Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Br J Ophthalmol ; 93(6): 765-70, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19218250

ABSTRACT

AIMS: To evaluate the efficacy of a single posterior sub-Tenon capsule injection of triamcinolone acetonide (PSTA) before panretinal photocoagulation (PRP). METHODS: This 6-month study involved the randomisation of 82 eyes of 41 patients, with bilateral severe non-proliferative diabetic retinopathy or proliferative diabetic retinopathy to a single PSTA 20 mg or to no injection before PRP. The primary end-point was change in best-corrected visual acuity (BCVA) at 6 months compared with that at baseline using the logarithm of the minimum angle of resolution (logMAR). Secondary end-points were changes in retinal thickness and intraocular pressure. RESULTS: The mean changes in logMAR BCVA at 6 months compared with that at baseline were a worsening of 0.010 (SD 0.029) in the control group (no injection) and an improvement of 0.072 (0.028) in the PSTA group (p = 0.04). The mean changes in foveal thickness at 6 months compared with baseline measurements were an increase of 32.8 (82.8) mum in the control group and a lessening of 9.7 (85.6) mum in the PSTA group (p = 0.03). CONCLUSIONS: PSTA before PRP appears to be beneficial in preventing PRP-induced visual loss in eyes with diabetic retinopathy by reducing the chance of macular thickening.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetic Retinopathy/drug therapy , Laser Coagulation/methods , Triamcinolone Acetonide/therapeutic use , Adult , Aged , Chemotherapy, Adjuvant , Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , Diabetic Retinopathy/surgery , Female , Follow-Up Studies , Fovea Centralis/pathology , Glucocorticoids/therapeutic use , Humans , Intraocular Pressure/drug effects , Laser Coagulation/adverse effects , Macular Edema/etiology , Macular Edema/pathology , Macular Edema/prevention & control , Male , Middle Aged , Visual Acuity/drug effects , Young Adult
2.
J Biol Chem ; 276(33): 31099-104, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11390407

ABSTRACT

It is known well that activation of the hexosamine pathway causes insulin resistance, but how this activation influences pancreatic beta-cell function remains unclear. In this study, we found that in isolated rat islets adenovirus-mediated overexpression of glutamine:fructose-6-phosphate amidotransferase (GFAT), the first and rate-limiting enzyme of the hexosamine pathway, leads to deterioration of beta-cell function, which is similar to that found in diabetes. Overexpression of GFAT or treatment with glucosamine results in impaired glucose-stimulated insulin secretion and reduction in the expression levels of several beta-cell specific genes (insulin, GLUT2, and glucokinase). Additionally, the DNA binding activity of PDX-1, an important transcription factor for these three genes, was markedly reduced. These phenomena were not mimicked by the induction of O-linked glycosylation with an inhibitor of O-GlcNAcase, PUGNAc. It was also found that glucosamine increases hydrogen peroxide levels and that several hexosamine pathway-mediated changes were suppressed by treatment with the antioxidant N-acetyl-l-cysteine. In conclusion, activation of the hexosamine pathway leads to deterioration of beta-cell function through the induction of oxidative stress rather than O-linked glycosylation. Thus, the hexosamine pathway may contribute to the deterioration of beta-cell function found in diabetes.


Subject(s)
Diabetes Mellitus/physiopathology , Hexosamines/metabolism , Islets of Langerhans/physiopathology , Oxidative Stress , Animals , Cells, Cultured , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/biosynthesis , Glycosylation , Hyperglycemia/physiopathology , Male , Rats , Rats, Sprague-Dawley
3.
Diabetes ; 50(2): 444-54, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272159

ABSTRACT

Systemic hypertension exacerbates diabetic retinopathy and other coexisting ocular disorders through mechanisms that remain largely unknown. Increased vascular permeability and intraocular neovascularization characterize these conditions and are complications primarily mediated by vascular endothelial growth factor (VEGF). Because systemic hypertension increases vascular stretch, we evaluated the expression of VEGF, VEGF-R2 (kinase insert domain-containing receptor [KDR]), and VEGF-R1 (fms-like tyrosine kinase [Flt]) in bovine retinal endothelial cells (BRECs) undergoing clinically relevant cyclic stretch and in spontaneously hypertensive rat (SHR) retina. A single exposure to 20% symmetric static stretch increased KDR mRNA expression 3.9 +/- 1.1-fold after 3 h (P = 0.002), with a gradual return to baseline within 9 h. In contrast, BRECs exposed to cardiac-profile cyclic stretch at 60 cpm continuously accumulated KDR mRNA in a transcriptionally mediated, time-dependent and stretch-magnitude-dependent manner. Exposure to 9% cyclic stretch increased KDR mRNA expression 8.7 +/- 2.9-fold (P = 0.011) after 9 h and KDR protein concentration 1.8 +/- 0.3-fold (P = 0.005) after 12 h. Stretched-induced VEGF responses were similar. Scatchard binding analysis demonstrated a 180 +/- 40% (P = 0.032) increase in high-affinity VEGF receptor number with no change in affinity. Cyclic stretch increased basal thymidine uptake 60 +/- 10% (P < 0.001) and VEGF-stimulated thymidine uptake by 2.6 +/- 0.2-fold (P = 0.005). VEGF-NAb reduced cyclic stretch-induced thymidine uptake by 65%. Stretched-induced KDR expression was not inhibited by AT1 receptor blockade using candesartan. Hypertension increased retinal KDR expression 67 +/- 42% (P < 0.05) in SHR rats compared with normotensive WKY control animals. When hypertension was reduced using captopril or candesartan, retinal KDR expression returned to baseline levels. VEGF reacted similarly, but Flt expression did not change. These data suggest a novel molecular mechanism that would account for the exacerbation of diabetic retinopathy by concomitant hypertension, and may partially explain the principal clinical manifestations of hypertensive retinopathy itself. Furthermore, these data imply that anti-VEGF therapies may prove therapeutically effective for hypertensive retinopathy and/or ameliorating the deleterious effects of coexistent hypertension on VEGF-associated disorders such as diabetic retinopathy.


Subject(s)
Hypertension/physiopathology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Retinal Vessels/metabolism , Animals , Antihypertensive Agents/pharmacology , Benzimidazoles/pharmacology , Biphenyl Compounds , Blood Pressure/drug effects , Captopril/pharmacology , Cattle , Cells, Cultured , Diabetic Angiopathies/complications , Diabetic Angiopathies/physiopathology , Diabetic Retinopathy/complications , Diabetic Retinopathy/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Hypertension/complications , Hypertension/drug therapy , Hypertension/pathology , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Retinal Vessels/pathology , Stress, Mechanical , Tetrazoles/pharmacology
4.
J Biol Chem ; 275(52): 40725-31, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11018037

ABSTRACT

Fibroblastic proliferation accompanies many angiogenesis-related retinal and systemic diseases. Since connective tissue growth factor (CTGF) is a potent mitogen for fibrosis, extracellular matrix production, and angiogenesis, we have studied the effects and mechanism by which vascular endothelial growth factor (VEGF) regulates CTGF gene expression in retinal capillary cells. In our study, VEGF increased CTGF mRNA levels in a time- and concentration-dependent manner in bovine retinal endothelial cells and pericytes, without the need of new protein synthesis and without altering mRNA stability. VEGF activated the tyrosine receptor phosphorylation of KDR and Flt1 and increased the binding of phosphatidylinositol 3-kinase (PI3-kinase) p85 subunit to KDR and Flt1, both of which could mediate CTGF gene induction. VEGF-induced CTGF expression was mediated primarily by PI3-kinase activation, whereas PKC and ERK pathways made only minimal contributions. Furthermore, overexpression of constitutive active Akt was sufficient to induce CTGF gene expression, and inhibition of Akt activation by overexpressing dominant negative mutant of Akt abolished the VEGF-induced CTGF expression. These data suggest that VEGF can increase CTGF gene expression in bovine retinal capillary cells via KDR or Flt receptors and the activation of PI3-kinase-Akt pathway independently of PKC or Ras-ERK pathway, possibly inducing the fibrosis observed in retinal neovascular diseases.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Growth Substances/biosynthesis , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins , Lymphokines/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Retina/metabolism , Animals , Cattle , Cells, Cultured , Connective Tissue Growth Factor , Cycloheximide/pharmacology , Growth Substances/genetics , Immediate-Early Proteins/genetics , Isoenzymes/physiology , Phosphorylation , Protein Kinase C/physiology , Proto-Oncogene Proteins c-akt , RNA, Messenger/analysis , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
5.
Diabetes ; 49(7): 1239-48, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10909984

ABSTRACT

Enhanced actions or levels of endothelin-1 (ET-1), a potent vasoconstrictor, have been associated with decreased blood flow in the retina and peripheral nerves of diabetic animals and may be related to the development of pathologies in these tissues. Hyperglycemia has been postulated to increase ET-1 secretion in endothelial cells. We have characterized the mechanism by which elevation of glucose is increasing ET-1 mRNA expression in capillary bovine retinal endothelial cells (BREC) and bovine retinal pericytes (BRPC). Elevation of glucose, but not mannitol, from 5.5 to 25 mmol/l for 3 days increased membranous protein kinase C (PKC) activities and ET-1 mRNA in parallel levels by 2-fold in BREC and BRPC. These effects were reversed by decreasing glucose levels to 5.5 mmol/l for an additional 2 days. Glucose-induced ET-1 overexpression was inhibited by a general PKC inhibitor, GF109203X, and a mitogen-activated protein kinase kinase inhibitor, PD98059, but not by wortmannin, a phosphatidylinositol 3-kinase inhibitor. By immunoblot analysis, PKC-beta2 and -delta isoforms in BREC were significantly increased relative to other isoforms in the membranous fractions when glucose level was increased. Overexpression of PKC-beta1 and -delta isoforms but not PKC-zeta isoform by adenovirus vectors containing the respective cDNA enhanced in parallel PKC activities, proteins, and basal and glucose-induced ET-1 mRNA expression by at least 2-fold. These results showed that enhanced ET-1 expression induced by hyperglycemia in diabetes is partly due to activation of PKC-beta and -delta isoforms, suggesting that inhibition of these PKC isoforms may prevent early changes in diabetic retinopathy and neuropathy.


Subject(s)
Endothelin-1/genetics , Endothelium, Vascular/physiology , Glucose/pharmacology , Protein Kinase C/metabolism , Retinal Vessels , Transcription, Genetic/drug effects , Animals , Capillaries , Cattle , Cells, Cultured , Endothelium, Vascular/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Indoles/pharmacology , Isoenzymes/metabolism , Maleimides/pharmacology , Pericytes/drug effects , Pericytes/physiology , Protein Kinase C beta , Protein Kinase C-alpha , Protein Kinase C-delta , RNA, Messenger/genetics
7.
Invest Ophthalmol Vis Sci ; 41(5): 1192-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10752960

ABSTRACT

PURPOSE: Angiotensin II (AII) has been shown to play a role in many vascular diseases. In the study described, the effect of AII on vascular endothelial growth factor (VEGF) expression and related intracellular signaling mechanism was investigated in bovine retinal microcapillary pericytes. METHODS: Cultured bovine retinal microvascular endothelial cells and pericytes were prepared. VEGF expression was determined by Northern blot analysis and immunoprecipitation assay. Cell proliferation was assessed by DNA content growth assay. Reporter gene studies were performed to identify the AII responsible transcription-activating region of VEGF gene. RESULTS: Angiotensin II induced a significant increase in VEGF mRNA in a time- and dose-dependent manner. Angiotensin II type I receptor antagonist inhibited this effect. Angiotensin II activates the transcription of VEGF gene without changing the mRNA half-life, and the AII responsible region was found in the 5'-flanking region of the VEGF gene. Angiotensin II also increased the expression of c-fos and c-jun mRNA, and antisense oligonucleotides against c-Fos blocked the AII-induced VEGF mRNA expression. The conditioned media of AII-stimulated pericyte cultures had a growth-promoting effect on endothelial cells, and this effect was inhibited almost completely by VEGF neutralizing antibody. CONCLUSIONS: These findings suggest that AII might induce angiogenic activity through a paracrine function of VEGF in retinal microvascular cells.


Subject(s)
Angiotensin II/pharmacology , Endothelial Growth Factors/genetics , Lymphokines/genetics , Pericytes/drug effects , RNA, Messenger/biosynthesis , Retina/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Blotting, Northern , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Growth Factors/biosynthesis , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Genes, fos/genetics , Genes, jun/genetics , Lymphokines/biosynthesis , Oligonucleotides, Antisense/pharmacology , Pericytes/metabolism , Retina/metabolism , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Invest Ophthalmol Vis Sci ; 40(9): 1891-8, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10440240

ABSTRACT

PURPOSE: To investigate the distribution of inflammatory mediators such as interleukin (IL)-1beta and tumor necrosis factor (TNF)-alpha and angiogenic cytokines such as vascular endothelial growth factor (VEGF) and to identify their cellular source in surgically excised choroidal neovascular membranes (CNVMs) of various origins. METHODS: Immunoperoxidase staining was performed on paraffin-embedded sections of 11 surgically excised CNVMs to identify cellular distribution and localization of cytokines. Immunofluorescent double staining was performed to detect the cellular source of cytokines. RESULTS: Cytokeratin-positive cells were detected in the RPE layer, in stromal cells, and around neovascular vessels. Macrophages identified by their cellular marker CD68 showed almost the same distribution as cytokeratin-positive cells, although they were most prominent in the stroma. A substantial number of neovascular vessels were also immunoreactive to IL-1beta and TNF-alpha. Immunofluorescent double staining revealed that the RPE layers immunopositive for cytokeratin were also immunopositive for all cytokines, whereas stromal cells immunostained for CD68 were positive for IL-1beta and TNF-alpha, but not for VEGF. CONCLUSIONS: These results indicate that IL-1beta and TNF-alpha secreted by macrophages may promote, at least in part, angiogenesis in CNVMs by stimulating VEGF production in RPE cells.


Subject(s)
Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Endothelial Growth Factors/metabolism , Interleukin-1/metabolism , Lymphokines/metabolism , Macrophages/physiology , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Choroidal Neovascularization/pathology , Female , Fluorescent Antibody Technique, Indirect , Humans , Immunoenzyme Techniques , Male , Pigment Epithelium of Eye/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Invest Ophthalmol Vis Sci ; 40(9): 2122-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10440269

ABSTRACT

PURPOSE: Estrogen is known to promote angiogenesis in gonads. The presence of estrogen receptors in the vascular endothelium of organs other than gonads has been reported. The goal of this study was to determine whether estrogen promotes the proliferation of retinal microvascular endothelial cells and to explore the mechanism of it. METHODS: DNA was quantitated using primary cultures of bovine retinal endothelial cells that were incubated with different doses of 17 beta-estradiol (E2), VEGF, or both. The changes in expression level of VEGF and VEGF receptor-2 (VEGFR2) were measured using northern blot analysis after treatment with E2. The presence of estrogen receptors in the endothelial cells was studied by immunohistochemistry and western blot analysis. RESULTS: 17 Beta-estradiol (E2) increased the DNA level in bovine retinal capillary endothelial cells (BRECs) by 177% at 1 nM (P < 0.05) and 150% at 10 nM (P < 0.05) by comparison with unstimulated BREC. One hundred nanomole tamoxifen completely blocked the E2-induced DNA synthesis in BRECs. Ten nanomole E2 augmented vascular endothelial growth factor (VEGF)-induced DNA synthesis in BRECs significantly (160%, P < 0.01). Ten nanomole E2 also increased VEGF mRNA expression, which peaked after 24 hours (6.7 times, P < 0.05), and VEGF receptor-2 (VEGFR2) mRNA expression, which peaked after 9 hours (2.4 times, P < 0.05). The mRNA expression level of VEGFR2 peaked with 10 nM E2 (P < 0.05) and that of VEGF reached maximum with 1 nM E2 (15 times, P < 0.001). VEGFR2 and VEGF proteins increased in parallel with their mRNA levels. Immunocytochemistry showed estrogen receptor expression in BRECs, and western blot analysis indicated the presence of a 67-kDa protein that was compatible with the estrogen receptor. CONCLUSIONS: These findings suggest that E2 may stimulate BREC growth by the receptor-mediated pathway and that E2 may augment the VEGF-dependent angiogenesis partly through the upregulation of VEGFR2.


Subject(s)
DNA/biosynthesis , Endothelium, Vascular/drug effects , Estradiol/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Retinal Vessels/drug effects , Animals , Blotting, Northern , Blotting, Western , Cattle , Cell Division/drug effects , Cells, Cultured , DNA Replication/drug effects , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Estrogen Antagonists/pharmacology , Fluorescent Antibody Technique, Indirect , Lymphokines/genetics , Lymphokines/metabolism , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Estrogen/metabolism , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Retinal Vessels/metabolism , Tamoxifen/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
J Immunol ; 163(1): 374-9, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10384138

ABSTRACT

Anterior uveitis associated with Behcet's disease and ankylosing spondylitis preferentially occurs in adult men, which may suggest the effects of sex hormones on acute anterior uveitis. Recently, estrogen receptors in the vascular endothelium have been reported to be involved in several pathological conditions. In the present study, we examined the gender differences in susceptibility to endotoxin-induced uveitis (EIU) and the effects of estrogen on anterior inflammation. EIU was induced in adult male, female, and ovariectomized female Lewis rats (200 g) by hind footpad injection of 200 microg of LPS. In EIU, cellular infiltration was more marked in male than in female rats, and ovariectomy increased cellular infiltration. Treatment with 10 microg of 17beta-estradiol significantly reduced the cell number in male and ovariectomized female rats with EIU. Estrogen receptor immunoreactivity was found in the nucleus of vascular endothelium and in some stromal cells of the iris-ciliary body. Semiquantitative PCR revealed that E-selectin and IL-6 gene expressions were increased in rats following LPS injection, and an overdose of tamoxifen, an estrogen receptor antagonist, reversed the effect of 17beta-estradiol on E-selectin, but not its effect on IL-6. These observations suggested that the down-modulation of these inflammatory genes by estrogen may contribute to the reduction in cellular infiltration in acute anterior uveitis.


Subject(s)
Cell Movement/drug effects , E-Selectin/biosynthesis , Estradiol/pharmacology , Interleukin-6/antagonists & inhibitors , Lipopolysaccharides/toxicity , Uveitis, Anterior/pathology , Uveitis, Anterior/prevention & control , Animals , Ciliary Body/metabolism , E-Selectin/analysis , E-Selectin/genetics , Endothelium, Vascular/metabolism , Eye Proteins/metabolism , Female , Gene Expression , Immunohistochemistry , Interleukin-6/biosynthesis , Interleukin-6/genetics , Iris/metabolism , Male , Rats , Rats, Inbred Lew , Receptors, Estrogen/metabolism , Uveitis, Anterior/metabolism
11.
Br J Pharmacol ; 127(2): 537-45, 1999 May.
Article in English | MEDLINE | ID: mdl-10385256

ABSTRACT

1. Tranilast, first developed as an anti-allergic drug, has been reported to inhibit vascular endothelial growth factor (VEGF)-induced angiogenesis and vasopermeability. To further clarify the inhibitory mechanism, we investigated the effects of tranilast on VEGF binding and subsequent intracellular signalling pathway linked to angiogenic activities and gene expression of bovine retinal microcapillary endothelial cells. 2. Tranilast significantly (P<0.01) inhibited VEGF, basic fibroblast growth factor (bFGF), and hypoxia conditioned media-induced BREC proliferation in a dose dependent manner with IC50's of 22, 82 and 10 microM, respectively. 3. VEGF-induced migration was also inhibited by tranilast in a dose dependent manner, with IC50 of 18 microM, and complete inhibition was observed at 300 microM (P<0.01). Tranilast suppressed VEGF-induced tube formation in a dose dependent manner with maximum (46%) inhibition observed at 300 microM (P<0.05). 4. Tranilast inhibited phorbol myristate acetate (PMA)-dependent stimulation of [3H]-thymidine incorporation and VEGF- and PMA-induced gene expression of integrin alpha v and c-fos in BREC. 5. Tranilast suppressed VEGF- and PMA-stimulated PKC activity in BREC. 6. Tranilast did not affect VEGF binding or VEGF-induced phosphorylation of tyrosine residues of VEGF receptor- and phospholipase Cgamma and their associated proteins. 7. These data suggest that tranilast might prove an effective inhibitor to prevent retinal neovascularization in ischaemic retinal diseases, and that its inhibitory effect might be through suppression of PKC-dependent signal transduction in BREC.


Subject(s)
Anti-Allergic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Neovascularization, Physiologic/drug effects , Protein Kinase C/antagonists & inhibitors , Retinal Vessels/metabolism , Signal Transduction/drug effects , ortho-Aminobenzoates/pharmacology , Animals , Capillaries/drug effects , Capillary Permeability/drug effects , Cattle , Cell Division/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , DNA/biosynthesis , Endothelial Growth Factors/antagonists & inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Lymphokines/antagonists & inhibitors , Phosphorylation , Protein Kinase C/physiology , Retinal Vessels/cytology , Retinal Vessels/drug effects , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology , Tyrosine/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
J Biol Chem ; 274(22): 15732-9, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10336473

ABSTRACT

Recent studies have shown that the angiopoietin-Tie2 system is a predominant regulator of vascular integrity. In this study, we investigated the effect of two known angiogenic stimuli, hypoxia and vascular endothelial growth factor (VEGF), on these molecules. VEGF induced both a time- and concentration-dependent increase in angiopoietin-2 (Ang2) mRNA expression in bovine microvascular endothelial cells. This up-regulation was derived primarily from an increased transcription rate as evidenced by nuclear run-on assay and mRNA decay study. The increased Ang2 expression upon VEGF treatment was almost totally abolished by inhibition of tyrosine kinase or mitogen-activated protein kinase and partially by suppression of protein kinase C. Hypoxia also directly increased Ang2 mRNA expression. In contrast, Ang1 and Tie2 responded to neither of these stimuli. The enhanced Ang2 expression following VEGF stimulation and hypoxia was accompanied by de novo protein synthesis as detected by immunoprecipitation. In a mouse model of ischemia-induced retinal neovascularization, Ang2 mRNA was up-regulated in the ischemic inner retinal layer, and remarkable expression was observed in neovascular vessels. These data suggest that both hypoxia- and VEGF-induced neovascularization might be facilitated by selective induction of Ang2, which deteriorates the integrity of preexisting vasculature.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Lymphokines/pharmacology , Proteins/genetics , Angiopoietin-2 , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cattle , Cell Hypoxia , Cells, Cultured , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Kinetics , Mice , Neovascularization, Physiologic/drug effects , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Retina/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Am J Pathol ; 154(2): 343-54, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10027393

ABSTRACT

Thrombospondin-1 is an extracellular matrix protein that inhibits endothelial cell proliferation, migration, and angiogenesis. This study was performed to investigate the role of thrombospondin-1 in ischemic retinal neovascularization. In a murine model of retinal neovascularization, thrombospondin-1 mRNA was increased from postnatal day 13 (P13), with a threefold peak response observed on P15, corresponding to the time of development of retinal neovascularization. Prominent expression of thrombospondin-1 was observed in neovascular cells, specifically, cells adjacent to the area of nonperfusion. It has been suggested that vascular endothelial growth factor (VEGF) plays a major role in ischemia-induced retinal neovascularization of this model, so we studied the effects of VEGF on thrombospondin-1 expression. In bovine retinal microcapillary endothelial cells, VEGF induced a biphasic response of thrombospondin-1 expression; VEGF decreased thrombospondin-1 mRNA 0.41-fold after 4 hours, whereas it increased, with a threefold peak response, after 24 hours. VEGF-induced endothelial cell proliferation was completely inhibited by exogenous thrombospondin-1 and increased by 37.5% with anti-thrombospondin-1 antibody. The present findings suggest that, in the ischemic retina, retinal neovascular cells increase thrombospondin-1 expression, and VEGF may stimulate endogenous thrombospondin-1 induction, which inhibits endothelial cell growth. VEGF-mediated thrombospondin-1 induction in ischemia-induced angiogenesis may be a negative feedback mechanism.


Subject(s)
Ischemia/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Thrombospondin 1/metabolism , Animals , Animals, Newborn , Cattle , Cells, Cultured , Cycloheximide/pharmacology , Disease Models, Animal , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Infant, Newborn , Ischemia/pathology , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Pregnancy , RNA, Messenger/biosynthesis , Rats , Retinal Neovascularization/etiology , Retinal Neovascularization/pathology , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Thrombospondin 1/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Microvasc Res ; 56(3): 183-91, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9828156

ABSTRACT

Although the vascular endothelial growth factor (VEGF)/VEGF receptor system plays a critical role in the pathogenesis of ischemic retinal neovascular diseases such as diabetic retinopathy, regulation of VEGF receptor expression in ischemic retina has not been fully investigated in vivo. Accordingly, we studied the regulation of Flt-1 (VEGFR-1) and KDR/Flk-1 (VEGFR-2) expression in a mouse model of ischemia-induced retinal neovascularization. Immunohistochemistry for Flt-1 and KDR/Flk-1 revealed that, in hypoxic retina, the immunoreactivity of KDR/Flk-1 was increased in both intensity and extent of involvement in the vessels near the avascular area, particularly at the neovascular tufts, but that the pattern of Flt-1 expression in hypoxic retina was almost the same as that of control animals. The number of KDR/Flk-1-positive vessels was significantly increased in hypoxic retina (P < 0.01). In addition, expression of both Flt-1 and KDR/Flk-1 was observed in nonvascular cells of the neural retina. Northern blot analysis demonstrated that the mRNA levels of KDR/Flk-1 were greater in the neovascular retina of hypoxic animals than in control animals. We suggest that the increased expression of KDR/Flk-1 in vascular cells might potentiate the VEGF-mediated angiogenesis that accompanies many ischemic retinal diseases.


Subject(s)
Eye/blood supply , Ischemia/complications , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Growth Factor/biosynthesis , Retinal Neovascularization/metabolism , Animals , Endothelial Growth Factors/metabolism , Immunohistochemistry , Lymphokines/metabolism , Mice , Mice, Inbred C57BL , Receptors, Vascular Endothelial Growth Factor , Retinal Neovascularization/etiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Am J Ophthalmol ; 126(3): 409-16, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9744374

ABSTRACT

PURPOSE: To evaluate the retinal thickness analyzer, a new image analyzer involving laser-slit biomicroscopy, for quantitative assessment of retinal thickness in patients with macular edema secondary to retinal vein occlusion. METHODS: Fifty-eight patients (central retinal vein occlusion, 22 eyes; branch retinal vein occlusion, 36 eyes) with a clinical diagnosis of macular edema associated with retinal vein occlusion were evaluated. Retinal thickness analysis was performed with the retinal thickness analyzer. A green helium-neon laser slit directed into the eye was scanned to generate optical cross-sections of the retina. Images were analyzed by an automated software algorithm, and a detailed map of the retinal thickness was obtained. RESULTS: Laser-slit images obtained with the retinal thickness analyzer in patients with macular edema associated with retinal vein occlusion disclosed intraretinal changes such as retinal edema, cystoid spaces, and hemorrhages. Linear regression analysis showed a significant relationship between the macular thickness in the central fovea and corrected visual acuity reported on a logMAR scale (P = .002, adjusted R2 = 0.40 in central retinal vein occlusion; P = .002, adjusted R2 = 0.34 in branch retinal vein occlusion). CONCLUSION: We confirmed the capability of the retinal thickness analyzer to provide rapid and precise evaluation of macular thickening in patients with retinal vein occlusion.


Subject(s)
Edema/pathology , Macula Lutea/pathology , Retina/pathology , Retinal Diseases/pathology , Retinal Vein Occlusion/complications , Adult , Aged , Diagnostic Techniques, Ophthalmological/instrumentation , Edema/etiology , Female , Fluorescein Angiography , Fundus Oculi , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Retinal Diseases/etiology , Visual Acuity
16.
Invest Ophthalmol Vis Sci ; 39(9): 1620-30, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9699551

ABSTRACT

PURPOSE: The selectin family is a group of early-reactive adhesion molecules that plays a role in the rolling phase of leukocytes in cellular infiltration. It has been reported that P-selectin is expressed on vascular endothelium in the iris-ciliary body 15 minutes after lipopolysaccharide (LPS) treatment in endotoxin-induced uveitis (EIU) and may contribute to the initial phase of ocular inflammation. The objective of the present study was to identify the expression pattern of E-selectin, another member of the selectin family, and to investigate the role of E-selectin during the course of EIU. METHODS: Endotoxin-induced uveitis was induced in male Lewis rats by a footpad injection of 200 microg LPS. The time-dependent expressions of E-selectin in EIU in the iris- ciliary body and the retina were studied by immunohistochemistry using wholemounts and paraffin-embedded sections and by monitoring the level of E-selectin mRNA expression. A monoclonal antibody to E-selectin and a control antibody were each injected intravenously to evaluate the effects of E-selectin inhibition on ocular inflammation at the time of maximum uveitis. In the anterior uvea, the effect was evaluated by the number of infiltrated cells and by the protein concentration in the aqueous humor 24 hours after LPS treatment; in the retina, the myeloperoxidase (MPO) activity was measured 48 hours after LPS treatment. The effect of the combined injection of anti-P-selectin and anti-E-selectin antibodies was also studied. It was then determined whether the delayed inhibition of E-selectin (6, 12, or 24 hours after LPS injection) could contribute to the early resolution of the uveitis. RESULTS: E-selectin immunoreactivity was observed on the vessel walls of the iris and retina 7 hours after LPS treatment in wholemounts and paraffin-embedded sections and remained positive for 24 hours after LPS treatment. The expression of E-selectin messenger RNA gene peaked at 6 hours and again at 18 hours after LPS treatment in the iris- ciliary body and retina. The expression returned to the basal level 24 hours after LPS treatment in the iris- ciliary body and 48 hours after LPS treatment in the retina. The selective inhibition of E-selectin significantly blocked the cellular infiltration into the aqueous humor (P < 0.005) but had a milder effect on the protein concentration in the aqueous humor (P=0.0536). The inhibition of E-selectin and P-selectin almost abrogated cellular infiltration (P < 0.001). Myeloperoxidase activity in the retina 48 hours after LPS treatment was again significantly decreased by the inhibition of E-selectin alone and by the inhibition of E-selectin and P-selectin (P < 0.0001). A single injection of anti-E-selectin antibody 6, 12, or 24 hours after LPS injection effectively blocked cellular infiltration in the aqueous humor 24 and 48 hours after LPS treatment. CONCLUSIONS: In EIU, E-selectin may be expressed on the vascular endothelium and remain after the period of expression of P-selectin and until approximately the time of maximum uveitis. The present results suggest that, in contrast to the role of P-selectin as an initiator of cellular infiltration, E-selectin may contribute to continuing cellular infiltration into the inflammatory site during inflammation, and its inhibition may contribute to the early resolution of the uveitis.


Subject(s)
Chemotaxis, Leukocyte/physiology , E-Selectin/physiology , Lipopolysaccharides , Neutrophils/physiology , Salmonella , Uveitis/metabolism , Animals , Antibodies, Monoclonal , Ciliary Body/metabolism , Ciliary Body/pathology , E-Selectin/genetics , Electrophoresis, Agar Gel , Immunoenzyme Techniques , Iris/metabolism , Iris/pathology , Male , Peroxidase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Retina/metabolism , Retina/pathology , Uveitis/chemically induced , Uveitis/pathology
17.
Microvasc Res ; 55(3): 230-40, 1998 May.
Article in English | MEDLINE | ID: mdl-9657923

ABSTRACT

P-selectin is one of the adhesion molecules involved in leukocyte rolling during an inflammatory reaction. The aim of this study was to examine the role of P-selectin in leukocyte-endothelial interactions in retinal microcirculation during ocular inflammation, known as endotoxin-induced uveitis (EIU), in vivo. EIU was induced in Lewis rats by footpad injection of lipopolysaccharide (LPS). At the time of LPS treatment or 12 h later, anti-rat P-selectin mAb (ARP) was injected intravenously, and its effect on leukocyte behavior in the retina was studied after intravital staining with acridine orange using a scanning laser ophthalmoscope. P-selectin gene expression in the retina was also studied by a semiquantitative polymerase chain reaction (PCR) method. Administration of ARP at the time of LPS treatment significantly reduced the number of rolling leukocytes at 6 and 12 h by 68% (P < 0.05) and 83% (P < 0.01), respectively, and the number of cells infiltrating the vitreous at 48 h by 61% (P < 0.05). Interestingly, ARP significantly inhibited the vasodilation observed during EIU. In contrast, delayed administration of ARP blocked neither cellular infiltration nor vasodilation. P-selectin gene expression was upregulated during the course of EIU. In conclusion, P-selectin may significantly contribute to the development of inflammation in the early stage of endotoxin-induced ocular inflammation.


Subject(s)
Cell Communication/immunology , Endothelium, Vascular/pathology , Leukocytes/pathology , P-Selectin/immunology , P-Selectin/physiology , Retinitis/pathology , Uveitis/pathology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Movement/immunology , Endothelium, Vascular/physiopathology , Female , Leukocyte Count , Microcirculation/pathology , Microcirculation/physiopathology , Rats , Rats, Inbred Lew , Retinitis/physiopathology , Uveitis/physiopathology
18.
Invest Ophthalmol Vis Sci ; 39(6): 1028-35, 1998 May.
Article in English | MEDLINE | ID: mdl-9579483

ABSTRACT

PURPOSE: Integrins alphavbeta3 and alphavbeta5 are cell-to-matrix adhesion molecules that have been reported to mediate vascular cell proliferation and migration. The authors investigated the regulation of expression of these angiogenic integrins by hypoxia and vascular endothelial growth factor (VEGF) in retinal microvascular endothelial cells in culture. METHODS: Cultured bovine retinal capillary endothelial cells were exposed to human recombinant VEGF under normoxic (95% air, 5% CO2) conditions to assess the effects of VEGF. Hypoxia studies were performed under lower oxygen concentration (0.5%-1.5% O2) induced by nitrogen replacement in constant 5% CO2 conditions. Integrin family mRNA and protein expression were assessed by northern blot analysis and immunoprecipitation. RESULTS: VEGF (25 ng/ml) increased integrin alphav, beta3, and 35 mRNA after 24 hours 6.1+/-0.8-fold (P < 0.001), 5.9+/-1.1-fold (P < 0.001), and 1.9+/-0.2-fold (P < 0.01), respectively. Similarly, hypoxia stimulated gene expression of integrin alphav and beta3 after 24 hours by 5.1+/-1.7-fold (P < 0.01) and 3.0+/-0.5-fold (P < 0.01), respectively, and integrin beta5 after 9 hours 1.4+/-0.2-fold (P < 0.05). This hypoxia-induced, integrin alphav mRNA elevation was inhibited significantly by anti-VEGF neutralizing antibody. Also, a conditioned medium from confluent endothelial cells maintained under hypoxic conditions for 24 hours produced a 7.1+/-1.1-fold increase (P < 0.001) in integrin alphav mRNA expression after 24 hours, which was reversed by anti-VEGF neutralizing antibody. Induction of integrin alphav by VEGF and hypoxia was confirmed in the protein level. CONCLUSIONS: These data suggest that hypoxia stimulates expression of vascular integrins alphavbeta3 and alphavbeta5 in retinal microvascular endothelial cells partially through autocrine-paracrine action of VEGF induced by the hypoxic state.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Integrins/metabolism , Lymphokines/pharmacology , Receptors, Vitronectin/metabolism , Retinal Vessels/metabolism , Animals , Antigens, CD/metabolism , Blotting, Northern , Cattle , Cell Hypoxia/physiology , Cells, Cultured , DNA Primers/chemistry , Electrophoresis, Agar Gel , Endothelial Growth Factors/immunology , Endothelium, Vascular/drug effects , Gene Expression , Integrin alphaV , Lymphokines/immunology , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Retinal Vessels/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Circ Res ; 82(5): 619-28, 1998 Mar 23.
Article in English | MEDLINE | ID: mdl-9529167

ABSTRACT

Angiotensin II (Ang II) plays a role in the development of many vascular diseases. In the present study, we have investigated the effect of Ang II on vascular endothelial growth factor (VEGF) receptor expression and VEGF-induced angiogenic activity in bovine retinal microcapillary endothelial cells (BRECs). Ang II induced a significant increase of kinase domain-containing receptor/total liver kinase (KDR/Flk-1) mRNA in a time- and dose-dependent manner, with a maximal 4.3+/-0.8-fold increase after a 4-hour stimulation. Ang II increased the rate of KDR gene transcription by 5.4-fold, whereas the half-life of KDR mRNA was not increased significantly. The increase depended partially on new protein synthesis. The Ang II-induced KDR mRNA increase was inhibited by either [Sar1,Ile8]angiotensin or angiotensin type 1 receptor antagonists but was not significantly altered by angiotensin type 2 receptor antagonists. The PKC inhibitor reduced Ang II-induced KDR mRNA expression by 70+/-15%. The tyrosine kinase inhibitor reduced the Ang II- and phorbol 12-myristate 13-acetate-induced KDR mRNA increases by 35+/-8% and 44+/-26%, respectively. Ang II increased by 3.1-fold the 35S-labeled KDR/Flk-1 immunoprecipitated by a specific antibody to KDR/Flk-1. Scatchard analysis demonstrated that Ang II induced a significant increase of binding sites without changing binding affinity. Ang II enhanced VEGF-induced cell growth and tube formation. Ang II itself had no effect on cell growth, tube formation, or mRNA levels of VEGF and tms-like tyrosine kinase (Flt-1) in BRECs. These findings suggest that Ang II might potentiate VEGF-induced angiogenic activity through an increase of the VEGF receptor KDR/Flk-1.


Subject(s)
Angiotensin II/pharmacology , Endothelium, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Animals , Aorta/cytology , Capillaries/cytology , Capillaries/drug effects , Capillaries/enzymology , Cattle , Cell Division/drug effects , Cells, Cultured , Endothelium, Vascular/enzymology , Gene Expression/drug effects , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/metabolism , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , Receptors, Vascular Endothelial Growth Factor , Retina/cytology , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects
20.
Invest Ophthalmol Vis Sci ; 38(8): 1610-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9224289

ABSTRACT

PURPOSE: P-selectin is one of the early-reactive adhesion molecules that play a part in the rolling phase of leukocytes in cellular infiltration. The study objective was to determine whether P-selectin is involved in the development of endotoxin-induced uveitis (EIU). METHODS: Endotoxin-induced uveitis was initiated in male Lewis rats by injecting 200 micrograms lipopolysaccharide (LPS) into the foot pad. Expression of P-selectin in the iris-ciliary body was studied by immunohistochemistry using wholemounts and paraffin embedded sections of iris-ciliary body prepared at various time intervals. A monoclonal antibody (mAb) against P-selectin and a ligand for P-selectin, sialyl Lewis X oligosaccharide (SLeX-OS), was intravenously injected to evaluate the effects of selectin inhibition. The effect of treatment was evaluated by the number of infiltrated cells and protein concentration in the aqueous humor at 24 hours after LPS treatment. RESULTS: P-selectin immunoreactivities were observed in the vessels of the iris in whole iris-ciliary body mounts and on the surface of the microvascular endothelium in paraffin-embedded sections. Activity was most prominent at 15 minutes and at 5 to 7 hours after LPS treatment and was moderate from 1 to 4 hours after treatment. The selective inhibition of P-selectin significantly blocked the cellular infiltration into aqueous humor, but this infiltration was even more effectively inhibited by SLeX-OS. Protein concentration in the aqueous humor was not inhibited by selectin as much as was cellular infiltration. CONCLUSIONS: In the early phase of EIU, P-selectin may be expressed on the vascular endothelium in the iris in a biphasic pattern that modulates the rolling phase of leukocytes. The expression of this molecule may be essential for succeeding processes of cellular infiltration and may determine the subsequent states of ocular inflammation.


Subject(s)
Endotoxins , P-Selectin/physiology , Uveitis/chemically induced , Animals , Aqueous Humor/metabolism , Endothelium, Vascular/metabolism , Eye Proteins/metabolism , Immunoenzyme Techniques , Iris/blood supply , Lipopolysaccharides , Male , Microcirculation , Osmolar Concentration , Rats , Rats, Inbred Lew , Time Factors , Uveitis/metabolism , Uveitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...