Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 707
Filter
1.
Exp Eye Res ; 216: 108945, 2022 03.
Article in English | MEDLINE | ID: mdl-35038456

ABSTRACT

The VEGF-A-induced functional impairment of the barrier formed by retinal endothelial cells (REC) can be prevented and even - at least temporarily - reverted by trapping the growth factor in a complex with a VEGF-binding protein or by inhibiting the activity of the VEGF receptor 2 (VEGFR2). In an approach to emulate the clinically relevant situation of constant exposure to effectors, we investigated (1) whether prolonged exposure to VEGF-A165 for up to six days results in a different type of disturbance of the barrier formed by immortalized bovine REC (iBREC) and (2) whether alterations of the barrier induced by VEGF-A165 can indeed be sustainably reverted by subsequent treatment with the VEGF-A-binding proteins ranibizumab or brolucizumab. As a measure of barrier integrity, the cell index (CI) of iBREC cultivated on gold electrodes was monitored continuously. CI values declined shortly after addition of the growth factor and then remained low for more than six days over which considerable amounts of both extra- and intracellular VEGF-A were measured. Interestingly, the specific VEGFR2 inhibitor nintedanib normalized the lowered CI when added to iBREC pre-treated with VEGF-A165 for one day, but failed to do so when cells had been exposed to the growth factor for six days. Expression of the tight junction (TJ) protein claudin-5 was unchanged early after addition of VEGF-A165 but higher after prolonged treatment, whereas decreased amounts of the TJ-protein claudin-1 remained low, and increased expression of the plasmalemma vesicle-associated protein (PLVAP) remained high during further exposure. After two days, the characteristic even plasma membrane stainings of claudin-1 or claudin-5 appeared weaker or disordered, respectively. After six days the subcellular localization of claudin-5 was similar to that of control cells again, but claudin-1 remained relocated from the plasma membrane. To counteract these effects of VEGF-A165, brolucizumab or ranibizumab was added after one day, resulting in recovery of the then lowered CI to normal values within a few hours. However, despite the VEGF antagonist being present, the CI declined again two days later to values that were just slightly higher than without VEGF inhibition during further assessment for several days. At this stage, neither the supernatants nor whole cell extracts from iBREC treated with VEGF-A165 and its antagonists contained significant amounts of free VEGF-A. Treatment of VEGF-A165-challenged iBREC with ranibizumab or brolucizumab normalized expression of claudin-1 and claudin-5, but not completely that of PLVAP. Interestingly, the characteristic VEGF-A165-induced relocalization of claudin-1 from the plasma membrane was reverted within one day by any of the VEGF antagonists, but reappeared despite their presence after further exposure for several days. Taken together, barrier dysfunction induced by VEGF-A165 results from deregulated para- and transcellular flow but the precise nature or magnitude of underlying changes on a molecular level clearly depend on the time of exposure, evolving into a stage of VEGF-A165-independent barrier impairment. These findings also provide a plausible explanation for resistance to treatment with VEGF-A antagonists frequently observed in clinical practice.


Subject(s)
Endothelial Cells/drug effects , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Retinal Vessels/cytology , Tight Junctions/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Biological Transport , Blotting, Western , Cattle , Cell Movement/drug effects , Cells, Cultured , Claudin-1/metabolism , Claudin-5/metabolism , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Ranibizumab/therapeutic use
2.
J Ethnopharmacol ; 283: 114652, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34626779

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lycium barbarum L., a classical traditional Chinese Medicine, has long been used to treat ocular diseases. Lycium barbarum polysaccharides (LBP) is an effective component of Lycium barbarum L. with a wide range of pharmacological activities. This research aims to investigate the inhibition of high glucose-induced angiogenesis by LBP in RF/6A cells. MATERIALS AND METHODS: A high-glucose-induced angiogenesis model was established using monkey retinal vascular endothelial (RF/6A) cells. Different dosages administration times of LBP and glucose concentrations were tested. Under the optimized conditions, RF/6A cells were treated with LBP for 48 h, followed by another 48-h culture in high glucose (25 mmol/L) medium. The effect and mechanism of LBP were investigated following the treatment. RESULTS: The expression of miR-15a-5p and miR-15a-3p in RF/6A cells decreased significantly after 48 h of 25 or 50 mmol/L high glucose treatment. The expression of miR-15a-5p was higher than that of miR-15a-3p. Mimic-miR-15a-5p or 600 mg/L LBP could increase the apoptosis of cells and the total length of vascular branches. The expression of VEGFA, VEGFR2, and ANG2 proteins was reduced, while the expression of ANG1 protein was elevated. Expression of ASM mRNA and protein was also inhibited. CONCLUSIONS: LBP attenuates diabetic retinal angiogenesis by rescuing the expression of miR-15a-5p in RF/6A cells.


Subject(s)
Diabetic Retinopathy/drug therapy , Drugs, Chinese Herbal/pharmacology , MicroRNAs/genetics , Neovascularization, Pathologic/drug therapy , Animals , Apoptosis/drug effects , Cell Line , Diabetic Retinopathy/genetics , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Glucose/metabolism , Haplorhini , Neovascularization, Pathologic/genetics , Retinal Vessels/cytology , Retinal Vessels/drug effects
3.
Mol Biotechnol ; 64(2): 171-177, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34554391

ABSTRACT

Diabetic retinopathy (DR) is a serious complication of diabetes mellitus. The purpose of this study was to investigate the potential functional role of long non-coding RNA TUG1 in high glucose (HG)-stimulated human retinal microvascular endothelial cells (hRMECs). The results demonstrated that following 72 h of HG stimulation, enhanced proliferation, migration, and tube formation process were observed in hRMECs. Moreover, HG treatment markedly increased TUG1 expression in hRMECs, and knockdown of TUG1 notably restrained the aberrant phenotypes of hRMECs induced by HG. Mechanistically, TUG1 may serve as a competing endogenous RNA (ceRNA) for miR-145, thereby blocking the repression on VEGF-A in hRMECs. Rescue experiments further indicated that inhibition of miR-145 abolished the beneficial role of TUG1 knockdown in HG-treated hRMECs. Our data suggested that knockdown of TUG1 protects hRMECs against HG stimulation partly by regulating miR-145/VEGF-A axis.


Subject(s)
MicroRNAs/genetics , RNA, Long Noncoding/genetics , Retinal Vessels/cytology , Cell Movement/genetics , Cells, Cultured , Diabetic Retinopathy/genetics , Endothelial Cells , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glucose/pharmacology , Humans , Retinal Vessels/drug effects , Retinal Vessels/pathology , Vascular Endothelial Growth Factor A/genetics
4.
Can J Physiol Pharmacol ; 100(2): 142-150, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34559978

ABSTRACT

The endothelial-mesenchymal transition (EndMT) participates in the progression of diabetic retinopathy (DR), but cell-intrinsic factors modulating this process remain elusive. In this study, we explored the role of lysophosphatidic acid (LPA) - producing enzyme, acylglycerol kinase (AGK), in the EndMT of human retinal microvascular endothelial cells (HRECs) under high-glucose (HG) conditions. We found that AGK was significantly elevated in HG-treated cells. In addition, AGK knockdown reversed the HG-induced EndMT in HRECs, which was evidenced by the increased endothelial markers (CD31 and VE-cadherin) and decreased mesenchymal markers (FSP1 and α-SMA). Furthermore, downregulation of AGK inhibited the HG-induced activation of transforming growth factor ß (TGF-ß)/Notch pathways, whereas exogenous TGF-ß1 (10 ng/mL) impeded the inhibitory effects of AGK knockdown on HG-induced EndMT in HRECs. Additionally, the silencing of AGK abolished the HG-induced upregulation of LPA and its receptor, LPA receptor 1 (LPAR1), and overexpression of LPAR1 further rescued the AGK knockdown-mediated inhibition of the EndMT process. In conclusion, we demonstrate that downregulation of AGK suppresses HG-induced EndMT in HRECs through regulating the LPAR1/TGF-ß/Notch signaling pathway, indicating that AGK might be a potential therapeutic target for the treatment of DR.


Subject(s)
Down-Regulation/genetics , Down-Regulation/physiology , Endothelial Cells/physiology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Glucose/adverse effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysophosphatidic Acid/genetics , Receptors, Notch/metabolism , Retinal Vessels/cytology , Signal Transduction/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Cells, Cultured , Gene Expression Regulation/genetics , Humans , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Notch/genetics , Transforming Growth Factor beta/genetics
5.
Cell Mol Life Sci ; 79(1): 37, 2021 Dec 31.
Article in English | MEDLINE | ID: mdl-34971428

ABSTRACT

The roles of nitric oxide (NO) and endothelial NO synthase (eNOS) in the regulation of angiogenesis are well documented. However, the involvement of eNOS in the sprouting of endothelial tip-cells at the vascular front during sprouting angiogenesis remains poorly defined. In this study, we show that downregulation of eNOS markedly inhibits VEGF-stimulated migration of endothelial cells but increases their polarization, as evidenced by the reorientation of the Golgi in migrating monolayers and by the fewer filopodia on tip cells at ends of sprouts in endothelial cell spheroids. The effect of eNOS inhibition on EC polarization was prevented in Par3-depleted cells. Importantly, downregulation of eNOS increased the expression of polarity genes, such as PARD3B, PARD6A, PARD6B, PKCΖ, TJP3, and CRB1 in endothelial cells. In retinas of eNOS knockout mice, vascular development is retarded with decreased vessel density and vascular branching. Furthermore, tip cells at the extremities of the vascular front have a marked reduction in the number of filopodia per cell and are more oriented. In a model of oxygen-induced retinopathy (OIR), eNOS deficient mice are protected during the initial vaso-obliterative phase, have reduced pathological neovascularization, and retinal endothelial tip cells have fewer filopodia. Single-cell RNA sequencing of endothelial cells from OIR retinas revealed enrichment of genes related to cell polarity in the endothelial tip-cell subtype of eNOS deficient mice. These results indicate that inhibition of eNOS alters the polarity program of endothelial cells, which increases cell polarization, regulates sprouting angiogenesis and normalizes pathological neovascularization during retinopathy.


Subject(s)
Neovascularization, Pathologic , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/physiology , Retina/metabolism , Retinal Neovascularization , Retinal Vessels , Animals , Cattle , Cell Line , Cell Movement , Cell Polarity , Endothelial Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/cytology , Retina/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/cytology , Retinal Vessels/pathology
6.
Int J Biol Sci ; 17(15): 4316-4326, 2021.
Article in English | MEDLINE | ID: mdl-34803500

ABSTRACT

Background: Insulin therapy is the major treatment of glycaemic control in type I diabetes mellitus (DM) and advanced type II DM patients who fail to respond to oral hypoglycemic agents. Nonetheless, insulin therapy is deemed unsuccessful in controlling the incidence of diabetic retinopathy (DR) and is likely a risk factor. Berberine, an isoquinoline alkaloid, has caught great attention towards its anti-diabetic mechanisms. This study aims to investigate the effect of berberine in decelerating DR progression in insulin-treated DM. Methods: To better understand the therapeutic potential of berberine in the presence of insulin, we elaborated the action of mechanism whether berberine inhibited retinal expression of HIF-1α and VEGF through regulating AKT/mTOR pathway. Suppression of insulin-induced neovasculature of retina endothelial cells by berberine was also studied. Lastly, the in vivo efficacy and safety of berberine as adjuvant therapy for the treatment of DR were systemically investigated in experimental type I and type II DM mice with insulin treatment. Results: Among various types of retinal cells, the activity of HIF-1α and VEGF in retinal endothelial cells could be particularly and exclusively stimulated by insulin intervention, which could be inhibited by berberine treatment in a dose- and time-dependent manner. Berberine suppressed Akt/mTOR activity in these cells, and restoration of Akt/mTOR signalling attenuated berberine's inhibition on HIF-1α and VEGF expression. Berberine suppressed the progression of DR in experimental type I and type II diabetic mice receiving insulin therapy. Conclusion: Berberine improves insulin-induced diabetic retinopathy in type I and II diabetes through inhibiting insulin-induced activation of retinal endotheliocytes via Akt/mTOR/ HIF-1α/VEGF pathway.


Subject(s)
Berberine/pharmacology , Diabetic Retinopathy/chemically induced , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Diabetes Mellitus, Experimental , Endothelial Cells/drug effects , Endothelial Cells/physiology , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Proto-Oncogene Proteins c-akt/genetics , Retinal Vessels/cytology , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Exp Eye Res ; 213: 108861, 2021 12.
Article in English | MEDLINE | ID: mdl-34822853

ABSTRACT

Aberrant angiogenesis lies at the heart of a wide range of ocular pathologies such as proliferative diabetic retinopathy, wet age-related macular degeneration and retinopathy of prematurity. This study explores the anti-angiogenic activity of a novel small molecule investigative compound capable of inhibiting profilin1-actin interaction recently identified by our group. We demonstrate that our compound is capable of inhibiting migration, proliferation and angiogenic activity of microvascular endothelial cells in vitro as well as choroidal neovascularization (CNV) ex vivo. In mouse model of laser-injury induced CNV, intravitreal administration of this compound diminishes sub-retinal neovascularization. Finally, our preliminary structure-activity relationship study (SAR) demonstrates that this small molecule compound is amenable to improvement in biological activity through structural modifications.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Retinal Neovascularization/drug therapy , Actins/antagonists & inhibitors , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Choroidal Neovascularization/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Humans , Intravitreal Injections , Mice , Mice, Inbred C57BL , Profilins/antagonists & inhibitors , Retinal Neovascularization/metabolism , Retinal Vessels/cytology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Wet Macular Degeneration/drug therapy , Wet Macular Degeneration/metabolism
8.
Exp Eye Res ; 213: 108846, 2021 12.
Article in English | MEDLINE | ID: mdl-34801534

ABSTRACT

PURPOSE: Diabetic retinopathy is a vision-threatening complication of diabetes characterized by endothelial injury and vascular dysfunction. The loss of the endothelial glycocalyx, a dynamic layer lining all endothelial cells, contributes to several microvascular pathologies, including an increase in vascular permeability, leukocyte plugging, and capillary occlusion, and may drive the progression of retinopathy. Previously, a significant decrease in glycocalyx thickness has been observed in diabetic retinas. However, the effects of diabetes on specific components of the retinal glycocalyx have not yet been studied. Therefore, the aim of our study was to investigate changes in synthesis, expression, and shedding of retinal glycocalyx components induced by hyperglycemia, which could provide a novel therapeutic target for diabetic retinopathy. METHODS: Primary rat retinal microvascular endothelial cells (RRMECs) were grown under normal glucose (5 mM) or high-glucose (25 mM) conditions for 6 days. The mRNA and protein levels of the glycocalyx components were examined using qRT-PCR and Western blot analysis, respectively. Further, mass spectrometry was used to analyze protein intensities of core proteins. In addition, the streptozotocin-induced Type 1 diabetic rat model was used to study changes in the expression of the retinal glycocalyx in vivo. The shedding of the glycocalyx was studied in both culture medium and in plasma using Western blot analysis. RESULTS: A significant increase in the shedding of syndecan-1 and CD44 was observed both in vitro and in vivo under high-glucose conditions. The mRNA levels of syndecan-3 were significantly lower in the RRMECs grown under high glucose conditions, whereas those of syndecan-1, syndecan-2, syndecan-4, glypican-1, glypican-3, and CD44 were significantly higher. The protein expression of syndecan-3 and glypican-1 in RRMECs was reduced considerably following exposure to high glucose, whereas that of syndecan-1 and CD44 increased significantly. In addition, mass spectrometry data also suggests a significant increase in syndecan-4 and a significant decrease in glypican-3 protein levels with high glucose stimulation. In vivo, our data also suggest a significant decrease in the mRNA transcripts of syndecan-3 and an increase in mRNA levels of glypican-1 and CD44 in the retinas of diabetic rats. The diabetic rats exhibited a significant reduction in the retinal expression of syndecan-3 and CD44. However, the expression of syndecan-1 and glypican-1 increased significantly in the diabetic retina. CONCLUSIONS: One of the main findings of our study was the considerable diversity of glucose-induced changes in expression and shedding of various components of endothelial glycocalyx, for example, increased endothelial and retinal syndecan-1, but decreased endothelial and retinal syndecan-3. This indicates that the reported decrease in the retinal glycocalyx in diabetes in not a result of a non-specific shedding mechanism. Moreover, mRNA measurements indicated a similar diversity, with increases in endothelial and/or retinal levels of syndecan-1, glypican-1, and CD44, but a decrease for syndecan-3, with these increases in mRNA potentially a compensatory reaction to the overall loss of glycocalyx.


Subject(s)
Diabetic Retinopathy/metabolism , Glycocalyx/metabolism , Hyperglycemia/metabolism , Retina/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Glucose/pharmacology , Glypicans/metabolism , Hyaluronan Receptors/metabolism , Insulin/blood , Male , Mass Spectrometry , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Retinal Vessels/cytology , Syndecans/metabolism
9.
Exp Eye Res ; 213: 108828, 2021 12.
Article in English | MEDLINE | ID: mdl-34742690

ABSTRACT

Pigment epithelium derived factor (PEDF), an endogenous inhibitor of angiogenesis, targets the growth of aberrant blood vessels in many tissues, including the eye. In this study we show that PEDF prevented early mitogenic signals of vascular endothelial growth factor (VEGF-A) in primate retinal endothelial cells, blocking proliferation, migration and tube formation. PEDF inhibited the phosphorylation and activation of five major downstream VEGF-A signaling partners, namely phosphoinositide-3-OH Kinase (PI3K), AKT, FAK, Src (Y416), and PLC-γ. It did so by binding to the extracellular domain of VEGF-R2, blocking VEGF-A-induced tyrosine phosphorylation (Tyr 951 and Tyr 1175), and inhibiting VEGF-R2 receptor kinase activity. PEDF had no effect on the transcription or translation of VEGF-R2 in cultured HUVECs. PEDF also bound to the extracellular domain of VEGF-R1. We conclude that PEDF blocks the growth of new blood vessels, in part, by reducing VEGF-A activation of its key mitogenic receptor, VEGF-R2, and by preventing its downstream signals in endothelial cells.


Subject(s)
Angiogenesis Inhibitors/physiology , Endothelial Cells/drug effects , Eye Proteins/physiology , Nerve Growth Factors/physiology , Retinal Vessels/cytology , Serpins/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Blood Vessels/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Primates , Real-Time Polymerase Chain Reaction , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Bioengineered ; 12(1): 7508-7518, 2021 12.
Article in English | MEDLINE | ID: mdl-34608841

ABSTRACT

Hsa_circ_0001850_circ_0001850 (circ-UBAP2) is reported to be upregulated in diabetic retinopathy (DR). However, its role in high glucose (HG)-triggered oxidative stress and vascular dysfunction in DR is unclear. This study aimed to investigate the potential of circUBAP2 in DR. The content of malondialdehyde (MDA), and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) were analyzed using the corresponding kits. Western blotting was performed to detect the protein expression of Nrf2, HO-1, and SOD-1. MTT assay was conducted to assess cell viability. A transwell migration assay was used to determine the migration ability of human retinal microvascular endothelial cells (hRMECs). A Matrigel tube formation assay was performed to analyze tube formation. The targeting relationships were verified using a luciferase reporter assay. We found that the circ-UBAP2 expression increased in DR patients and HG-treated hRMECs. Downregulation of circ-UBAP2 ameliorated HG-induced oxidative stress and dysfunction of hRMECs. Mechanistically, circ-UBAP2 sponges miR-589-5p, which is downregulated under hyperglycemic conditions. In addition, EGR1 was confirmed to be a target gene of miR-589-5p and was overexpressed in HG-treated hRMECs. In addition, EGR1 reversed the effects of miR-589-5p and induced oxidative stress and dysfunction in hRMECs. Taken together, knockdown of circ-UBAP2 relieved HG-induced oxidative stress and dysfunctions of the hRMECs through the miR-589-5p/EGR1 axis, which may offer a promising therapeutic target for DR.


Subject(s)
Diabetic Retinopathy/metabolism , MicroRNAs/genetics , Oxidative Stress/genetics , RNA, Circular/genetics , Retinal Vessels/cytology , Cells, Cultured , Down-Regulation/genetics , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Endothelium, Vascular/cytology , Glucose/adverse effects , Humans , MicroRNAs/metabolism , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/genetics , Oxidative Stress/drug effects , RNA, Circular/metabolism
11.
Invest Ophthalmol Vis Sci ; 62(12): 22, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34546322

ABSTRACT

Purpose: CD40 is an upstream inducer of inflammation in the diabetic retina. CD40 is upregulated in retinal endothelial cells in diabetes. The purpose of this study was to determine whether expression of CD40 in endothelial cells is sufficient to promote inflammatory responses in the retina of diabetic mice. Methods: Transgenic mice with CD40 expression restricted to endothelial cells (Trg-CD40 EC), transgenic control mice (Trg-Ctr), B6, and CD40-/- mice were made diabetic using streptozotocin. Leukostasis was assessed using FITC-conjugated ConA. Pro-inflammatory molecule expression was examined by real-time PCR, immunohistochemistry, ELISA, or flow cytometry. Release of ATP was assessed by ATP bioluminescence. Results: Diabetic B6 and Trg-CD40 EC mice exhibited increased retinal mRNA levels of ICAM-1, higher ICAM-1 expression in endothelial cells, and increased leukostasis. These responses were not detected in diabetic mice that lacked CD40 (CD40-/- and Trg-Ctr). Diabetic B6 but not Trg-CD40 EC mice upregulated TNF-α, IL-1ß, and NOS2 mRNA levels. CD40 stimulation in retinal endothelial cells upregulated ICAM-1 but not TNF-α, IL-1ß, or NOS2. CD40 ligation did not trigger ATP release by retinal endothelial cells or pro-inflammatory cytokine production in bystander myeloid cells. In contrast to diabetic B6 mice, diabetic Trg-CD40 EC mice did not upregulate P2X7 mRNA levels in the retina. Conclusions: Endothelial cell CD40 promotes ICAM-1 upregulation and leukostasis. In contrast, endothelial cell CD40 does not lead to pro-inflammatory cytokine and NOS2 upregulation likely because it does not activate purinergic-mediated pro-inflammatory molecule expression by myeloid cells or induce expression of these pro-inflammatory molecules in endothelial cells.


Subject(s)
CD40 Antigens/genetics , Cytokines/genetics , Diabetic Retinopathy/genetics , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/genetics , Nitric Oxide Synthase Type II/genetics , Receptors, Purinergic P2X7/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation/physiology , Humans , Leukostasis , Luminescent Measurements , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Vessels/cytology , Up-Regulation
12.
Mol Vis ; 27: 575-587, 2021.
Article in English | MEDLINE | ID: mdl-34531648

ABSTRACT

Purpose: The correlation between chemerin and diabetic retinopathy (DR) has been demonstrated previously. We aimed to investigate the potential inflammatory and angiogenic roles of chemerin in DR using rat primary retinal microvascular endothelial cells (RRMECs). Methods: RRMECs were incubated in low- and high-glucose media, and stable chemerin receptor (ChemR23) knockdown in RRMECs was established by lentiviral infection. Real-time quantitative PCR (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), and western blotting were employed to investigate the mRNA and protein expression of intercellular adhesion molecule-1 (ICAM-1), vascular endothelial growth factor (VEGF), tumor necrosis factor-α (TNF-α), and the interleukin-6 receptor (IL-6R) to explore the inflammatory and angiogenic effects of chemerin. A scratch assay was employed to evaluate the effect of chemerin on RRMEC migration. Results: Chemerin and TNF-α markedly increased the mRNA and protein expression of ICAM-1 in RRMECs (p<0.001). ChemR23 knockdown may have decreased the ICAM-1 expression under low- and high-glucose conditions (p<0.001). Even in the ChemR23-knockdown group, TNF-α significantly increased the mRNA and protein levels of ICAM-1 under low- and high-glucose conditions (p<0.001). Chemerin promoted VEGF expression under low- and high-glucose conditions. ChemR23 knockdown markedly decreased VEGF levels under low- and high-glucose conditions (p<0.05) and significantly decreased RRMEC migration (p<0.001). Conclusions: Chemerin promotes the expression of ICAM-1, the secretion of VEGF, and the migration of RRMECs via the activation of ChemR23.


Subject(s)
Chemokines/physiology , Diabetic Angiopathies/metabolism , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Receptors, Chemokine/metabolism , Animals , Blotting, Western , Cells, Cultured , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay , Glucose/pharmacology , Intercellular Adhesion Molecule-1/genetics , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-6/genetics , Retinal Vessels/cytology , Tumor Necrosis Factor-alpha/genetics , Vascular Endothelial Growth Factor A/genetics
13.
Mol Vis ; 27: 528-541, 2021.
Article in English | MEDLINE | ID: mdl-34526760

ABSTRACT

Purpose: To identify a novel mutation in KIF11 with clinical and functional analysis among 516 familial patients with exudative vitreoretinopathy (FEVR). Methods: Next-generation sequencing was performed on 516 patients with FEVR between January 2015 and October 2017. Clinical data were collected from patient charts, including sex, age at presentation, visual acuity if available, axial length, stage, and systemic clinical findings. Protein and mRNA levels were detected with western blotting and real-time quantitative PCR, respectively. Mass spectrometry was used to analyze the interacting protein of KIF11. Results: In total, 304 of 516 patients were identified with at least one mutation in FEVR causative genes. Mutations in KIF11 were identified in 14.47% of all carriers. The novel mutation p. H718L accounted for the greatest proportion (12/44; 27.30%) among all mutations in KIF11. Fundus presentations in these 12 individuals varied from the avascular zone of the peripheral retina to total retinal detachment. The p. H718L mutation can reduce the proliferation of human retinal endothelial cells (HRECs) compared to the wild type. The mRNA level of vascular endothelial growth factor-α, transforming growth factor-α, metalloproteinase-1, and angiopoietin-like 4 were depressed in the KIF11 (p. H718L) groups under hypoxia stimuli. Mass spectrometry results demonstrated that eukaryotic elongation factor 2 (EEF2) was an interacting protein of KIF11 and that the p. H718L mutation can attenuate the binding activity. Conclusions: Patients with the most frequent KIF11 mutation p. H718L showed typical FEVR presentations in this cohort. The mutation in KIF11 likely plays a role in the proliferation of HRECs, and the p. H718L mutation can reduce the proliferation.


Subject(s)
Familial Exudative Vitreoretinopathies/genetics , Kinesins/genetics , Mutation/genetics , Angiopoietin-Like Protein 4/genetics , Axial Length, Eye , Blotting, Western , Cell Proliferation/physiology , Child , Child, Preschool , Endothelium, Vascular/physiology , Familial Exudative Vitreoretinopathies/physiopathology , Female , Fluorescein Angiography , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Mass Spectrometry , Matrix Metalloproteinase 1/genetics , Pedigree , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Vessels/cytology , Transforming Growth Factor alpha/genetics , Vascular Endothelial Growth Factor A/genetics , Visual Acuity/physiology
14.
Invest Ophthalmol Vis Sci ; 62(12): 17, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34542556

ABSTRACT

Purpose: Investigate the contribution of the Wnt pathway to vascular endothelial growth factor (VEGF)/anti-VEGF-mediated control of endothelial cell permeability. Methods: High glucose-treated primary human retinal endothelial cells (HRECs) were exposed to either VEGF, or VEGF and then anti-VEGF. Changes in gene expression were assayed by RNAseq and qRT-PCR. Permeability was monitored by electrical cell-substrate impedance sensing (ECIS). Approaches to activate the Wnt pathway included treatment with LiCl and overexpression of constitutively activated ß-catenin. ß-catenin-dependent transcriptional activity was monitored in HRECs stably expressing a TCF/LEF-driven reporter. Results: VEGF/anti-VEGF altered expression of genes encoding many members of the Wnt pathway. A subset of these genes was regulated in a way that is likely to contribute to control of the endothelial cell barrier. Namely, the VEGF-induced alteration of expression of such genes was reversed by anti-VEGF, and such adjustments occurred at times corresponding to changes in barrier function. While pharmacological and molecular approaches to activate the Wnt pathway had no effect on basal permeability, they suppressed VEGF-induced relaxation. Furthermore, anti-VEGF-mediated restoration of barrier function was unaffected by activation of the Wnt pathway. Conclusions: VEGF/anti-VEGF engages multiple members of the Wnt pathway, and activating this pathway enforces the endothelial barrier by attenuating VEGF-induced relaxation. These data suggest that FDA-approved agents such as LiCl may be an adjuvant to anti-VEGF therapy for patients afflicted with blinding conditions including diabetic retinopathy.


Subject(s)
Cell Membrane Permeability/physiology , Endothelial Cells/metabolism , Retinal Vessels/cytology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Wnt Signaling Pathway/physiology , Adult , Angiogenesis Inhibitors/pharmacology , Cells, Cultured , Electric Impedance , Gene Expression Regulation/physiology , Glucose/pharmacology , Humans , Lentivirus/genetics , Lithium Chloride/pharmacology , Male , Real-Time Polymerase Chain Reaction , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Transfection , beta Catenin/metabolism
15.
Med Sci Monit ; 27: e933601, 2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34456330

ABSTRACT

BACKGROUND The aim of this study was to investigate distribution rules of radial peripapillary capillaries (RPCs) density and correlations with retinal nerve fiber layers (RNFL) thickness in normal subjects. MATERIAL AND METHODS We included 78 eyes of 78 healthy subjects examined by optical coherence tomography angiography (OCTA). RPCs density and RNFL thickness were measured automatically. Distributions of RPCs density and RNFL thickness were analyzed at different locations. Correlations of these 2 parameters and relationship with large vessels were evaluated by Spearman test. RESULTS Average density for overall, peripapillary, and inside disc RCPs was 56.12±2.51%, 58.56±2.84%, and 60.16±4.01%, respectively. Overall and peripapillary RCPs density were positively correlated with RNFL thickness (r=0.595, P.


Subject(s)
Capillaries/cytology , Nerve Fibers/physiology , Optic Disk/blood supply , Retinal Vessels/cytology , Adult , Female , Follow-Up Studies , Healthy Volunteers , Humans , Male , Middle Aged , Visual Fields , Young Adult
16.
FASEB J ; 35(9): e21842, 2021 09.
Article in English | MEDLINE | ID: mdl-34418159

ABSTRACT

Retinopathy of prematurity (ROP) remains one of the major causes of blindness in children worldwide. While current ROP treatments are mostly disruptive to reduce proliferative neovascularization by targeting the hypoxic phase, protection against early hyperoxia-induced retinal vascular loss represents an effective therapeutic window, but no such therapeutic strategy is available. Built upon our recent demonstration that the protection against oxygen-induced retinopathy by adenosine A2A receptor (A2A R) antagonists is most effective when administered at the hyperoxia (not hypoxic) phase, we here uncovered the cellular mechanism underlying the A2A R-mediated protection against early hyperoxia-induced retinal vascular loss by reversing the inhibition of cellular proliferation via possibly multiple signaling pathways. Specifically, we revealed two distinct stages of the hyperoxia phase with greater cellular proliferation and apoptosis activities and upregulation of adenosine signaling at postnatal 9 day (P9) but reduced cellular activities and adenosine-A2A R signaling at P12. Importantly, the A2A R-mediated protection at P9 was associated with the reversal of hyperoxia-induced inhibition of progenitor cells at the peripheral retina at P9 and of retinal endothelial proliferation at P9 and P12. The critical role of cellular proliferation in the hyperoxia-induced retinal vascular loss was validated by the increased avascular areas by siRNA knockdown of the multiple signaling molecules involved in modulation of cellular proliferation, including activin receptor-like kinase 1, DNA-binding protein inhibitor 1, and vascular endothelial growth factor-A.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Cell Proliferation/drug effects , Hyperoxia/metabolism , Protective Agents/pharmacology , Receptor, Adenosine A2A/metabolism , Retinal Neovascularization , Retinal Vessels/drug effects , Activin Receptors, Type II/metabolism , Animals , Apoptosis/drug effects , Inhibitor of Differentiation Protein 1/metabolism , Mice , Neovascularization, Pathologic , Oxygen/adverse effects , Retina/cytology , Retina/drug effects , Retina/pathology , Retinal Vessels/cytology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta2/metabolism , Vascular Endothelial Growth Factor A/metabolism
17.
Mol Vis ; 27: 191-205, 2021.
Article in English | MEDLINE | ID: mdl-33953532

ABSTRACT

Purpose: There are reports that a b-isoform of vascular endothelial growth factor-A 165 (VEGFA165b) is predominant in normal human vitreous, switching to the a-isoform (VEGFA165a) in the vitreous of some diseased eyes. Although these isoforms appear to have a different ability to activate the VEGF receptor 2 (VEGFR2) in various endothelial cells, the nature of their ability to activate intracellular signaling pathways is not fully characterized, especially in retinal endothelial cells. We determined their activation potential for two key intracellular signaling pathways (MAPK, AKT) over complete dose-response curves and compared potential effects on the expression of several VEGFA165 target genes in primary human retinal microvascular endothelial cells (HRMECs). Methods: To determine full dose-response curves for the activation of MAPK (ERK1/2), AKT, and VEGFR2, direct in-cell western assays were developed using primary HRMECs. Potential differences in dose-response effects on gene expression markers related to endothelial cell and leukocyte adhesion (ICAM1, VCAM1, and SELE) and tight junctions (CLDN5 and OCLN) were tested with quantitative PCR. Results: Activation dose-response analysis revealed much stronger activation of MAPK, AKT, and VEGFR2 by the a-isoform at lower doses. MAPK activation in primary HRMECs displayed a sigmoidal dose-response to a range of VEGFA 165 a concentrations spanning 10-250 pM, which shifted higher into the 100-5,000 pM range with VEGFA 165 b. Similar maximum activation of MAPK was achieved by both isoforms at high concentrations. Maximum activation of AKT by VEGFA 165 b was only half of the maximum activation from VEGFA 165 a. At a lower intermediate dose, where VEGFA 165 a activated intracellular signaling stronger than VEGFA 165 b, the changes in VEGFA target gene expression were generally greater with VEGFA 165 a. Conclusions: In primary HRMECs, VEGFA 165 a could maximally activate MAPK and AKT at lower concentrations where VEGFA 165 b had relatively little effect. The timing for maximum activation of MAPK was similar for the isoforms, which is different from that reported for non-retinal endothelial cells. Although differences in VEGFA 165 a and VEGFA 165 b are limited to the sequence of their six C-terminal six amino acids, this results in a large difference in their ability to activate at least two key intracellular signaling pathways and VEGF-target gene expression in primary human retinal endothelial cells.


Subject(s)
Endothelium, Vascular/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinal Vessels/cytology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Claudin-5/genetics , E-Selectin/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation/physiology , Humans , Immunoblotting , Intercellular Adhesion Molecule-1/genetics , Occludin/genetics , Polymerase Chain Reaction , Protein Isoforms , Transcriptional Activation/physiology , Vascular Cell Adhesion Molecule-1/genetics
18.
Mol Med Rep ; 23(6)2021 Jun.
Article in English | MEDLINE | ID: mdl-33899114

ABSTRACT

Mangiferin is a prominent active component that can be derived from several traditional herbs, including Mangifera indica L., Anemarrhena asphodeloides Bge., and Belamcanda chinensis (L.) DC., which displays antidiabetic properties. Diabetic retinopathy (DR), a serious complication caused by diabetes, is the leading cause of blindness. The present study aimed to evaluate the beneficial effects of mangiferin on high glucose (HG)/hypoxia­induced rat retinal capillary endothelial cell (RRCEC) angiogenesis, as well as the underlying mechanisms. To establish an in vitro model of DR, RRCECs were exposed to 30 mM glucose and hypoxia. Following treatment with different doses of mangiferin (0.05, 0.1 or 0.2 µM), RRCEC viability, migration and angiogenesis were assessed by performing Cell Counting Kit 8, immunofluorescence, wound healing, Transwell and tube formation assays. Western blotting was conducted to evaluate protein expression levels. Furthermore, LY294002 and IGF­1, an inhibitor and activator of the PI3K/AKT/mTOR signaling pathway, respectively, were used to verify the potential mechanisms underlying mangiferin. The results demonstrated that mangiferin notably inhibited HG/hypoxia­induced RRCEC migration and angiogenesis. HG/hypoxia­induced upregulation of hypoxia­inducible factor­1α, vascular endothelial growth factor, matrix metallopeptidase (MMP)2 and MMP9 expression levels and the phosphorylation of PI3K, AKT and mTOR in RRCECs was significantly reversed following treatment with mangiferin. Additionally, further activation of the PI3K/AKT signaling pathway by IGF­1 inhibited the beneficial effects of mangiferin on RRCECs, whereas deactivation of the PI3K/AKT signaling pathway by LY294002 displayed the opposite results. Collectively, the results of the present study suggested that mangiferin suppressed RRCEC angiogenesis via modulating the PI3K/AKT/mTOR signaling pathway, which could serve as an effective treatment strategy for DR.


Subject(s)
Cell Hypoxia , Endothelial Cells/metabolism , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinases/metabolism , Retinal Vessels/cytology , Xanthones/pharmacology , Animals , Cell Movement , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/physiology , Glucose/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
19.
Exp Eye Res ; 207: 108585, 2021 06.
Article in English | MEDLINE | ID: mdl-33887222

ABSTRACT

Diabetic retinopathy (DR), characterized by intraretinal vessel formation, is a major complication in diabetes. Neovascularization is an important characteristic of DR, but its formation mechanism remains unclear. In this research, Malat1, miR-205-5p, and VEGF-A levels in high glucose (HG) treat-human retinal microvascular endothelial cells (hRMECs) was detected with qRT-PCR. CCK-8 assay, transwell assay, and tube formation assay was applied to access hRMEC viability, migration, and angiogenesis. Expression level of endothelial-mesenchymal transition (EndMT) markers (VE-cadherin, FSP1, and α-SMA) was detected by western blotting assay. Interaction among Malat1, miR-205-5p, and VEGF-A was confirmed by dual-luciferase reporter assay. Furthermore, in vivo DR mouse model was induced, and the effect of Malat1 on DR and EndMT markers was confirmed through hematoxylin-eosin (HE) staining and western blotting. As a result, Malat1 and VEGF-A was upregulated while miR-205-5p was suppressed under HG conditions. Malat1 could sponge miR-205-5p to regulate VEGF-A expression. Malat1 knockdown inhibited hRMEC proliferation, migration, and tube formation by targeting miR-205-5p under HG conditions. Furthermore, inhibition of Malat1 prevented the HG-induced EndMT process. In summary, Malat1 knockdown diminished hRMEC dysfunctions by regulating miR-205-5p/VEGF-A, providing a useful insight for exploring new therapeutic target for DR.


Subject(s)
Diabetic Retinopathy/prevention & control , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Glucose/pharmacology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Retinal Neovascularization/prevention & control , Vascular Endothelial Growth Factor A/genetics , Actins/metabolism , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Cells, Cultured , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Mice, Inbred C57BL , MicroRNAs/metabolism , Real-Time Polymerase Chain Reaction , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Vessels/cytology , S100 Calcium-Binding Protein A4/metabolism , Transfection , Vascular Endothelial Growth Factor A/metabolism
20.
Aging (Albany NY) ; 13(4): 5342-5357, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33536350

ABSTRACT

Diabetes-induced oxidative stress is vital in initiating neuronal damage in the diabetic retina, leading to diabetic retinopathy (DR). This study investigates the possible effects of coumestrol (CMS) on streptozotocin (STZ)-induced DR. First, we established a rat model of DR by STZ injection and a cell model involving high-glucose (HG) exposure of human retinal microvascular endothelial cells (hRMECs). We characterized the expression patterns of oxidative stress indicators, pro-inflammatory cytokines, and pro-apoptotic proteins in hRMECs. Polymerase chain reaction showed sirtuin 1 (SIRT1) to be poorly expressed in the retinal tissues of STZ-treated rats and HG-exposed hRMECs, but its expression was upregulated upon treatment with CMS treatment. Furthermore, CMS treatment attenuated the STZ-induced pathologies such as oxidative stress, inflammation, and cell apoptosis. Consistent with the in vivo results, CMS activated the expression of SIRT1, thereby inhibiting oxidative stress, inflammation, and apoptosis of HG-treated hRMECs. From these findings, we concluded that CMS ameliorated DR by inhibiting inflammation, apoptosis and oxidative stress through activation of SIRT1.


Subject(s)
Apoptosis/drug effects , Coumestrol/pharmacology , Diabetic Retinopathy/metabolism , Endothelial Cells/drug effects , Oxidative Stress/drug effects , Phytoestrogens/pharmacology , Retina/drug effects , Sirtuin 1/drug effects , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Glucose/toxicity , Humans , Inflammation/metabolism , Rats , Retina/metabolism , Retina/pathology , Retinal Vessels/cytology , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...