Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 41
1.
BMC Genomics ; 25(1): 352, 2024 Apr 09.
Article En | MEDLINE | ID: mdl-38594623

BACKGROUND: Posterior capsular opacification (PCO) is the main reason affecting the long-term postoperative result of cataract patient, and it is well accepted that fibrotic PCO is driven by transforming growth factor beta (TGFß) signaling. Ferroptosis, closely related to various ocular diseases, but has not been explored in PCO. METHODS: RNA sequencing (RNA-seq) was performed on both TGF-ß2 treated and untreated primary lens epithelial cells (pLECs). Differentially expressed genes (DEGs) associated with ferroptosis were analyzed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) to investigate their biological function. Additionally, protein-to-protein interactions among selected ferroptosis-related genes by PPI network and the top 10 genes with the highest score (MCC algorithm) were selected as the hub genes. The top 20 genes with significant fold change values were validated using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: Our analysis revealed 1253 DEGs between TGF-ß2 treated and untreated pLECs, uncovering 38 ferroptosis-related genes between two groups. Among these 38 ferroptosis-related genes,the most prominent GO enrichment analysis process involved in the response to oxidative stress (BPs), apical part of cell (CCs),antioxidant activity (MFs). KEGG were mainly concentrated in fluid shear stress and atherosclerosis, IL-17 and TNF signaling pathways, and validation of top 20 genes with significant fold change value were consistent with RNA-seq. CONCLUSIONS: Our RNA-Seq data identified 38 ferroptosis-related genes in TGF-ß2 treated and untreated pLECs, which is the first observation of ferroptosis related genes in primary human lens epithelial cells under TGF-ß2 stimulation.


Capsule Opacification , Ferroptosis , Humans , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology , Transcriptome , Epithelial-Mesenchymal Transition/genetics , Ferroptosis/genetics , Blotting, Western , Capsule Opacification/genetics , Capsule Opacification/metabolism , Epithelial Cells/metabolism
2.
Int J Biol Macromol ; 259(Pt 2): 129290, 2024 Feb.
Article En | MEDLINE | ID: mdl-38199534

Posterior capsule opacification (PCO) is the most common postoperative complication of cataract surgery. Transforming growth factor-ß (TGF-ß) is related to epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) that is proven to induce PCO formation in clinical and experimental studies. In this study, CRISPR sequences targeting exon of TGF-ßRII were knocked out with lentiviral transfection in LECs. Rabbits' PCO model was established and recombinant adeno-associated virus (AAV) for transferring the gRNA of TGF ßRII were intravitreally injected. SgRNA inhibited TGF-ßRII expression and human LECs proliferation. In TGF-ßRII knockout group, LECs motility and migration were suppressed, N-cadherin and vimentin expressions were significantly decreased, whereas E-cadherin was increased. The animal model showed that TGF-ßRII knockout in vivo was effective in suppressing PCO. The current study suggested that the CRISPR/Cas9 endonuclease system could suppress TGF-ßRII secretion, which participates in the EMT procedure of LECs in vitro and PCO in vivo. These findings might provide a new gene-editing approach and insight into a novel therapeutic strategy for PCO.


Capsule Opacification , Lens, Crystalline , Animals , Humans , Rabbits , Capsule Opacification/genetics , Capsule Opacification/metabolism , CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems , Lens, Crystalline/metabolism , Epithelial Cells , Epithelial-Mesenchymal Transition/genetics , Epithelium/metabolism , Cell Movement , Cell Proliferation
3.
Mol Cell Biochem ; 479(4): 743-759, 2024 Apr.
Article En | MEDLINE | ID: mdl-37171723

Fibrotic cataract, including anterior subcapsular cataract (ASC) and posterior capsule opacification, always lead to visual impairment. Epithelial-mesenchymal transition (EMT) is a well-known event that causes phenotypic alterations in lens epithelial cells (LECs) during lens fibrosis. Accumulating studies have demonstrated that microRNAs are important regulators of EMT and fibrosis. However, the evidence explaining how microRNAs modulate the behavior and alter the cellular phenotypes of the lens epithelium in fibrotic cataract is insufficient. In this study, we found that hsa-let-7c-3p is downregulated in LECs in human ASC in vivo as well as in TGFß2-induced EMT in vitro, indicating that hsa-let-7c-3p may participate in modulating the profibrotic processes in the lens. We then demonstrated that overexpression of hsa-let-7c-3p markedly suppressed human LEC proliferation and migration and attenuated TGFß2-induced EMT and injury-induced ASC in a mouse model. In addition, hsa-let-7c-3p mediated lens fibrosis by directly targeting the CDH11 gene, which encodes cadherin-11 protein, an important mediator in the EMT signaling pathway. It decreased cadherin-11 protein expression at the posttranscriptional level but not at the transcriptional level by binding to a specific site in the 3-untranslated region (3'-UTR) of CDH11 mRNA. Moreover, blockade of cadherin-11 expression with a specific short hairpin RNA reversed TGFß2-induced EMT in LECs in vitro. Collectively, these data demonstrated that hsa-let-7c-3p plays a clear role in attenuating ASC development and may be a novel candidate therapeutic for halting fibrosis and maintaining vision.


Cadherins , Capsule Opacification , Cataract , Lens, Crystalline , MicroRNAs , Animals , Humans , Mice , Capsule Opacification/genetics , Capsule Opacification/metabolism , Cataract/genetics , Cataract/metabolism , Cataract/pathology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Fibrosis , Lens, Crystalline/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
4.
Exp Eye Res ; 231: 109463, 2023 06.
Article En | MEDLINE | ID: mdl-37044287

Posterior capsule opacification (PCO) is a serious complication after cataract surgery. Diabetes could increase the occurrence of PCO, but the mechanism is still unclear. The purpose of this study is to investigate the role of small extracellular vesicles (sEVs) derived from diabetic aqueous humor in PCO process. Intraoperatively-derived aqueous humor sEVs from patients with diabetic related cataract (DRC) promoted the epithelial-mesenchymal transition (EMT) and metastasis of human lens epithelial cells (LECs). Via mouse PCO surgical model and DiI labeled fluorescence detection of sEVs, the sEVs derived from vascular endothelium were discovered directly contacting with LECs. Furthermore, we demonstrated that high-glucose-cultured human umbilical vein endothelial cells (HUVEC) -derived sEVs facilitated EMT process of HLE-B3 using co-culture model in vitro. miRNA-seq data and GEO datasets analysis revealed that miR-1246 was essential in EMT process with diabetes. The miR-1246 was highly expressed in diabetic aqueous humor sEVs and high-glucose-treated vascular-endothelial-cell-derived sEVs. Moreover, miR-1246 promoted the metastasis and EMT process of HLE-B3 cells by directly targeting GSK-3ß. Inhibiting miR-1246 could negatively regulated EMT. This finding might serve as a potential therapy for diabetic PCO.


Capsule Opacification , Diabetes Mellitus , Lens, Crystalline , MicroRNAs , Humans , Mice , Animals , Capsule Opacification/genetics , Capsule Opacification/pathology , Glycogen Synthase Kinase 3 beta , Endothelial Cells/pathology , MicroRNAs/genetics , Epithelial Cells/pathology , Diabetes Mellitus/pathology , Glucose , Epithelial-Mesenchymal Transition
5.
Curr Eye Res ; 48(7): 618-626, 2023 07.
Article En | MEDLINE | ID: mdl-36946600

PURPOSE: Posterior capsule opacification (PCO) is the most common postoperative complication after cataract surgery and cannot yet be eliminated. Here, we investigated the inhibitory effects of telomerase reverse transcriptase (TERT) gene silencing on PCO in a rabbit model. METHODS: After rabbit lens epithelial cells (LECs) were treated with adenovirus containing short hairpin RNAs (shRNA) targeting TERT (shTERT group), adenovirus containing scramble nonsense control shRNA (shNC group) or PBS (control group), quantitative real-time polymerase chain reaction and Western blotting were used to measure the expression levels of TERT, and a scratch assay was performed to assess the LEC migration. New Zealand white rabbits underwent sham cataract surgery followed by an injection of adenovirus carrying shTERT into their capsule bag. The intraocular pressure and anterior segment inflammation were evaluated on certain days, and EMT markers (α-SMA and E-cadherin) were evaluated by Western blotting and immunofluorescence. The telomerase activity of the capsule bag was detected by ELISA. At 28 d postoperatively, hematoxylin and eosin staining of the cornea and iris and electron microscopy of the posterior capsule were performed. RESULTS: Application of shTERT to LECs downregulated the expression levels of TERT mRNA and protein. The scratch assay results showed a decrease in the migration of LECs in the shTERT group. In vivo, shTERT decreased PCO formation after cataract surgery in rabbits and downregulated the expression of EMT markers, as determined by Western blotting and immunofluorescence. In addition, telomerase activity was suppressed in the capsule bag. Despite slight inflammation in the iris, histologic results revealed no toxic effects in the cornea and iris. CONCLUSION: TERT silencing effectively reduces the migration and proliferation of LECs and the formation of PCO. Our findings suggest that TERT silencing may be a potential preventive strategy for PCO.


Capsule Opacification , Cataract , Telomerase , Rabbits , Animals , Capsule Opacification/genetics , Capsule Opacification/prevention & control , Capsule Opacification/metabolism , RNA, Small Interfering/genetics , Adenoviridae/genetics , Telomerase/genetics , Telomerase/metabolism , Inflammation/metabolism , Epithelial Cells/metabolism , Cataract/metabolism
6.
Contrast Media Mol Imaging ; 2022: 5383146, 2022.
Article En | MEDLINE | ID: mdl-35909588

The abnormal proliferation, migration, and epithelial-mesenchymal transformation (EMT) of lens epithelial cells (LECs) are the main reasons for vision loss caused by posterior capsular opacification (PCO) after cataract surgery. Insulin-like growth factor-1 (IGF-1) was found to be associated with the pathogenesis of cataracts, but its biological role in PCO is poorly understood. In the present study, IGF-1 overexpression facilitated the proliferation, migration, and EMT, whereas knockdown of IGF-1 markedly suppressed the proliferation, migration, and TGF-ß2-induced EMT of LECs. Additionally, to evaluate valuable microRNAs (miRNAs) which target IGF-1 to modulate LEC-EMT, we predicted miR-3666 might regulate IGF-1 by binding its 3'UTR according to the bioinformatics database. Furthermore, we verified that miR-3666 directly targeted IGF-1 by luciferase reporter assay. By using miR-3666 mimics, cell proliferation, migration, and invasion were suppressed, while being enhanced by the reduction of miR-3666. Knockout of IGF1 reverses the effect of the miR-3666 inhibitor on the malignant behavior of LECs. These results indicate the role of miR-3666/IGF-1 in LEC-EMT that offers new strategies for the therapy and prevention of PCO.


Capsule Opacification , Insulin-Like Growth Factor I , Lens, Crystalline , MicroRNAs , Capsule Opacification/genetics , Capsule Opacification/metabolism , Capsule Opacification/pathology , Cell Movement/genetics , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , MicroRNAs/genetics , MicroRNAs/metabolism
7.
Cells ; 11(16)2022 08 17.
Article En | MEDLINE | ID: mdl-36010635

The integrity of lens epithelial cells (LECs) lays the foundation for lens function and transparency. By contrast, epithelial-mesenchymal transition (EMT) of LECs leads to lens fibrosis, such as anterior subcapsular cataracts (ASC) and fibrotic forms of posterior capsule opacification (PCO). However, the underlying mechanisms remain unclear. Here, we aimed to explore the role of long non-coding RNA (lncRNA) H19 in regulating TGF-ß2-induced EMT during lens fibrosis, revealing a novel lncRNA-based regulatory mechanism. In this work, we identified that lncRNA H19 was highly expressed in LECs, but downregulated by exposure to TGF-ß2. In both human lens epithelial explants and SRA01/04 cells, knockdown of H19 aggravated TGF-ß2-induced EMT, while overexpressing H19 partially reversed EMT and restored lens epithelial phenotypes. Semi-in vivo whole lens culture and H19 knockout mice demonstrated the indispensable role of H19 in sustaining lens clarity through maintaining LEC features. Bioinformatic analyses further implied a potential H19-centered regulatory mechanism via Smad-dependent pathways, confirmed by in vitro experiments. In conclusion, we uncovered a novel role of H19 in inhibiting TGF-ß2-induced EMT of the lens by suppressing Smad-dependent signaling, providing potential therapeutic targets for treating lens fibrosis.


Capsule Opacification , RNA, Long Noncoding , Animals , Capsule Opacification/genetics , Capsule Opacification/metabolism , Epithelial Cells/metabolism , Fibrosis , Humans , Mice , Phenotype , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transforming Growth Factor beta2/metabolism
8.
Bioengineered ; 13(6): 14357-14367, 2022 06.
Article En | MEDLINE | ID: mdl-35758265

Epithelial-melancholy transition (EMT) is the main cause of organ fibrosis and a common pathogenetic mechanism in most cataracts. This study aimed to explore the molecular mechanism of Toll-like receptor (TLR)-3 in the occurrence and development of post-cataract EMT and to provide new ideas for the prevention and treatment of posterior capsule opacification (PCO). In the presence or absence of TLR3, the human lens epithelial cell (LEC) line, SRA01/04, was treated with the transforming growth factor (TGF)-ß2. Cell counting kit-8 (CCK-8) and Transwell assays were used to analyze the cell proliferation, migration, and invasion. The expression levels of proteins and RNAs were detected by western blotting and quantitative polymerase chain reaction (qPCR) experiments. Functional gain and loss studies showed that TLR3 regulates the proliferation, migration, and invasion of LECs and EMT induced by TGF-ß2. Moreover, TLR3 regulates the expression of Jagged-1, Notch-1, and Notch-3 These findings indicate that TLR3 prevents the progression of lens fibrosis by targeting the Jagged-1/Notch signaling pathway to regulate the proliferation, migration, and invasion of LECs, and TGF-ß2-induced EMT. Therefore, the TLR3-Jagged-1/Notch signaling axis may be a potential therapeutic target for the treatment of fibrotic cataracts.


Capsule Opacification , Receptors, Notch , Toll-Like Receptor 3 , Capsule Opacification/genetics , Capsule Opacification/metabolism , Capsule Opacification/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Fibrosis , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Transforming Growth Factor beta2/pharmacology
9.
J Biochem Mol Toxicol ; 36(9): e23144, 2022 Sep.
Article En | MEDLINE | ID: mdl-35730126

Posterior capsular opacification (PCO) is the major complication after cataract surgery and can result in secondary vision loss. Circular RNAs (circRNAs) are reported to play critical regulatory roles in multiple cell biological processes. The most common working mechanism of circRNAs is by acting as microRNA sponges. Here, we analyzed the role and mechanism of circRNA RNA polymerase III subunit A (POLR3A) in PCO. Cell viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Cell motility was assessed by transwell and wound healing assays. Dual-luciferase reporter and RNA-pull-down assays were performed to verify the interaction between microRNA-31 (miR-31) and circ-POLR3A or thioredoxin interacting protein (TXNIP). PCO cell model was established by treating SRA01/04 cells with transforming growth factor-ß2 (TGF-ß2). We found that TGF-ß2 enhanced SRA01/04 cell viability, migration, and invasion abilities. Circ-POLR3A expression was upregulated in PCO tissues and TGF-ß2-induced SRA01/04 cells. TGF-ß2 promoted the viability and motility of SRA01/04 cells largely by upregulating circ-POLR3A. Circ-POLR3A negatively regulated the miR-31 level by directly interacting with it. Circ-POLR3A absence-induced influences in TGF-ß2-induced SRA01/04 cells were partly reversed by silencing miR-31. miR-31 is directly bound to the 3'-untranslated region of TXNIP. TXNIP overexpression largely attenuated miR-31 overexpression-mediated effects in TGF-ß2-induced SRA01/04 cells. Circ-POLR3A could elevate the protein expression of TXNIP by sponging miR-31. Exosomes were involved in mediating the delivery of circ-POLR3A in SRA01/04 cells. In conclusion, circ-POLR3A contributed to TGF-ß2-induced promotion of cell viability, migration, and invasion of SRA01/04 cells by targeting miR-31/TXNIP axis.


Capsule Opacification , MicroRNAs , 3' Untranslated Regions , Capsule Opacification/genetics , Capsule Opacification/metabolism , Carrier Proteins/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , MicroRNAs/metabolism , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , RNA Polymerase III/pharmacology , RNA, Circular/genetics , Thioredoxins , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology , Transforming Growth Factors/genetics , Transforming Growth Factors/metabolism , Transforming Growth Factors/pharmacology
10.
Curr Eye Res ; 47(5): 735-746, 2022 05.
Article En | MEDLINE | ID: mdl-35392747

BACKGROUND: Circular RNA (circRNA) is a novel star factor in the research of ocular diseases including cataract and the most common postoperative complication posterior capsule opacification (PCO). Hsa_circRNA_0060640 (circ_0060640) is an age-related cataract-related circRNA. However, its role in cataractogenesis is unrevealed yet. METHODS: PCO in vitro model was established in human lens epithelium cells (hLECs) induced by transforming growth factor-beta2 (TGF-ß2). RNA and protein expressions were respectively detected by quantitative PCR and western blotting. Direct interaction between two RNAs was predicted by Starbase tool and confirmed by dual-luciferase reporter assay. MTS and EdU assays measured cell proliferation; Transwell, starch wound and western blotting assays evaluated cell motility and epithelial-mesenchymal transition (EMT). RESULTS: Circ_0060640 expression is higher in anterior lens capsule tissues from human cataractous eyes and TGF-ß2-stimulated hLECs cells line SRA01/04. RNA interference of circ_0060640 could prevent SRA01/04 cells from TGF-ß2-induced cell proliferation, migration and invasion, accompanied with decreased N-cadherin and α-smooth muscle actin and increased E-cadherin. Mechanistically, circ_0060640 directly controls microRNA (miR)-214-3p expression and then regulates gene expression of collagen type I alpha2 chain (COL1A2). Notably, COL1A2 inhibition is underlying the protective role of circ_0060640 silencing and miR-214-3p ectopic expression in TGF-ß2-stimulated SRA01/04 cells. CONCLUSION: Circ_0060640 is a novel cataract-related gene and its silencing could block TGF-ß2-evoked hLECs proliferation, motility and EMT in vitro via targeting miR-214-3p-COL1A2 axis. Therefore, targeting circ_0060640 via RNA interference might be a treatment strategy for PCO development.


Capsule Opacification , MicroRNAs , Capsule Opacification/genetics , Capsule Opacification/metabolism , Cell Movement , Cell Proliferation , Collagen Type I/genetics , Epithelial-Mesenchymal Transition , Epithelium/metabolism , Humans , MicroRNAs/genetics , RNA, Circular/genetics , Transforming Growth Factor beta2/pharmacology
11.
Biochem Biophys Res Commun ; 598: 62-68, 2022 04 02.
Article En | MEDLINE | ID: mdl-35151205

The cause of posterior capsular opacification (PCO) is the dysfunction of lens epithelial cells (LECs). Circular RNA (circRNA) was found to regulate cell biological functions, including LECs. However, the role of circ-GGA3 in PCO formation is unclear. Quantitative real-time PCR was used to measure the expression of circ-GGA3, miR-497-5p and SMAD4. Cell proliferation, invasion and migration were determined via MTT assay, EdU staining, transwell assay and wound healing assay. The protein expression of epithelial-mesenchymal transition (EMT) markers, fibrosis markers, TGF-ß/SMAD pathway markers and SMAD4 were determined by western blot assay. The interaction between miR-497-5p and circ-GGA3 or SMAD4 was confirmed using dual-luciferase reporter assay. Circ-GGA3 was highly expressed in PCO patients, and its silencing inhibited the proliferation, invasion, migration, EMT process and fibrosis of TGF-ß2-induced LECs. Circ-GGA3 could sponge miR-497-5p to regulate SMAD4. Further experiments revealed that miR-497-5p inhibitor recovered the negative regulation of circ-GGA3 knockdown on the biological functions of TGF-ß2-induced LECs, and SMAD4 overexpression also abolished the suppressive effect of miR-497-5p. In addition, circ-GGA3/miR-497-5p/SMAD4 axis could activate the TGF-ß/SMAD pathway. Our results indicated that circ-GGA3 could enhance the biological functions of LECs, suggesting that circ-GGA3 might be a potential target for PCO therapy.


Capsule Opacification/genetics , Lens, Crystalline/cytology , MicroRNAs/genetics , RNA, Circular/genetics , Smad4 Protein/genetics , Capsule Opacification/pathology , Case-Control Studies , Cells, Cultured , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/pharmacology
12.
Curr Eye Res ; 47(3): 372-381, 2022 03.
Article En | MEDLINE | ID: mdl-34961410

OBJECTIVE: Posterior capsular opacification (PCO) is a common postoperative ocular complication after cataract surgery. Little research focused on the regulation of circular RNAs (circRNAs) in PCO. This study was designed to investigate the function of circRNA-muskelin (circ-MKLN1) in PCO. METHODS: SRA01/04 cells were treated with transforming growth factor (TGF)-ß2. Cell viability was analyzed by Cell Counting Kit-8 (CCK-8) assay. Transwell assay was used for cell migration and invasion detection. Cell migration was also measured by wound healing assay. Epithelial-mesenchymal transition (EMT)-related proteins and connective tissue growth factor (CTGF) were quantified using western blot. RESULTS: Cell viability, migration, invasion and EMT process in SRA01/04 cells were facilitated by TGF-ß2. Circ-MKLN1 expression was enhanced in 17 PCO lens samples relative to 19 normal lens samples and TGF-ß2-treated SRA01/04 cells contrasted to control cells. Downregulation of circ-MKLN1 inhibited the effects of TGF-ß2 on SRA01/04 cells. Circ-MKLN1 targeted miR-377-3p and the regulation of si-circ-MKLN1 for the TGF-ß2-induced influences was related to the upregulation of miR-377-3p. CTGF was the target gene for miR-377-3p. CTGF knockdown also abolished the TGF-ß2-mediated cell growth, migration and invasion of SRA01/04 cells. The function of miR-377-3p was achieved by reducing the CTGF level. TGF-ß2-induced CTGF expression promotion was alleviated by si-circ-MKLN1 through upregulating the expression of miR-377-3p. CONCLUSION: These results showed that circ-MKLN1 contributed to the progression of PCO in vitro by increasing the CTGF expression via sponging miR-377-3p. Circ-MKLN1 might be important for improving the molecular target therapy in PCO.


Capsule Opacification , Lens, Crystalline , MicroRNAs , Capsule Opacification/genetics , Capsule Opacification/metabolism , Cell Adhesion Molecules/metabolism , Cell Proliferation , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Lens, Crystalline/metabolism , MicroRNAs/genetics , MicroRNAs/pharmacology , RNA, Circular/genetics , Transforming Growth Factor beta2/pharmacology
13.
Life Sci ; 286: 120061, 2021 Dec 01.
Article En | MEDLINE | ID: mdl-34666037

AIMS: The epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) synthesis and cell migration of residual lens cells constitute the canonical mechanisms of posterior capsular opacification (PCO). Recently, myofibroblast cell apoptosis is also observed in the rabbit PCO model. However, whether cell apoptosis is a key factor affecting PCO and regulates EMT/ECM synthesis/cell migration remains obscure. MAIN METHODS: Flow cytometry was utilized to assess cell cycle and apoptosis. EMT marker α-smooth muscle actin (α-SMA), ECM markers fibronectin (Fn), type 1 collagen (COL-1) and apoptosis-associated proteins in the presence or absence of EMT/ECM inhibitor (LY2109761), apoptosis inhibitor (ZVAD) or apoptosis activator (BTSA1) were detected by Western blotting. Downstream effector genes in apoptosis-induced lens epithelial cell lines (LECs) were analyzed by RNA-seq. Gene silencing and overexpression in LECs were performed to validate the role of effector genes. We measured cell migration capability using Wound healing and Transwell assays. KEY FINDINGS: We found that TGF-ß2 induced cell apoptosis. ZVAD inhibited α-SMA expression in the ex vivo capsule model and decreased the expression of both EMT and ECM markers in TGF-ß2-treated LECs. RNA-seq revealed that FILIP1L was significantly decreased in apoptosis-activated cells. We further validated that the knockdown of FILIP1L could enhance EMT and ECM synthesis and promote cell migration and that FILIP1L overexpression could reverse these effects. Apoptosis might contribute to TGF-ß2-induced EMT and ECM synthesis during PCO, and these contributions are mediated by FILIP1L. SIGNIFICANCE: Our findings uncover the role of apoptosis in PCO development and provide new drug targets.


Capsule Opacification/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Apoptosis/drug effects , Capsule Opacification/genetics , Cell Cycle , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , China , Collagen Type I/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix/drug effects , Extracellular Matrix/physiology , Fibronectins/metabolism , Flow Cytometry/methods , Humans , Intracellular Signaling Peptides and Proteins/physiology , Lens, Crystalline/metabolism , Male , Posterior Capsule of the Lens/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Swine , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology
14.
Biomolecules ; 11(8)2021 08 04.
Article En | MEDLINE | ID: mdl-34439816

Cataracts are a leading cause of blindness worldwide. Surgical removal of cataracts is a safe and effective procedure to restore vision. However, a large number of patients later develop vision loss due to regrowth of lens cells and subsequent degradation of the visual axis leading to visual disability. This postsurgical complication, known as posterior capsular opacification (PCO), occurs in up to 30% of cataract patients and has no clinically proven pharmacological means of prevention. Despite the availability of many compounds capable of preventing early steps in PCO development, there is currently no effective means to deliver such therapies into the eye for a suitable duration. To model a solution to this unmet medical need, we fabricated acrylic substrates as intraocular lens (IOL) mimics scaled to place into the capsular bag of the mouse lens following a mock-cataract surgery. Substrates were coated with a hydrophilic crosslinked acrylate nanogel designed to elute Sorbinil, an aldose reductase inhibitor previously shown to suppress PCO. Insertion of the Sorbinil-eluting device into the lens capsule at the time of cataract surgery resulted in substantial prevention of cellular changes associated with PCO development. This model demonstrates that a cataract inhibitor can be delivered into the postsurgical lens capsule at therapeutic levels.


Capsule Opacification/prevention & control , Cataract Extraction/adverse effects , Drug Carriers , Enzyme Inhibitors/pharmacology , Imidazolidines/pharmacology , Lenses, Intraocular , Actins/genetics , Actins/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Capsule Opacification/etiology , Capsule Opacification/genetics , Capsule Opacification/pathology , Cataract/genetics , Cataract/metabolism , Cataract/pathology , Cataract Extraction/methods , Disease Models, Animal , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Humans , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/surgery , Mice , Nanogels/administration & dosage , Nanogels/chemistry , Signal Transduction , Vimentin/genetics , Vimentin/metabolism
15.
Exp Eye Res ; 209: 108676, 2021 08.
Article En | MEDLINE | ID: mdl-34146586

The transcriptome of mammalian tissues differs between males and females, and these differences can change across the lifespan, likely regulating known sexual dimorphisms in disease prevalence and severity. Cataract, the most prevalent disease of the ocular lens, occurs at similar rates in young individuals, but its incidence is elevated in older women compared to men of the same age. However, the influence of sex on the lens transcriptome was unknown. RNAseq based transcriptomic profiling of young adult C57BL/6J mouse lens epithelial and fiber cells revealed that few genes are differentially expressed between the sexes. In contrast, lens cells from aged (24 month old) male and female C57BL/6J mice differentially expressed many genes, including several whose expression is lens preferred. Like cataracts, posterior capsular opacification (PCO), a major sequela of cataract surgery, may also be more prevalent in women. Lens epithelial cells isolated from mouse eyes 24 h after lens fiber cell removal exhibited numerous transcriptomic differences between the sexes, including genes implicated in complement cascades and extracellular matrix regulation, and these differences are much more pronounced in aged mice than in young mice. These results provide an unbiased basis for future studies on how sex affects the lens response to aging, cataract development, and cataract surgery.


Capsule Opacification/genetics , Cataract Extraction/adverse effects , Extracellular Matrix/genetics , Gene Expression Regulation , Lens, Crystalline/metabolism , Postoperative Complications/epidemiology , Transcriptome/genetics , Animals , Capsule Opacification/metabolism , Capsule Opacification/surgery , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Female , Gene Expression Profiling/methods , Lens Capsule, Crystalline/metabolism , Lens Capsule, Crystalline/pathology , Male , Mice , Mice, Inbred C57BL , Postoperative Complications/genetics , Postoperative Complications/metabolism , Sex Factors , Wound Healing/genetics
16.
Exp Eye Res ; 207: 108572, 2021 06.
Article En | MEDLINE | ID: mdl-33844960

BACKGROUND: Posterior capsular opacification (PCO) is the major vision-disrupting complication arising after cataract surgery. Circular RNAs (circRNAs) are biological active RNAs which were involved in various physiological functions. So far, the role of circRNA caspase recruitment domain family member 6 (circ-CARD6) in PCO is still unclear. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) was applied to detect the expression of circ-CARD6, microRNA 31 (miR-31) and fibroblast growth factor 7 (FGF7) message RNA (mRNA). Western blot was used to analyze the protein expression. Transmission electron microscopy (TEM) was employed to capture the exosome image. The proliferation and metastasis were analyzed by cell counting kit-8 (CCK8), transwell and wound healing assays. The potential binding sequences between miR-31 and circ-CARD6 or FGF7 were respectively predicted by Circinteractome and Targetscan online tool, and verified by dual-luciferase reporter and RNA binding protein immunoprecipitation (RIP) assays. RESULTS: Exosome-transmitted circ-CARD6 was highly expressed in PCO tissues and TGF-ß2-treated SRA01/04 cells. Circ-CARD6 deletion repressed the proliferation, metastasis, EMT process and MAPK pathway, which was reversed by anti-miR-31 in TGF-ß2-treated SRA01/04 cells. Meanwhile, circ-CARD6 sponged miR-31 which directly targeted FGF7 in TGF-ß2-treated SRA01/04 cells. FGF7 overexpression allayed miR-31 overexpression-induced suppression in proliferation, metastasis, EMT process and MAPK pathway. Besides, circ-CARD6 regulated FGF7 expression by sponging miR-31. CONCLUSION: Circ-CARD6 promoted PCO development via miR-31/FGF7 axis. This finding might contribute to the development of the targeted therapy for PCO.


CARD Signaling Adaptor Proteins/genetics , Capsule Opacification/genetics , Exosomes/genetics , Fibroblast Growth Factor 7/genetics , MicroRNAs/genetics , Posterior Capsule of the Lens/pathology , Blotting, Western , Capsule Opacification/pathology , Epithelial Cells/cytology , Gene Expression Regulation/physiology , Humans , Lens, Crystalline/cytology , Microscopy, Electron, Transmission , RNA, Circular/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Transfection , Wound Healing/physiology
17.
Mol Med Rep ; 22(1): 436-444, 2020 07.
Article En | MEDLINE | ID: mdl-32377733

Posterior capsular opacification (PCO) is the main complication following cataract surgery. The proliferation of the residual lens epithelial cells (LECs) serves an important role in PCO formation. The authors' previous study revealed that vitamin C inhibited the proliferation of human LECs by increasing the rapid degradation of hypoxia­inducible factor­1 (HIF­1α), and hence inhibited the expression of vascular endothelial growth factor (VEGF). The present study aimed to further investigate the mechanisms underlying the effects of vitamin C on the expression levels of VEGF. The present study demonstrated that the HIF­1 inhibitor BAY 87­2243 significantly inhibited the cell proliferation and the expression levels of VEGF in LECs through the use of colony formation, western blotting and ELISA assays. Moreover, it was revealed that vitamin C could further inhibit the cell proliferation and the expression levels of VEGF in LECs following the cotreatment with the HIF­1 inhibitor. The proline hydroxylation of HIF­1α by prolyl hydroxylases (PHDs) was previously discovered to be responsible for the rapid degradation of HIF­1α. Thus, the present study subsequently used three PHD inhibitors to investigate their effects on the expression levels of VEGF; it was found that the PHD2 specific inhibitor increased the expression levels of VEGF to the greatest extent. Moreover, the genetic knockdown of PHD2 by lentiviral transfection also significantly increased the expression levels of VEGF, whereas the PHD2 specific inhibitor did not alter the expression levels of VEGF in the PHD2 knockdown LECs. AKT kinase activity is an important mediator known to upregulate VEGF expression. Using an immunoprecipitation assay to isolate endogenous AKT, it was demonstrated that AKT was prolyl hydroxylated by PHD2, which inhibited its activity. It was also revealed that vitamin C enhanced the proline­hydroxylation and inhibited the activity of AKT. Furthermore, an AKT inhibitor increased the effects of vitamin C on the expression levels of VEGF. However, the AKT inhibitor did not affect the expression levels of glucose transporter 1, which is a HIF­1α target gene. In conclusion, the findings of the present study suggested that vitamin C may inhibit the expression levels of VEGF via HIF­1α­dependent and AKT­dependent pathways in LECs.


Ascorbic Acid/pharmacology , Down-Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lens, Crystalline/drug effects , Vascular Endothelial Growth Factor A/genetics , Vitamins/pharmacology , Capsule Opacification/drug therapy , Capsule Opacification/genetics , Capsule Opacification/metabolism , Cell Line , Cell Proliferation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
18.
Sci China Life Sci ; 63(12): 1928-1937, 2020 Dec.
Article En | MEDLINE | ID: mdl-32399769

The purpose of this study was to investigate the role of a human lens microRNA (miR-497-5p) in regulating epithelialmesenchymal transition (EMT) under the control of transforming growth factor beta (TGF-ß). A microRNA array was used to evaluate the microRNA profiles of untreated and TGF-ß-treated human lens epithelial cells in culture. This showed that TGF-ß treatment led to the upregulation of 96 microRNAs and downregulation of 39 microRNAs. Thirteen microRNAs were predicted to be involved in the pathogenesis of posterior capsule opacification (PCO). Meanwhile, overexpression of miR-497-5p suppressed cell proliferation and EMT 48 h post-transfection, and inhibition of miR-497-5p accelerated cell proliferation and EMT. Treatment with TGF-ß inhibited the expression of miR-497-5p, but not cell proliferation. miR-497-5p was also found to regulate the level of CCNE1 and FGF7, which are reported to be actively involved in EMT. CCNE1 and FGF7 were bona fide targets of miR-497-5p. The results suggest that miR-497-5p participates in the direct regulation of lens epithelial cell EMT and is regulated by TGF-ß. miR-497-5p may be a novel target for PCO therapy.


Epithelial-Mesenchymal Transition/drug effects , Lens, Crystalline/drug effects , MicroRNAs/genetics , Transforming Growth Factor beta/pharmacology , Capsule Opacification/genetics , Capsule Opacification/metabolism , Capsule Opacification/pathology , Cell Line , Cell Proliferation/drug effects , Cyclin E/genetics , Cyclin E/metabolism , Epithelial Cells , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Gene Expression Regulation/drug effects , Humans , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , MicroRNAs/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism
19.
J Biosci ; 452020.
Article En | MEDLINE | ID: mdl-32345785

Transforming growth factor ß2 (TGF-ß2)/Smad signaling is widely accepted as a key inducer of proliferation and epithelial-mesenchymal transition (EMT) of human lens epithelial cells (LECs), contributing to the development of posterior capsule opacification (PCO). Increasing evidence shows that microRNAs (miRNAs) play important roles in PCO pathogenesis. Herein, we aimed to explore the role and molecular mechanism of let-7a-5p on TGF-ß2-induced proliferation and EMT in LECs. qRT-PCR was performed to detect the expression of let-7a-5p and Smad2 mRNA. Western blot was used to determine the Smad2 level and the induction of EMT. The targeted correlation between let-7a-5p and Smad2 was confirmed using dual-luciferase reporter and RNA immunoprecipitation assays. CCK-8 assay was employed to determine cell proliferation, and transwell assays were performed to assess cell migration and invasion. We found that TGF-ß2 induced EMT of LECs, and TGF-ß2 upregulated Smad2 expression and reduced let-7a-5p expression in LECs. Smad2 was a direct target of let-7a-5p. Moreover, let-7a-5p upregulation repressed proliferation, migration, invasion and EMT in TGF-ß2-induced LECs. But, Smad2 expression restoration abrogated the inhibitory effect of let-7a-5p upregulation. In conclusion, our data indicated that let-7a-5p upregulation repressed TGF-ß2-induced proliferation, migration, invasion and EMT at least partly by targeting Smad2 in LECs, highlighting that let-7a-5p might act as a promising therapeutic target to intervene to the progression of PCO.


Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta2/metabolism , Capsule Opacification/genetics , Capsule Opacification/metabolism , Cell Line , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Humans , Lens, Crystalline/cytology , Posterior Capsule of the Lens/metabolism , Signal Transduction/genetics , Smad2 Protein/genetics
20.
Matrix Biol ; 90: 79-108, 2020 08.
Article En | MEDLINE | ID: mdl-32173580

Fibrotic posterior capsular opacification (PCO), one of the major complications of cataract surgery, occurs when lens epithelial cells (LCs) left behind post cataract surgery (PCS) undergo epithelial to mesenchymal transition, migrate into the optical axis and produce opaque scar tissue. LCs left behind PCS robustly produce fibronectin, although its roles in fibrotic PCO are not known. In order to determine the function of fibronectin in PCO pathogenesis, we created mice lacking the fibronectin gene (FN conditional knock out -FNcKO) from the lens. While animals from this line have normal lenses, upon lens fiber cell removal which models cataract surgery, FNcKO LCs exhibit a greatly attenuated fibrotic response from 3 days PCS onward as assessed by a reduction in surgery-induced cell proliferation, and fibrotic extracellular matrix (ECM) production and deposition. This is correlated with less upregulation of Transforming Growth Factor ß (TGFß) and integrin signaling in FNcKO LCs PCS concomitant with sustained Bone Morphogenetic Protein (BMP) signaling and elevation of the epithelial cell marker E cadherin. Although the initial fibrotic response of FNcKO LCs was qualitatively normal at 48 h PCS as measured by the upregulation of fibrotic marker protein αSMA, RNA sequencing revealed that the fibrotic response was already quantitatively attenuated at this time, as measured by the upregulation of mRNAs encoding molecules that control, and are controlled by, TGFß signaling, including many known markers of fibrosis. Most notably, gremlin-1, a known regulator of TGFß superfamily signaling, was upregulated sharply in WT LCs PCS, while this response was attenuated in FNcKO LCs. As exogenous administration of either active TGFß1 or gremlin-1 to FNcKO lens capsular bags rescued the attenuated fibrotic response of fibronectin null LCs PCS including the loss of SMAD2/3 phosphorylation, this suggests that fibronectin plays multifunctional roles in fibrotic PCO development.


Capsule Opacification/pathology , Cataract Extraction/adverse effects , Fibronectins/genetics , Fibronectins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Capsule Opacification/etiology , Capsule Opacification/genetics , Capsule Opacification/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Profiling , Gene Expression Regulation , Gene Knockout Techniques , Mice , Sequence Analysis, RNA , Signal Transduction , Transforming Growth Factor beta/metabolism
...