Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Endocr J ; 2024 May 28.
Article in English | MEDLINE | ID: mdl-38811189

ABSTRACT

Endothelial-to-mesenchymal transition (EndMT) is a pivotal event in diabetic retinopathy (DR). This study explored the role of circRNA zinc finger protein 532 (circZNF532) in regulating EndMT in DR progression. Human retinal microvascular endothelial cells (HRMECs) were exposed to high glucose (HG) to induce the DR cell model. Actinomycin D-treated HRMECs were used to confirm the mRNA stability of phosphoinositide-3 kinase catalytic subunit δ (PIK3CD). The interaction between TATA-box-binding protein-associated factor 15 (TAF15) and circZNF532/PIK3CD was subsequently analyzed using RNA immunoprecipitation (RIP), RNA pull-down. It was found that HG treatment accelerated EndMT process, facilitated cell migration and angiogenesis, and enhanced PIK3CD and p-AKT levels in HRMECs, whereas si-circZNF532 transfection neutralized these effects. Further data showed that circZNF532 recruited TAF15 to stabilize PIK3CD, thus elevating PIK3CD expression. Following rescue experiments suggested that PIK3CD overexpression partially negated the inhibitory effect of circZNF532 silencing on EndMT, migration, and angiogenesis of HG-treated HRMECs. In conclusion, our results suggest that circZNF532 recruits TAF15 to stabilize PIK3CD, thereby facilitating EndMT in DR.

2.
Semin Ophthalmol ; 39(4): 312-319, 2024 May.
Article in English | MEDLINE | ID: mdl-38192082

ABSTRACT

The purpose of our study was to investigate the role of TGF-ß1 in the endothelial-to-mesenchymal transition (EndoMT) and fibrosis in high glucose (HG)-treated human retinal microvascular endothelial cells (HRMECs). HRMECs were cultured not only under normal glucose (NG) conditions with or without TGF-ß1, but also under HG conditions with or without the TGF-ß1 inhibitor SB431542. The expression of TGF-ß1 was detected by real time-PCR and enzyme-linked immunosorbent assay. Morphological changes and migration of the HRMECs were observed using electron microscopy and scratch-wound assay. Endothelial markers, such as CD31 and vascular endothelial (VE)-cadherin, and the acquisition of fibrotic markers, such as alpha smooth muscle actin (α-SMA) and fibroblast-specific protein-1 (FSP-1), were determined by immunofluorescent staining and western blot. The level of TGF-ß1 was significantly upregulated in HG-treated HRMECs. And HG stimulation promoted obvious morphological changes and the migration ability in HRMECs. Our results also demonstrated increased expression of α-SMA and FSP-1, and decreased expression of CD31 and VE-cadherin, in HG-treated HRMECs. These EndoMT-related changes were promoted by TGF-ß1 and abrogated by SB431542. The results of this study demonstrated the important role of TGF-ß1 in HG-induced vitreoretinal fibrosis. EndoMT is likely to be involved in the associated effects.


Subject(s)
Benzamides , Dioxoles , Endothelial Cells , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Fibrosis , Glucose/metabolism , Glucose/pharmacology , Epithelial-Mesenchymal Transition/physiology
3.
Cell Signal ; 113: 110955, 2024 01.
Article in English | MEDLINE | ID: mdl-38084838

ABSTRACT

Diabetic retinopathy (DR) is a leading cause of blindness, and ferroptosis may be an essential component of the pathological process of DR. In this study, we aimed to screen five hub genes (TLR4, CAV1, HMOX1, TP53, and IL-1B) using bioinformatics analysis and experimentally verify their expression and effects on ferroptosis and cell function. The online Gene Expression Omnibus microarray expression profiling datasets GSE60436 and GSE1025485 were selected for investigation. Ferroptosis-related genes that might be differentially expressed in DR were identified. Then, Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and protein-protein interaction (PPI) network analyses were conducted to characterize the differentially-expressed ferroptosis-related genes. After tissue-specific analyses and external dataset validation of hub genes, the mRNA and protein levels of hub genes in retinal microvascular endothelial cells (HRMECs) symbiotic with high glucose were verified using real-time quantitative PCR (qRT-PCR) and immunocytochemistry (ICC). Finally, hub genes were knocked down using siRNA, and changes in ferroptosis and cell function were observed. Based on the differential expression analysis, 19 ferroptosis-related genes were identified. GO and KEGG enrichment analyses showed that ferroptosis-related genes were significantly enriched in reactive oxygen species metabolic processes, necrotic cell death, hypoxia responses, iron ion responses, positive regulation of cell migration involved in sprouting angiogenesis, NF-kappa B signaling pathway, ferroptosis, fluid shear stress, and atherosclerosis. Subsequently, PPI network analysis and critical module construction were used to identify five hub genes. Based on bioinformatics analysis of mRNA microarrays, qRT-PCR confirmed higher mRNA expression of five genes in the DR model, and immunocytochemistry confirmed their higher protein expression. Finally, siRNA interference was used to verify the effects of five genes on ferroptosis and cell function. Based on bioinformatics analysis, five potential genes related to ferroptosis were identified, and their upregulation may affect the onset or progression of DR. This study sheds new light on the pathogenesis of DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Ferroptosis , Humans , Diabetic Retinopathy/genetics , Endothelial Cells , Ferroptosis/genetics , Computational Biology , RNA, Messenger , RNA, Small Interfering
4.
Int J Mol Sci ; 23(13)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35806359

ABSTRACT

The pathophysiology of retinopathy of prematurity (ROP) is postulated to first involve delayed intraretinal vascularization, followed by intravitreal neovascularization (IVNV). Although intravitreal agents that reduce the bioactivity of vascular endothelial growth factor (VEGF) are used to treat IVNV, concerns exist regarding their effects on intraretinal vascularization. In an experimental ROP model, VEGF receptor 2 (VEGFR2) knockdown in retinal endothelial cells reduced IVNV and promoted intraretinal vascularization, whereas knockdown of a downstream effector, signal transducer and activator of transcription 3 (STAT3) in retinal endothelial cells only reduced IVNV. In this study, we tested the hypothesis that the different pathways involved in VEGF-triggered VEGFR2 signaling and VEGF-triggered STAT3 signaling in retinal endothelial cells would allow us to delineate signaling pathways involved in IVNV from those involved in intraretinal vascularization in ROP. To address our hypothesis, we used RNA-sequencing and pathway enrichment analysis to determine changes in the transcriptome of cultured human retinal microvascular endothelial cells (HRMECs). Of the enriched pathways, inactivation of oncostatin M signaling was predicted by either KDR or STAT3 knockdown in the presence of VEGF. Activation of kinetochore metaphase signaling was predicted by KDR knockdown, whereas inactivation was predicted by STAT3 knockdown in the presence of VEGF. Inactivation of signaling by the Rho family of GTPases was predicted by KDR knockdown, but activation was predicted by STAT3 knockdown in the presence of VEGF. Taken together, our data identified unique signaling pathway differences between VEGF-triggered VEGFR2 and VEGF-triggered STAT3 in HRMECs that might have implications in ROP.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Infant, Newborn , Neovascularization, Pathologic/metabolism , RNA-Seq , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
5.
Biochem Biophys Res Commun ; 533(4): 1406-1412, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33092793

ABSTRACT

Dysregulation of endothelial cell proliferation and migration are hallmarks of angiogenic diseases. Among them, excessive ocular angiogenesis is a major cause of blindness. Vascular endothelial growth factor (VEGF)-VEGF receptor 2 (VEGFR2) signaling plays crucial roles in angiogenesis, endothelial cell proliferation and migration. Here, we showed that ankyrin repeat and FYVE domain containing 1 (ANKFY1), a Rab5-GTP-interacting protein, is required for retinal endothelial cell proliferation and migration. ANKFY1 knockdown significantly suppressed cell growth of human retinal microvascular endothelial cells (HRMECs) in the presence or absence of VEGF. HRMEC migration was also inhibited by depletion of ANKFY1. Western blot analysis showed that ANKFY1 knockdown reduced cell surface VEGFR2 level. In contrast, qRT-PCR analysis indicated that ANKFY1 knockdown had no effect on VEGFR2 mRNA levels. We also found that the attenuation of the protein kinase B/endothelial nitric oxide synthase (Akt/eNOS) pathway in ANKFY1 knockdown HRMECs. In conclusion, our findings revealed novel functions of ANKFY1 in cell growth and migration of retinal endothelial cells.


Subject(s)
Endothelium, Vascular/cytology , Phosphate-Binding Proteins/metabolism , Retina/cytology , Retina/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/metabolism , Gene Knockdown Techniques , Humans , Integrin beta1/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphate-Binding Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Gene ; 760: 144992, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32721474

ABSTRACT

BACKGROUND AND AIM: Diabetic retinopathy is a severe diabetic complication and a major cause of blindness. In this study, we explored the role of circ_0001879 in retinal vascular dysfunction under diabetic conditions. METHODS: Human retinal microvascular endothelial cells (HRMECs) were divided into normal glucose group (NG, 5.5 mmol/L d-glucose), high glucose group (HG, 25 mmol/L d-glucose), and osmotic control group (5.5 mmol/L d-glucose + 19.5 mmol/L mannitol). The expression of circ_0001879 and miR-30-3p was assessed via qRT-PCR. The circ_0001879/miR-30-3p roles in retinal vascular dysfunction were investigated through Cell Counting Kit-8 and Transwell assay. Bioinformatics analysis and luciferase reporter assays were applied to examine interactions between circ_0001879 and miR-30-3p in HRMECs. RESULTS: The relative circ_0001879 expression was remarkably increased in diabetic retinas group than that in the control group. Silencing circ_0001879 suppressed the proliferation and migration of HRMECs under high-glucose conditions. In addition, circ_0001879 acted as a binding platform and miRNA sponge for miR-30-3p. Circ_0001879 modulated the function of HRMECs via targeting miR-30-3p. CONCLUSION: Silencing circ_0001879 inhibited the proliferation and migration of HRMECs under high-glucose conditions via modulating miR-30-3p, which might shed new light on a novel potentially marker and molecular therapeutic target for diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/pathology , Glucose/administration & dosage , MicroRNAs/genetics , Retinal Vessels/pathology , Animals , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glucose/metabolism , Humans , Male , MicroRNAs/metabolism , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Vessels/drug effects , Retinal Vessels/metabolism
7.
Biosci Rep ; 40(7)2020 07 31.
Article in English | MEDLINE | ID: mdl-32519748

ABSTRACT

OBJECTIVE: Diabetic retinopathy (DR) is one of the most severe and common complications of diabetes mellitus. The present study aimed to investigate the molecular mechanism of MEG3, miR-19b and SOCS6 in human retinal microvascular endothelial cells (hRMECs) under high glucose conditions. METHODS: HRMECs were cultured in 5 or 30 mM D-glucose medium. qRT-PCR and Western blotting were used to determine the mRNA expression and protein levels. MTT assay and flow cytometry analysis were performed to detect the viability and apoptosis of hRMECs, respectively. TNF-α, IL-6 and IL-1ß levels in cell supernatants were detected by ELISA. The activity of caspase-3/7 was also determined. A luciferase reporter assay was performed to confirm the targeting relationship between miR-19b and SOCS6, as well as MEG3 and miR-19b. RESULTS: Our study demonstrated that miR-19b was increased and SOCS6 was decreased in HG-induced hRMECs. Knockdown of SOCS6 inhibited cell viability and reversed the promotion of cell viability induced by knockdown of miR-19b. Additionally, miR-19b directly targeted and negatively regulated SOCS6. Moreover, miR-19b promoted the cell apoptosis rate and caspase-3/7 activity and increased inflammatory factors through the SOCS6-mediated JAK2/STAT3 signalling pathway. In addition, MEG3 attenuated HG-induced apoptosis of hRMECs by targeting the miR-19b/SOCS6 axis. CONCLUSION: These findings indicate that MEG3 inhibited HG-induced apoptosis and inflammation by regulating the miR-19b/SOCS6 axis through the JAK2/STAT3 signalling pathway in hRMECs. Thus, these findings might provide a new target for the treatment of DR.


Subject(s)
Diabetic Retinopathy/genetics , Endothelial Cells/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Cell Line , Culture Media/metabolism , Diabetic Retinopathy/immunology , Diabetic Retinopathy/pathology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Gene Knockdown Techniques , Glucose/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Janus Kinase 2/metabolism , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Microvessels/cytology , Retinal Vessels/cytology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/metabolism
8.
Nutrients ; 11(3)2019 Mar 04.
Article in English | MEDLINE | ID: mdl-30836664

ABSTRACT

Retinal apoptosis plays a critical role in the progression of diabetic retinopathy (DR), a common diabetic complication. Currently, the tight control of blood glucose levels is the standard approach to prevent or delay the progression of DR. However, prevalence of DR among diabetic patients remains high. Focusing on natural nutrients or herbal medicines that can prevent or delay the onset of diabetic complications, we administered an ethanol extract of the aerial portion of Osteomeles schwerinae (OSSCE), a Chinese herbal medicine, over a period of 17 weeks to spontaneously diabetic Torii (SDT) rats. OSSCE was found to ameliorate retinal apoptosis through the regulation of advanced glycation end product (AGE) accumulation, oxidative stress, and mitochondrial function via the inhibition of NF-κB activity, in turn, through the downregulation of PKCδ, P47phox, and ERK1/2. We further demonstrated in 25 mM glucose-treated human retinal microvascular endothelial cells (HRMECs) that hyperoside (3-O-galactoside-quercetin), quercitrin (3-O-rhamnoside-quercetin), and 2″-O-acetylvitexin (8-C-(2″-O-acetyl-glucoside)-apigenin) were the active components of OSSCE that mediated its pharmacological action. Our results provide evidence that OSSCE is a powerful agent that may directly mediate a delay in the development or disease improvement in patients of DR.


Subject(s)
Diabetes Mellitus/drug therapy , Diabetic Retinopathy/drug therapy , Drugs, Chinese Herbal/pharmacology , Ethanol/pharmacology , Plant Extracts/pharmacology , Animals , Apoptosis/drug effects , Diabetes Mellitus/etiology , Diabetic Retinopathy/prevention & control , Endothelial Cells/drug effects , Glycation End Products, Advanced/drug effects , Humans , Male , Rats , Retina/drug effects
9.
J Ethnopharmacol ; 185: 162-70, 2016 Jun 05.
Article in English | MEDLINE | ID: mdl-26988565

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tauroursodeoxycholic acid (TUDCA), one of the main ingredients from bear gall which hold "Clearing heat and detoxification, Removing liver fire for improving eyesight" functions, is formed by the conjugation of ursodeoxycholic acid (UDCA) with taurine. However, the limited information of TUDCA on protecting diabetic retinopathy (DR) has been known. The present study was conducted to evaluate the protection of TUDCA on high glucose-induced human retinal microvascular endothelial cells (HRMECs) dysfunction and streptozotocin (STZ)-induced diabetic retinopathy (DR) rats and the possible mechanism underlying was also explored. MATERIALS AND METHODS: The proliferation of high glucose-induced HRMECs was determined by MTT assay. DR rats' model was established by an administration of high-glucose-fat diet and an intraperitoneal injection of STZ (30mg/kg). The cell supernatant and rats' serum were collected for the assays of NO content by ELISA kits. Retinas were stained with hematoxylin and eosin (HE) to observe pathological changes. Immunohistochemical assay was applied to examine the protein expression of ICAM-1, NOS, NF-κB p65 and VEGF in rat retinas. Furthermore, western blot analysis was carried out to examine the protein expression of ICAM-1, NOS, NF-κB p65 and VEGF in high glucose-induced HRMECs. RESULTS: After treating with TUDCA, high glucose-induced HRMECs proliferation could be significantly inhibited. TUDCA (5.0µM, 25.0µM and 125.0µM) could decrease NO content in high glucose-induced HRMECs. Furthermore, TUDCA (500mg/kg/d and 250mg/kg/d) also decrease NO content in serum of DR rats. Additionally, both immunocytochemistry analysis and western blot analysis showed that the over-expression of ICAM-1, NOS, NF-κB p65 and VEGF were significantly decreased by TUDCA. CONCLUSION: The data indicated that TUDCA could ameliorate DR by decreasing NO content and down-regulating the protein expression of ICAM-1, NOS, NF-κB p65 and VEGF. Thus, our experimental results suggested that TUDCA might be a potential drug for the prevention and treatment of DR.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Retinopathy/prevention & control , Endothelial Cells/drug effects , Glucose/toxicity , Retinal Vessels/cytology , Taurochenodeoxycholic Acid/pharmacology , Animals , Gene Expression Regulation, Enzymologic , Humans , Intercellular Adhesion Molecule-1 , Male , Mice , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
10.
Exp Eye Res ; 135: 67-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25913458

ABSTRACT

Retinal diseases characterized by pathologic retinal angiogenesis are the leading causes of blindness worldwide. Although therapies directed toward vascular endothelial growth factor (VEGF) represent a significant step forward in the treatment of proliferative retinopathies, further improvements are needed. In the last few years, an intense research activity has focused around the use of herbal and traditional natural medicines as an alternative for slowing down the progression of proliferative retinopathies. In the present study, we investigated the antiangiogenic effects of acetyl-11-keto-ß-boswellic acid (AKBA), one of the active principles derived from the plant Boswellia serrata, used in Ayurvedic systems of medicine. We studied the antiangiogenic properties of AKBA using the mouse model of oxygen-induced retinopathy (OIR), which mimics the neovascular response seen in human retinopathy of prematurity. We first evaluated the effects of subcutaneously administered AKBA on the expression/activity of proteins which are known to play a role in the OIR model. In the retina, AKBA increased expression and activity of Src homology region 2 domain-containing phosphatase 1 and reduced the phosphorylation of the transcription factor signal transducer and activator of transcription 3 (STAT3) as well as VEGF expression and VEGF receptor (VEGFR)-2 phosphorylation. Likely as a result of these effects, AKBA significantly reduced retinal neovascularization in OIR mice without affecting retinal cell survival and retinal function. Using retinal explants cultured in hypoxia and an activator of STAT3 phosphorylation, we showed that the AKBA-induced inhibition of VEGFR-2 phosphorylation is likely to be mediated by a mechanism depending on an SHP-1/STAT3/VEGF axis. In the OIR model, neovascularization results from the activation of retinal endothelial cells, therefore we evaluated whether AKBA affected the angiogenic response of human retinal microvascular endothelial cells (HRMECs). We observed that AKBA reduced proliferation, migration and tube formation in HRMECs stimulated with exogenous VEGF, while it reduced migration and tube formation in untreated HRMECs. Taken together, our results demonstrate the antiangiogenic effects of AKBA in a model of pathologic neovascularization, providing a rationale for further investigation of AKBA as a promising therapeutic agent to reduce the impact of proliferative retinopathies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Retinal Neovascularization/drug therapy , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Electroretinography/drug effects , Endothelial Cells/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Acta Pharm Sin B ; 4(6): 447-53, 2014 Dec.
Article in English | MEDLINE | ID: mdl-26579416

ABSTRACT

In this work, retinal penetration of fluorescein was achieved in vitro by covalent attachment of taurine to fluorescein, yielding the F-Tau conjugate. Nuclear magnetic resonance (NMR) and high resolution mass spectrometry (HRMS) were used to confirm the successful synthesis of F-Tau. The cellular uptake of F-Tau in adult retinal pigment epithelial cells (ARPE-19) and human retinal microvascular endothelial cells (hRMECs) was visualized via confocal scanning microscopy. The results indicated an improvement of solubility and a reduction of logP of F-Tau compared with fluorescein. As compared with fluorescein, F-Tau showed little toxicity, and was retained longer by cells in uptake experiments. F-Tau also displayed higher transepithelial permeabilities than fluorescein in ARPE-19 and hRMECs monolayer cells (P<0.05). These results showed that taurine may be a useful ligand for targeting small-molecule hydrophobic pharmaceuticals into the retina.

12.
Acta Pharmaceutica Sinica B ; (6): 447-453, 2014.
Article in English | WPRIM (Western Pacific) | ID: wpr-329703

ABSTRACT

In this work, retinal penetration of fluorescein was achieved in vitro by covalent attachment of taurine to fluorescein, yielding the F-Tau conjugate. Nuclear magnetic resonance (NMR) and high resolution mass spectrometry (HRMS) were used to confirm the successful synthesis of F-Tau. The cellular uptake of F-Tau in adult retinal pigment epithelial cells (ARPE-19) and human retinal microvascular endothelial cells (hRMECs) was visualized via confocal scanning microscopy. The results indicated an improvement of solubility and a reduction of logP of F-Tau compared with fluorescein. As compared with fluorescein, F-Tau showed little toxicity, and was retained longer by cells in uptake experiments. F-Tau also displayed higher transepithelial permeabilities than fluorescein in ARPE-19 and hRMECs monolayer cells (P<0.05). These results showed that taurine may be a useful ligand for targeting small-molecule hydrophobic pharmaceuticals into the retina.

SELECTION OF CITATIONS
SEARCH DETAIL
...