, Angiotensin, Type 1/physiology , Animals , Apoptosis/physiology , Caspase 3 , Culture Media, Serum-Free/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genes, Dominant , Humans , In Situ Nick-End Labeling , Inhibitor of Apoptosis Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Neoplasm Proteins , Oxygen/toxicity , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/genetics , Retinal Vessels/cytology , Signal Transduction/drug effects , Survivin , Swine
11.
J Biol Chem ; 278(34): 31964-71, 2003 Aug 22.
Article in English | MEDLINE | ID: mdl-12775712

ABSTRACT

The effects of insulin on vascular endothelial growth factor (VEGF) expression in cultured vascular cells and in angiogenesis were characterized. Insulin increased VEGF mRNA levels in mouse aortic smooth muscle cells from 10(-9) to 10(-7) m with an initial peak of 3.7-fold increases at 1 h and a second peak of 2.8-fold after 12 h. The first peak of VEGF expression was inhibited by LY294002, an inhibitor of phosphatidylinositol (PI) 3-kinase, and by the overexpression of dominant negative forms of p85 subunit of PI 3-kinase or Akt. Inhibitors of MEK kinase, PD98059, or overexpression of dominant negative forms of Ras was ineffective. In contrast, the chronic effect of insulin on VEGF expression was partially inhibited by both LY294002 or PD98059 as well as by the overexpression of dominant negatives of PI 3-kinase or Ras. The importance of PI 3-kinase-Akt pathway on VEGF expression was confirmed in mouse aortic smooth muscle cells isolated from insulin receptor substrate -1 knockout (IRS-1-/-) mice that showed parallel reductions of 46-49% in insulin-stimulated VEGF expression and PI 3-kinase-Akt activation. Insulin-induced activation of PI 3-kinase-Akt on hypoxia-induced VEGF expression and neovascularization was reduced by 40% in the retina of neonatal hypoxia model using IRS-1-/- mice. Thus, unlike other cells, insulin can regulate VEGF expression by both IRS-1/PI 3-kinase-Akt cascade and Ras-MAPK pathways in aortic smooth muscle cells. The in vivo results provide direct evidence that insulin can modulate hypoxia-induced angiogenesis via reduction in VEGF expression in vivo.


Subject(s)
Endothelial Growth Factors/genetics , Gene Expression Regulation/physiology , Insulin/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Neovascularization, Physiologic , Retinal Vessels/metabolism , Signal Transduction , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Insulin/physiology , Insulin Receptor Substrate Proteins , Mice , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/genetics , Phosphoproteins/physiology , Precipitin Tests , Promoter Regions, Genetic , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
FASEB J ; 17(1): 76-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12475915

ABSTRACT

Diabetic macular edema, resulting from increased microvascular permeability, is the most prevalent cause of vision loss in diabetes. The mechanisms underlying this complication remain poorly understood. In the current study, diabetic vascular permeability (blood-retinal barrier breakdown) is demonstrated to result from a leukocyte-mediated Fas-FasL-dependent apoptosis of the retinal vasculature. Following the onset of streptozotocin-induced diabetes, FasL expression was increased in rat neutrophils (P<0.005) and was accompanied by a simultaneous increase in Fas expression in the retinal vasculature. Static adhesion assays demonstrated that neutrophils from diabetic, but not control, rats induced endothelial cell apoptosis in vitro (P<0.005). The latter was inhibited via an antibody-based FasL blockade (P<0.005). In vivo, the inhibition of FasL potently reduced retinal vascular endothelial cell injury, apoptosis, and blood-retinal barrier breakdown (P<0.0001) but did not diminish leukocyte adhesion to the diabetic retinal vasculature. Taken together, these data are the first to identify leukocyte-mediated Fas-FasL-dependent retinal endothelial cell apoptosis as a major cause of blood-retinal barrier breakdown in early diabetes. These data imply that the targeting of the Fas-FasL pathway may prove beneficial in the treatment of diabetic retinopathy.


Subject(s)
Apoptosis , Blood-Retinal Barrier , Diabetes Mellitus, Experimental/immunology , Diabetic Retinopathy/immunology , Membrane Glycoproteins/antagonists & inhibitors , fas Receptor/physiology , Animals , Cell Adhesion , Cell Death , Coculture Techniques , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Endothelium, Vascular/cytology , Fas Ligand Protein , Leukocytes/immunology , Membrane Glycoproteins/physiology , Models, Immunological , Neutrophils/immunology , Rats , Retina/cytology
13.
Retina ; 22(4): 418-22, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12172107

ABSTRACT

PURPOSE: To investigate the expression of estrogen receptor in choroidal neovascular membranes (CNVMs) surgically excised from eyes with high myopia. METHODS: The CNVMs were surgically excised from two eyes with high myopia. Immunohistochemical analysis with a monoclonal antibody to estrogen receptor and in situ hybridization with an oligodeoxynucleotide sequence coding for estrogen receptor were used to study the cellular distribution of estrogen receptor and its messenger RNA in the CNVMs. Immunohistochemical localization of glial fibrillary acidic protein and vimentin in the CNVMs was compared with localization of estrogen receptor. RESULTS: Immunohistochemical analysis with monoclonal antibody to estrogen receptor showed widespread staining throughout the CNVMs. By in situ hybridization, the expression of estrogen receptor messenger RNA was predominantly observed in the CNVMs. Staining with antibody to vimentin was widespread throughout the CNVMs, which was similar to the localization of estrogen receptor. CONCLUSION: Estrogen receptor was expressed in the CNVMs in highly myopic eyes, suggesting that estrogen may have important functions in the formation of CNVMs.


Subject(s)
Choroidal Neovascularization/metabolism , Myopia/metabolism , Receptors, Estrogen/metabolism , Aged , Antibodies, Monoclonal , Choroidal Neovascularization/etiology , Choroidal Neovascularization/surgery , Female , Fluorescein Angiography , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunoenzyme Techniques , In Situ Hybridization , Myopia/complications , Myopia/surgery , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Vimentin/metabolism
14.
Invest Ophthalmol Vis Sci ; 43(6): 2007-14, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12037012

ABSTRACT

PURPOSE: It has been reported that 17beta-estradiol (E2) may enhance the proliferation of bovine retinal vascular endothelial cells (BRECs) by increasing the expression of VEGFR-2 and VEGF. The hypothesis in the current study was that estrogen may contribute to fetal vascular development and the cessation of exposure to estrogen of premature infants on birth may have an inhibitory effect on retinopathy of prematurity (ROP). Because ROP is thought to develop under relative hypoxia after exposure to high-dose oxygen, this study was conducted to investigate how estrogen modulates hypoxia-induced VEGF in BRECs and mouse ROP. METHODS: Gene expression of VEGF and hypoxia-inducible factor (HIF)-1alpha were studied in BRECs, with or without E2, under normoxia and hypoxia (1% O2). A binding assay was performed to determine whether estrogen interferes with HIF-1-mediated induction of VEGF. In a mouse ROP model, effects of E2 were evaluated by avascular area, subsequent extraretinal neovascularization, and retinal expression of the VEGF gene, by administering E2 during hyperoxia (75% O2) and/or after exposure to room air. RESULTS: Hypoxia-induced VEGF mRNA in BRECs was reduced dose dependently by 1 to 100 nM E2. E2 reduced hypoxia-induced binding of HIF-1 to the VEGF promoter site and reduced the HIF-1alpha mRNA level. In mouse ROP, injection of E2 during hyperoxia increased retinal VEGF mRNA and reduced the retinal avascular area at the end of hyperoxia. E2 treatment during the normoxia that followed reduced VEGF mRNA and extraretinal neovascularization. Treatment with E2 throughout both periods significantly improved retinopathy. CONCLUSIONS: Estrogen may function as a significant modulator of the level of VEGF mRNA under different oxygen conditions and could serve as a prophylactic agent for ROP.


Subject(s)
Endothelial Growth Factors/genetics , Endothelium, Vascular/drug effects , Estrogens/pharmacology , Hypoxia/metabolism , Lymphokines/genetics , Retinal Neovascularization/metabolism , Retinal Vessels/drug effects , Retinopathy of Prematurity/metabolism , Transcription Factors , Animals , Blotting, Northern , Cattle , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gene Expression Regulation/drug effects , Humans , Hypoxia/pathology , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Infant, Newborn , Lymphokines/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinopathy of Prematurity/pathology , Tamoxifen/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
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
16.
Circulation ; 105(3): 373-9, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-11804995

ABSTRACT

BACKGROUND: Inadequate angiogenic response to ischemia in the myocardium of diabetic patients could result in poor collateral formation. Yet, excessive neovascularization in the retina causes proliferative diabetic retinopathy. Since vascular endothelial growth factor (VEGF) is the major angiogenic factor expressed in response to hypoxia, we have characterized expression of VEGF and its receptors in retina, renal glomeruli, aorta, and myocardium in insulin-resistant and diabetic states. Methods and Results- The expression of mRNA and protein for VEGF and its receptors, VEGF-R1 and VEGF-R2, in the myocardium was decreased significantly by 40% to 70% in both diabetic and insulin-resistant nondiabetic rats. Twofold reductions in VEGF and VEGF-R2 were observed in ventricles from diabetic patients compared with nondiabetic donors. In contrast, expression of VEGF and its receptors were increased 2-fold in retina and glomeruli from diabetic or insulin-resistant rats. Insulin treatment of diabetic rats normalized changes in both cardiac and microvascular tissues. Insulin increased VEGF mRNA expression in cultured rat neonatal cardiac myocytes. CONCLUSIONS: The results documented for the first time that differential regulation of VEGF and its receptors exist between microvascular and cardiac tissues, which can be regulated by insulin. These results provide a potential explanation for concomitant capillary leakage and neovascularization in the retina and inadequate collateral formation in the myocardium of insulin-resistant and diabetic patients.


Subject(s)
Diabetes Mellitus/metabolism , Endothelial Growth Factors/metabolism , Insulin Resistance , Lymphokines/metabolism , Myocardium/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Animals , Cells, Cultured , Collateral Circulation , Coronary Circulation , Diabetes Mellitus/blood , Endothelial Growth Factors/genetics , Female , Gene Expression Regulation/drug effects , Humans , Insulin/pharmacology , Kidney Glomerulus/metabolism , Lymphokines/genetics , Male , Middle Aged , Neovascularization, Pathologic , Obesity , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Retina/metabolism , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Proc Natl Acad Sci U S A ; 99(2): 721-6, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-11805327

ABSTRACT

Retinal neovascularization is a major cause of blindness and requires the activities of several signaling pathways and multiple cytokines. Activation of protein kinase C (PKC) enhances the angiogenic process and is involved in the signaling of vascular endothelial growth factor (VEGF). We have demonstrated a dramatic increase in the angiogenic response to oxygen-induced retinal ischemia in transgenic mice overexpressing PKC beta 2 isoform and a significant decrease in retinal neovascularization in PKC beta isoform null mice. The mitogenic action of VEGF, a potent hypoxia-induced angiogenic factor, was increased by 2-fold in retinal endothelial cells by the overexpression of PKC beta 1 or beta 2 isoforms and inhibited significantly by the overexpression of a dominant-negative PKC beta 2 isoform but not by the expression of PKC alpha, delta, and zeta isoforms. Association of PKC beta 2 isoform with retinoblastoma protein was discovered in retinal endothelial cells, and PKC beta 2 isoform increased retinoblastoma phosphorylation under basal and VEGF-stimulated conditions. The potential functional consequences of PKC beta-induced retinoblastoma phosphorylation could include enhanced E2 promoter binding factor transcriptional activity and increased VEGF-induced endothelial cell proliferation.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Endothelial Growth Factors/physiology , Isoenzymes/physiology , Lymphokines/physiology , Neovascularization, Pathologic , Protein Kinase C/physiology , Retinal Vessels/growth & development , Retinoblastoma Protein/metabolism , Animals , Cattle , Cell Division , Cells, Cultured , E2F Transcription Factors , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Isoenzymes/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C beta , Retinal Vessels/cytology , Retinal Vessels/physiology , Signal Transduction , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
J Biol Chem ; 277(2): 1047-57, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11694503

ABSTRACT

Stretch-induced expression of vascular endothelial growth factor (VEGF) is thought to be important in mediating the exacerbation of diabetic retinopathy by systemic hypertension. However, the mechanisms underlying stretch-induced VEGF expression are not fully understood. We present novel findings demonstrating that stretch-induced VEGF expression in retinal capillary pericytes is mediated by phosphatidylinositol (PI) 3-kinase and protein kinase C (PKC)-zeta but is not mediated by ERK1/2, classical/novel isoforms of PKC, Akt, or Ras despite their activation by stretch. Cardiac profile cyclic stretch at 60 cpm increased VEGF mRNA expression in a time- and magnitude-dependent manner without altering mRNA stability. Stretch increased ERK1/2 phosphorylation, PI 3-kinase activity, Akt phosphorylation, and PKC-zeta activity. Signaling pathways were explored using inhibitors of PKC, MEK1/2, and PI 3-kinase; adenovirus-mediated overexpression of ERK, PKC-alpha, PKC-delta, PKC-zeta, and Akt; and dominant negative (DN) mutants of ERK, PKC-zeta, Ras, PI 3-kinase and Akt. Although stretch activated ERK1/2 through a Ras- and PKC classical/novel isoform-dependent pathway, these pathways were not responsible for stretch-induced VEGF expression. Overexpression of DN ERK and Ras had no effect on VEGF expression in these cells. In contrast, DN PI 3-kinase as well as pharmacologic inhibitors of PI 3-kinase blocked stretch-induced VEGF expression. Although stretch-induced PI 3-kinase activation increased both Akt phosphorylation and activity of PKC-zeta, VEGF expression was dependent on PKC-zeta but not Akt. In addition, PKC-zeta did not mediate stretch-induced ERK1/2 activation. These results suggest that stretch-induced expression of VEGF involves a novel mechanism dependent upon PI 3-kinase-mediated activation of PKC-zeta that is independent of stretch-induced activation of ERK1/2, classical/novel PKC isoforms, Ras, or Akt. This mechanism may play a role in the well documented association of concomitant hypertension with clinical exacerbation of neovascularization and vascular permeability.


Subject(s)
Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Retina/metabolism , Animals , Cattle , Cells, Cultured , Endothelial Growth Factors/genetics , Enzyme Inhibitors/pharmacology , Lymphokines/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Isoforms/metabolism , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , RNA Stability , Receptors, Platelet-Derived Growth Factor/metabolism , Retina/chemistry , Retina/cytology , Retina/drug effects , Stress, Mechanical , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , ras Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...