Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 709
Filter
1.
Exp Eye Res ; 247: 110046, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147191

ABSTRACT

Diabetic retinopathy, a leading cause of vision impairment, is marked by microvascular complications in the retina, including pericyte loss, a key indicator of early-stage disease. This study explores the therapeutic potential of exosomes derived from immortalized adipose-mesenchymal stem cells differentiated into pericyte-like cells in restoring the function of mouse retinal microvascular endothelial cells damaged by high glucose conditions, thereby contributing to the understanding of early diabetic retinopathy intervention strategies. To induce immortalized adipose-mesenchymal stem cells differentiation into pericyte-like cells, the study employed pericyte growth supplement. And confirmed the success of cell differentiation through the detection of α-smooth muscle actin and neural/glial antigen 2 expression by Western blot and immunofluorescence. Exosomes were isolated from the culture supernatant of immortalized adipose-mesenchymal stem cells using ultracentrifugation and characterized through Western blot for exosomal markers (CD9, CD81, and TSG101), transmission electron microscopy, and nanoparticle tracking analysis. Their influence on mouse retinal microvascular endothelial cells under high glucose stress was assessed through various functional assays. Findings revealed that exosomes, especially those from pericyte-like immortalized adipose-mesenchymal stem cells, were efficiently internalized by retinal microvascular endothelial cells and effectively counteracted high glucose-induced apoptosis. These exosomes also mitigated the rise in reactive oxygen species levels and suppressed the migratory and angiogenic properties of retinal microvascular endothelial cells, as demonstrated by Transwell and tube formation assays, respectively. Furthermore, they preserved endothelial barrier function, reducing hyperglycemia-induced permeability. At the molecular level, qRT-PCR analysis showed that exosome treatment modulated the expression of critical genes involved in angiogenesis (VEGF-A, ANG2, MMP9), inflammation (IL-1ß, TNF-α), gap junction communication (CX43), and cytoskeletal regulation (ROCK1), with the most prominent effects seen with exosomes from pericyte-like immortalized adipose-mesenchymal stem cells. High glucose increased the expression of pro-angiogenic and pro-inflammatory markers, which were effectively normalized post-exosome treatment. In conclusion, this research highlights the reparative capacity of exosomes secreted by pericyte-like differentiated immortalized adipose-mesenchymal stem cells in reversing the detrimental effects of high glucose on retinal microvascular endothelial cells. By reducing apoptosis, oxidative stress, inflammation, and abnormal angiogenic behavior, these exosomes present a promising avenue for therapeutic intervention in early diabetic retinopathy. Future studies can focus on elucidating the precise molecular mechanisms and exploring their translational potential in vivo.


Subject(s)
Blotting, Western , Cell Differentiation , Diabetic Retinopathy , Exosomes , Glucose , Mesenchymal Stem Cells , Pericytes , Retinal Vessels , Exosomes/metabolism , Pericytes/metabolism , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Retinal Vessels/cytology , Retinal Vessels/metabolism , Glucose/pharmacology , Diabetic Retinopathy/metabolism , Cells, Cultured , Adipose Tissue/cytology , Adipose Tissue/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/cytology , Microscopy, Electron, Transmission , Mice, Inbred C57BL , Apoptosis , Endothelial Cells/metabolism
2.
Exp Eye Res ; 247: 110062, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39187056

ABSTRACT

Exposure to VEGF-A165a over several days leads to a persistent dysfunction of the very tight barrier formed by immortalized endothelial cells of the bovine retina (iBREC). Elevated permeability of the barrier is indicated by low cell index values determined by electric cell-substrate impedance measurements, by lower amounts of claudin-1, and by disruption of the homogenous and continuous staining of vascular endothelial cadherin at the plasma membrane. Because of findings that suggest modulation of VEGF-A's detrimental effects on the inner blood-retina barrier by the angiogenic growth factor angiopoietin-2, we investigated in more detail in vitro whether this growth factor indeed changes the stability of the barrier formed by retinal endothelial cells or modulates effects of VEGF-A. In view of the clinical relevance of anti-VEGF therapy, we also studied whether blocking VEGF-A-driven signaling is sufficient to prevent barrier dysfunction induced by a combination of both growth factors. Although angiopoietin-2 stimulated proliferation of iBREC, the formed barrier was not weakened at a concentration of 3 nM: Cell index values remained high and expression or subcellular localization of claudin-1 and vascular endothelial cadherin, respectively, were not affected. Angiopoietin-2 enhanced the changes induced by VEGF-A165a and this was more pronounced at lower concentrations of VEGF-A165a. Specific inhibition of the VEGF receptors with tivozanib as well as interfering with binding of VEGF-A to its receptors with bevacizumab prevented the detrimental effects of the growth factors; dual binding of angiopoietin-2 and VEGF-A by faricimab was marginally more efficient. Uptake of extracellular angiopoietin-2 by iBREC can be efficiently prevented by addition of faricimab which is also internalized by the cells. Exposure of the cells to faricimab over several days stabilized their barrier, confirming that inhibition of VEGF-A signaling is not harmful to this cell type. Taken together, our results confirm the dominant role of VEGF-A165a in processes resulting in increased permeability of retinal endothelial cells in which angiopoietin-2 might play a minor modulating role.


Subject(s)
Angiopoietin-2 , Blood-Retinal Barrier , Cadherins , Cell Proliferation , Vascular Endothelial Growth Factor A , Animals , Cattle , Angiogenesis Inhibitors/pharmacology , Angiopoietin-2/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Proliferation/drug effects , Cells, Cultured , Claudin-1/metabolism , Electric Impedance , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/cytology , Peptide Fragments , Retinal Vessels/cytology , Retinal Vessels/metabolism , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
Cells Tissues Organs ; 213(5): 368-381, 2024.
Article in English | MEDLINE | ID: mdl-38320535

ABSTRACT

INTRODUCTION: Ocular diseases pose a significant health concern for donkeys. However, studies examining the microanatomy and cell populations of the donkey retina are scarce. The current study aimed to describe the vascular pattern of the donkey retina and document its cellular components. METHODS: The donkey retina specimens were obtained from different retinal regions and prepared for semithin sectioning and immunohistochemistry. RESULTS: The donkey has a paurangiotic retina in which retinal vessels are confined to a narrow area around the optic disc. Glial cells coexist with the blood vessels being very numerous in the vascular region and become scanty in the avascular ones. S-100-positive astrocytes could be observed in these avascular areas. Ganglion cells are organized in a single layer with the least population existing in the peripheral retina. Acidic fibroblast growth factor (AFGF) is immunoreactive in amacrine and ganglion cells. A subpopulation of amacrine cells reacted strongly to tyrosine hydroxylase (TH), and others reacted positively to S-100 protein. Ganglion cell nuclei exhibited a strong immunoreactivity to S-100 protein as well. Furthermore, glial fibrillary acidic protein (GFAP) is used to identify Müller cells that extend their processes across the retina from the inner to the outer limiting membrane. CONCLUSIONS: In conclusion, our findings provide novel insights into the normal retinal organization. The donkey retina shows the characteristic expression of immunohistochemical markers for the major cell types. In addition, the distribution of glial cells is comparable between the vascular and avascular regions.


Subject(s)
Equidae , Immunohistochemistry , Neuroglia , Retina , Animals , Neuroglia/metabolism , Neuroglia/cytology , Retina/metabolism , Retina/cytology , Neurons/metabolism , Neurons/cytology , Retinal Vessels/metabolism , Retinal Vessels/cytology , Glial Fibrillary Acidic Protein/metabolism , Cell Differentiation
4.
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
5.
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
6.
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
7.
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
8.
J Biol Chem ; 295(14): 4647-4660, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32086377

ABSTRACT

Vascular endothelial growth factor (VEGF) contributes to blood-retinal barrier (BRB) dysfunction in several blinding eye diseases, including diabetic retinopathy. Signaling via the secreted protein norrin through the frizzled class receptor 4 (FZD4)/LDL receptor-related protein 5-6 (LRP5-6)/tetraspanin 12 (TSPAN12) receptor complex is required for developmental vascularization and BRB formation. Here, we tested the hypothesis that norrin restores BRB properties after VEGF-induced vascular permeability in diabetic rats or in animals intravitreally injected with cytokines. Intravitreal co-injection of norrin with VEGF completely ablated VEGF-induced BRB permeability to Evans Blue-albumin. Likewise, 5-month diabetic rats exhibited increased permeability of FITC-albumin, and a single norrin injection restored BRB properties. These results were corroborated in vitro, where co-stimulation of norrin with VEGF or stimulation of norrin after VEGF exposure restored barrier properties, indicated by electrical resistance or 70-kDa RITC-dextran permeability in primary endothelial cell culture. Interestingly, VEGF promoted norrin signaling by increasing the FZD4 co-receptor TSPAN12 at cell membranes in an MAPK/ERK kinase (MEK)/ERK-dependent manner. Norrin signaling through ß-catenin was required for BRB restoration, but glycogen synthase kinase 3 α/ß (GSK-3α/ß) inhibition did not restore BRB properties. Moreover, levels of the tight junction protein claudin-5 were increased with norrin and VEGF or with VEGF alone, but both norrin and VEGF were required for enriched claudin-5 localization at the tight junction. These results suggest that VEGF simultaneously induces vascular permeability and promotes responsiveness to norrin. Norrin, in turn, restores tight junction complex organization and BRB properties in a ß-catenin-dependent manner.


Subject(s)
Blood-Retinal Barrier/metabolism , Capillary Permeability/drug effects , Eye Proteins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Animals , Blood-Retinal Barrier/drug effects , Cattle , Claudin-5/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Rats , Rats, Long-Evans , Retina/metabolism , Retinal Vessels/cytology , Retinal Vessels/metabolism , Signal Transduction/drug effects , Tetraspanins/genetics , Tetraspanins/metabolism , Up-Regulation/drug effects , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
9.
Angiogenesis ; 24(1): 7-11, 2021 02.
Article in English | MEDLINE | ID: mdl-33033849

ABSTRACT

Mouse retinal vasculature is a well-recognized and commonly used animal model for angiogenesis and microvascular remodeling. Morphological features of retinal vasculature reflect the vessel's biological functions, and are critical in understanding the physiological and pathological process of vascular development and disease. Here we developed a comprehensive software, Vessel Tech, using retinal vasculature images of postnatal mice. This pipeline can automatically process retinal vascular images, reconstruct vessel network with high accuracy and assess global and local vascular characteristics based on the recent machine-learning techniques. The development of Vessel Tech provides a powerful tool for vascular biologists.


Subject(s)
Retinal Vessels/diagnostic imaging , Software , Animals , Endothelial Cells/cytology , Image Processing, Computer-Assisted , Mice , Neural Networks, Computer , Retinal Vessels/cytology
10.
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
11.
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
12.
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
13.
J Vasc Res ; 58(1): 49-57, 2021.
Article in English | MEDLINE | ID: mdl-33022674

ABSTRACT

The neonatal mouse retinal vascularization model has been widely used in the vascular biology field to investigate mechanisms of angiogenesis and arterial-venous fate specification during blood vessel formation and maturation. Recent advances in next-generation sequencing can further elucidate mechanisms of blood vessel formation and remodeling in this, as well as other, vascular development models. However, an optimized method for isolating retinal endothelial cells that limits tissue digestion-induced cell damage is required for next-generation sequencing applications. In this study, we established a method for isolating neonatal retinal endothelial cells that optimizes cell viability and purity. The CD31+/CD45- endothelial cell population was fluorescence-activated cell sorting (FACS)-isolated from digested postnatal retinas, found to be highly enriched for endothelial cell gene expression, and exhibited no change in viability for 60 min post-FACS. Thus, this method for retinal endothelial cell isolation is compatible with next-generation sequencing applications. Combining this isolation method with next-generation sequencing will enable further delineation of mechanisms underlying vascular development and maturation.


Subject(s)
Cell Separation , Endothelial Cells/physiology , Flow Cytometry , Retinal Vessels/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Survival , Endothelial Cells/metabolism , Gene Expression Regulation , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Mice , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Real-Time Polymerase Chain Reaction , Time Factors , Workflow
14.
Exp Eye Res ; 202: 108300, 2021 01.
Article in English | MEDLINE | ID: mdl-33065089

ABSTRACT

Diabetic retinopathy (DR) is the leading cause of visual impairment and acquired blindness among adults worldwide. Retinal microvascular pericyte deficiency is one of the earliest pathological changes associated with DR, and long noncoding RNA myocardial infarction-associated transcript (MIAT) has been implicated as a crucial regulator of microvascular dysfunction in DR. Pyroptosis is a caspase-1-dependent proinflammatory form of cell death, and in the present study, we investigated the potential pyroptosis of primary human retinal pericytes (HRPCs) and the mechanism by which MIAT is involved in this process. We applied advanced glycation end product modified bovine serum albumin (AGE-BSA) to simulate the DR environment. The results suggested that AGE-BSA induced the active cleavage of caspase-1 and gasdermin D, the release of IL-1ß, IL-18 and LDH, and reduced cell viability, which was prevented by the inhibition of caspase-1, indicating the occurrence of caspase-1-mediated pyroptosis in HRPCs. Immunofluorescence images revealed the phenotypic characteristics of pyroptosis, including pyknosis, swelling and hyperpermeability in plasmolemma. MIAT and CASP1 expression were substantially increased, while that of miR-342-3p was decreased in AGE-BSA-treated HRPCs. MIAT knockdown inhibited pyroptosis in HRPCs, which was reinforced by cotreatment with miR-342-3p mimic but relieved by cotreatment with miR-342-3p inhibitor. Furthermore, HRPC pyroptosis was inhibited by treatment with the miR-342-3p mimic alone but enhanced by the miR-342-3p inhibitor. Luciferase reporter assay results demonstrated binding between MIAT and miR-342-3p, as well as between miR-342-3p and CASP1. MIAT antagonized the effect of miR-342-3p on the depression of its target CASP1 and promoted AGE-BSA-induced pericyte pyroptosis. These findings may promote a better understanding of retinal pericyte depletion pathogenesis and the development of new therapeutic strategies for the treatment of diabetic retinopathy.


Subject(s)
Caspase 1/metabolism , Diabetic Retinopathy/metabolism , MicroRNAs/metabolism , Pericytes/physiology , Pyroptosis/physiology , RNA, Long Noncoding/physiology , Retinal Vessels/cytology , Blotting, Western , Cell Survival/physiology , Cells, Cultured , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/physiology , Genes, Reporter/genetics , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
15.
Exp Eye Res ; 205: 108490, 2021 04.
Article in English | MEDLINE | ID: mdl-33607076

ABSTRACT

Microglia and Müller cells (MCs) are believed to be critically involved in hypoxia-induced blood-retinal barrier (BRB) disruption, which is a major pathogenic factor of various retinopathies. However, the underlying mechanism remains poorly defined. The inner BRB (iBRB) is primarily formed of microvascular endothelial cells (ECs) with tight junction (TJ), which are surrounded and supported by retinal glial cells. We developed a novel in vitro iBRB model sheet by sandwiching Transwell membrane with layered mouse brain microvascular ECs (bEnd.3) and mouse retinal MCs (QMMuC-1) on each side of the membrane. Using this model, we tested the hypothesis that under hypoxic condition, activated microglia produce inflammatory cytokines such as interleukin (IL)-1ß, which may promote vascular endothelial growth factor (VEGF) production from MCs, leading to TJ disruption. The iBRB model cell sheets were exposed to 1% oxygen for 6 h with or without mouse brain microglia (BV2) or IL-1ß. TJ structure and function were examined by zonula occludens (ZO)-1 immunostaining and fluorescein isothiocyanate permeability assay, respectively. Relative gene expression of IL-1ß in BV2 under normoxic and hypoxic conditions was examined by real-time reverse transcription-polymerase chain reaction. VEGF protein concentration in QMMuC-1 supernatants was measured by enzyme-linked immunosorbent assay. The bEnd.3 cell sheet incubated with BV2 in hypoxic condition or with IL-1ß in normoxic condition showed abnormal localization of ZO-1 and aberrated barrier function. Under normoxic condition, EC-MC iBRB model cell sheet showed lower permeability than bEnd.3 cell sheet. Under hypoxic conditions, the barrier function of EC-MC iBRB model cell sheet was more deteriorated compared to bEnd.3 cell sheet. Under hypoxic condition, incubation of EC-MC iBRB model cell sheet with BV2 cells or IL-1ß significantly increased barrier permeability, and hypoxia-treated BV2 cells expressed significantly higher levels of IL-1ß mRNA. Incubation of QMMuC-1 with IL-1ß increased VEGF production. These results suggest that under hypoxic condition, microglia are activated to release proinflammatory cytokines such as IL-1ß that promote VEGF production from MCs, leading to disruption of iBRB function. Modulating microglia and MCs function may be a novel approach to treat hypoxia-induced retinal BRB dysfunction.


Subject(s)
Blood-Retinal Barrier/physiology , Capillary Permeability/physiology , Endothelium, Vascular/metabolism , Ependymoglial Cells/physiology , Hypoxia/metabolism , Microglia/physiology , Tight Junctions/metabolism , Animals , Cell Line , Cells, Cultured , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Gene Expression , Immunohistochemistry , Interleukin-1beta/genetics , Mice , Models, Biological , RNA, Messenger/genetics , Retinal Vessels/cytology , Vascular Endothelial Growth Factor A/metabolism , Zonula Occludens-1 Protein
16.
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
17.
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
18.
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
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.
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
SELECTION OF CITATIONS
SEARCH DETAIL