Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.206
Filter
1.
J Vis Exp ; (208)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38975789

ABSTRACT

Lens epithelial cells (LECs) play multiple important roles in maintaining the homeostasis and normal function of the lens. LECs determine lens growth, development, size, and transparency. Conversely, dysfunctional LECs can lead to cataract formation and posterior capsule opacification (PCO). Consequently, establishing a robust primary LEC culture system is important to researchers engaged in lens development, biochemistry, cataract therapeutics, and PCO prevention. However, cultivating primary LECs has long presented challenges due to their limited availability, slow proliferation rate, and delicate nature. This study addresses these hurdles by presenting a comprehensive protocol for primary LEC culture. The protocol encompasses essential steps such as the formulation of an optimized culture medium, precise isolation of lens capsules, trypsinization techniques, subculture procedures, harvest protocols, and guidelines for storage and shipment. Throughout the culture process, cell morphology was monitored using phase-contrast microscopy. To confirm the authenticity of the cultured LECs, immunofluorescence assays were conducted to detect the presence and subcellular distribution of critical lens proteins, namely αA- and γ-crystallins. This detailed protocol equips researchers with a valuable resource for cultivating and characterizing primary LECs, enabling advancements in our comprehension of lens biology and the development of therapeutic strategies for lens-related disorders.


Subject(s)
Epithelial Cells , Lens, Crystalline , Trypsin , Epithelial Cells/cytology , Lens, Crystalline/cytology , Animals , Mice , Trypsin/chemistry , Trypsin/metabolism , Cell Culture Techniques/methods , Primary Cell Culture/methods
2.
Int Ophthalmol ; 44(1): 316, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38969958

ABSTRACT

BACKGROUND: Diabetic cataract (DC) is a common complication of diabetes and its etiology and progression are multi-factorial. In this study, the roles of specific protein 1 (SP1) and fibroblast growth factor 7 (FGF7) in DC development were explored. METHODS: DC cell model was established by treating SRA01/04 cells with high glucose (HG). MTT assay was conducted to evaluate cell viability. Transwell assay and wound-healing assay were performed to assess cell migration and invasion. Western blot assay and qRT-PCR assay were conducted to measure the expression of N-cadherin, E-cadherin, Collagen I, Fibronectin, SP1 and FGF7 expression. CHIP assay and dual-luciferase reporter assay were conducted to analyze the combination between FGF7 and SP1. RESULTS: FGF7 was upregulated in DC patients and HG-induced SRA01/04 cells. HG treatment promoted SRA01/04 cell viability, migration, invasion and epithelial-mesenchymal transition (EMT), while FGF7 knockdown abated the effects. Transcription factor SP1 activated the transcription level of FGF7 and SP1 overexpression aggravated HG-induced SRA01/04 cell injury. SP1 silencing repressed HG-induced SRA01/04 cell viability, migration, invasion and EMT, but these effects were ameliorated by upregulating FGF7. Additionally, SP1 knockdown inhibited the PI3K/AKT pathway by regulating the transcription level of FGF7. CONCLUSION: Transcription factor SP1 activated the transcription level of FGF7 and the PI3K/AKT pathway to regulate HG-induced SRA01/04 cell viability, migration, invasion and EMT.


Subject(s)
Cell Movement , Cell Survival , Epithelial Cells , Epithelial-Mesenchymal Transition , Fibroblast Growth Factor 7 , Glucose , Lens, Crystalline , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Sp1 Transcription Factor , Epithelial-Mesenchymal Transition/drug effects , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Glucose/pharmacology , Epithelial Cells/metabolism , Fibroblast Growth Factor 7/metabolism , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/pharmacology , Lens, Crystalline/metabolism , Lens, Crystalline/cytology , Cataract/metabolism , Cells, Cultured , Gene Expression Regulation
3.
Differentiation ; 138: 100792, 2024.
Article in English | MEDLINE | ID: mdl-38935992

ABSTRACT

The role extracellular matrix (ECM) in multiple events of morphogenesis has been well described, little is known about its specific role in early eye development. One of the first morphogenic events in lens development is placodal thickening, which converts the presumptive lens ectoderm from cuboidal to pseudostratified epithelium. This process occurs in the anterior pre-placodal ectoderm when the optic vesicle approaches the cephalic ectoderm and is regulated by transcription factor Pax6 and secreted BMP4. Since cells and ECM have a dynamic relationship of interdependence and modulation, we hypothesized that the ECM evolves with cell shape changes during lens placode formation. This study investigates changes in optic ECM including both protein distribution deposition, extracellular gelatinase activity and gene expression patterns during early optic development using chicken and mouse models. In particular, the expression of Timp2, a metalloprotease inhibitor, corresponds with a decrease in gelatinase activity within the optic ECM. Furthermore, we demonstrate that optic ECM remodeling depends on BMP signaling in the placode. Together, our findings suggest that the lens placode plays an active role in remodeling the optic ECM during early eye development.


Subject(s)
Extracellular Matrix , Gene Expression Regulation, Developmental , Lens, Crystalline , PAX6 Transcription Factor , Animals , Extracellular Matrix/metabolism , Mice , Lens, Crystalline/metabolism , Lens, Crystalline/growth & development , Lens, Crystalline/cytology , PAX6 Transcription Factor/metabolism , PAX6 Transcription Factor/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Chick Embryo , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction , Chickens/genetics , Eye/metabolism , Eye/growth & development , Eye/embryology
4.
Sci Rep ; 14(1): 12160, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38802452

ABSTRACT

The knowledge on responses of human lens epithelial cells (HLECs) to ionizing radiation exposure is important to understand mechanisms of radiation cataracts that are of concern in the field of radiation protection and radiation therapy. However, biological effects in HLECs following protracted exposure have not yet fully been explored. Here, we investigated the temporal kinetics of γ-H2AX foci as a marker for DNA double-strand breaks (DSBs) and cell survival in HLECs after exposure to photon beams at various dose rates (i.e., 150 kVp X-rays at 1.82, 0.1, and 0.033 Gy/min, and 137Cs γ-rays at 0.00461 Gy/min (27.7 cGy/h) and 0.00081 Gy/min (4.9 cGy/h)), compared to those in human lung fibroblasts (WI-38). In parallel, we quantified the recovery for DSBs and cell survival using a biophysical model. The study revealed that HLECs have a lower DSB repair rate than WI-38 cells. There is no significant impact of dose rate on cell survival in both cell lines in the dose-rate range of 0.033-1.82 Gy/min. In contrast, the experimental residual γ-H2AX foci showed inverse dose rate effects (IDREs) compared to the model prediction, highlighting the importance of the IDREs in evaluating radiation effects on the ocular lens.


Subject(s)
Cell Survival , DNA Breaks, Double-Stranded , Dose-Response Relationship, Radiation , Epithelial Cells , Histones , Lens, Crystalline , Humans , Epithelial Cells/radiation effects , Epithelial Cells/metabolism , Lens, Crystalline/radiation effects , Lens, Crystalline/cytology , DNA Breaks, Double-Stranded/radiation effects , Histones/metabolism , Cell Survival/radiation effects , Radiation, Ionizing , Cell Line , DNA Repair/radiation effects , Fibroblasts/radiation effects , Fibroblasts/metabolism , X-Rays , Gamma Rays/adverse effects
5.
Stem Cell Res Ther ; 15(1): 141, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745238

ABSTRACT

BACKGROUND: Previous studies have suggested that macrophages are present during lens regeneration in newts, but their role in the process is yet to be elucidated. METHODS: Here we generated a transgenic reporter line using the newt, Pleurodeles waltl, that traces macrophages during lens regeneration. Furthermore, we assessed early changes in gene expression during lens regeneration using two newt species, Notophthalmus viridescens and Pleurodeles waltl. Finally, we used clodronate liposomes to deplete macrophages during lens regeneration in both species and tested the effect of a subsequent secondary injury after macrophage recovery. RESULTS: Macrophage depletion abrogated lens regeneration, induced the formation of scar-like tissue, led to inflammation, decreased iris pigment epithelial cell (iPEC) proliferation, and increased rates of apoptosis in the eye. Some of these phenotypes persisted throughout the last observation period of 100 days and could be attenuated by exogenous FGF2 administration. A distinct transcript profile encoding acute inflammatory effectors was established for the dorsal iris. Reinjury of the newt eye alleviated the effects of macrophage depletion, including the resolution of scar-like tissue, and re-initiated the regeneration process. CONCLUSIONS: Together, our findings highlight the importance of macrophages for facilitating a pro-regenerative environment in the newt eye by regulating fibrotic responses, modulating the overall inflammatory landscape, and maintaining the proper balance of early proliferation and late apoptosis of the iPECs.


Subject(s)
Fibrosis , Lens, Crystalline , Macrophages , Regeneration , Salamandridae , Animals , Macrophages/metabolism , Regeneration/drug effects , Lens, Crystalline/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/injuries , Apoptosis/drug effects , Cell Proliferation/drug effects
6.
Redox Biol ; 73: 103216, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38820983

ABSTRACT

Long-lived lens fiber cells require a robust cellular protective function against oxidative insults to maintain their hemostasis and viability; however, the underlying mechanism is largely obscure. In this study, we unveiled a new mechanism that protects lens fiber cells against oxidative stress-induced cell death. We found that mechano-activated connexin (Cx) hemichannels (HCs) mediate the transport of glutathione (GSH) into chick embryonic fibroblasts (CEF) and primary lens fiber cells, resulting in a decrease in the accumulation of intracellular reactive oxygen species induced by both H2O2 and ultraviolet B, providing protection to lens fiber cells against cell apoptosis and necrosis. Furthermore, HCs formed by both homomeric Cx50 or Cx46 and heteromeric Cx50/Cx46 were mechanosensitive and could transport GSH into CEF cells. Notably, mechano-activated Cx50 HCs exhibited a greater capacity to transport GSH than Cx46 HCs. Consistently, the deficiency of Cx50 in single lens fiber cells led to a higher level of oxidative stress. Additionally, outer cortical short lens fiber cells expressing full length Cxs demonstrated greater resistance to oxidative injury compared to central core long lens fibers. Taken together, our results suggest that the activation of Cx HCs by interstitial fluid flow in cultured epithelial cells and isolated fiber cells shows that HCs can serve as a pathway for moving GSH across the cell membrane to offer protection against oxidative stress.


Subject(s)
Connexins , Glutathione , Lens, Crystalline , Oxidative Stress , Connexins/metabolism , Connexins/genetics , Glutathione/metabolism , Animals , Lens, Crystalline/metabolism , Lens, Crystalline/cytology , Reactive Oxygen Species/metabolism , Chick Embryo , Biological Transport , Apoptosis , Fibroblasts/metabolism , Hydrogen Peroxide/metabolism , Cells, Cultured
7.
Curr Eye Res ; 47(5): 747-752, 2022 05.
Article in English | MEDLINE | ID: mdl-35317686

ABSTRACT

PURPOSE: Cataract is a leading visual disease characterized by enhanced oxidative stress and increased apoptosis of human lens epithelial cells (HLECs). TRIM3 is a tumor suppressor in many cancers. However, its role in cataract remains unknown. In this study, we aimed to explore the role of TRIM3 in H2O2-injured HLECs and the underlying mechanisms involved. METHODS: HLECs were treated with different H2O2 concentrations to induce apoptosis. A lentivirus was designed to overexpress TRIM3 and p53, and TRIM3 knockdown was prepared. A P53 inhibitor, PFTα, was used to knockdown p53. Cell viability and apoptosis were detected by CCK-8 and flow cytometric analyses, respectively. TRIM3, p53, Bcl2, and Bax expression levels were determined by qRT-qPCR and western blotting. RESULTS: It was found that H2O2-treated HLECs had markedly decreased cell viability and TRIM3 expression. TRIM3 overexpression attenuated the H2O2-induced HLEC apoptosis, while TRIM3 knockdown promoted it. P53, a downstream target of TRIM3, was found to be negatively regulated by TRIM3 via ubiquitination in HLECs. Furthermore, p53 overexpression abolished the effect of TRIM3 overexpression on H2O2-induced HLEC apoptosis, while PFTα alleviated the TRIM3 knockdown-mediated HLEC apoptosis. CONCLUSION: This study demonstrates that TRIM3 inhibited the H2O2-induced apoptosis of HLECs by decreasing p53 via ubiquitination.


Subject(s)
Apoptosis , Carrier Proteins , Epithelial Cells , Lens, Crystalline , Tumor Suppressor Protein p53 , Carrier Proteins/metabolism , Cataract/metabolism , Cells, Cultured , Epithelial Cells/cytology , Gene Knockdown Techniques , Humans , Hydrogen Peroxide/toxicity , Lens, Crystalline/cytology , Oxidative Stress , Tumor Suppressor Protein p53/genetics , Ubiquitination
8.
Bioengineered ; 13(3): 5903-5914, 2022 03.
Article in English | MEDLINE | ID: mdl-35184653

ABSTRACT

Paeoniflorin (Pae) has been reported to serve an important role in complications associated with diabetes. To the best of our knowledge, the role of Pae in diabetic cataracts has not yet been reported. Human lens epithelial SRA01/04 cells were induced by high glucose (HG) and subsequently treated with Pae. Cell viability was detected using the MTT assay. Moreover, LDH levels were detected. Immunofluorescence (IF) and Western blotting were used to determine the protein expression levels of N-cadherin and E-cadherin. ELISA was performed to determine oxidative stress-related indicator levels. TUNEL and Western blotting detected the apoptotic rate. The mRNA and protein expression levels of sirtuin 1 (SIRT1) in SRA01/04 cells were measured via reverse transcription-quantitative PCR and Western blotting, respectively. Subsequently, cell transfection techniques were used to inhibit the expression of SIRT1 in cells. MTT, ELISA, IF, Western blotting and TUNEL assays were used to investigate the mechanisms of epithelial-mesenchymal transition (EMT) and oxidative damage with Pae in the diabetic cataract. Pae significantly increased cell viability and possibly inhibit the EMT and oxidative damage of SRA01/04 cells induced by HG. Pae was demonstrated to upregulate SIRT1 expression levels. The results therefore suggested that the downregulation of SIRT1 reversed the protective effect of Pae on EMT and oxidative damage in SRA01/04 cells induced by HG. In conclusion, Pae may inhibit EMT of lens epithelial cells and reduce oxidative damage in diabetic cataracts via the upregulation of SIRT1.


Subject(s)
Cataract , Diabetes Mellitus , Epithelial-Mesenchymal Transition , Lens, Crystalline , Oxidative Stress , Sirtuin 1 , Cataract/genetics , Cataract/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Glucosides , Humans , Lens, Crystalline/cytology , Monoterpenes , Sirtuin 1/genetics , Sirtuin 1/metabolism , Up-Regulation
9.
Biochem Biophys Res Commun ; 598: 62-68, 2022 04 02.
Article in English | MEDLINE | ID: mdl-35151205

ABSTRACT

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.


Subject(s)
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
10.
Chem Biol Interact ; 355: 109804, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35123994

ABSTRACT

Recent reports have challenged the notion that the lens is immune-privileged. However, these studies have not fully identified the molecular mechanism(s) that promote immune surveillance of the lens. Using a mouse model of targeted glutathione (GSH) deficiency in ocular surface tissues, we have investigated the role of oxidative stress in upregulating cytokine expression and promoting immune surveillance of the eye. RNA-sequencing of lenses from postnatal day (P) 1-aged Gclcf/f;Le-CreTg/- (KO) and Gclcf/f;Le-Cre-/- control (CON) mice revealed upregulation of many cytokines (e.g., CCL4, GDF15, CSF1) and immune response genes in the lenses of KO mice. The eyes of KO mice had a greater number of cells in the aqueous and vitreous humors at P1, P20 and P50 than age-matched CON and Gclcw/w;Le-CreTg/- (CRE) mice. Histological analyses revealed the presence of innate immune cells (i.e., macrophages, leukocytes) in ocular structures of the KO mice. At P20, the expression of cytokines and ROS content was higher in the lenses of KO mice than in those from age-matched CRE and CON mice, suggesting that oxidative stress may induce cytokine expression. In vitro administration of the oxidant, hydrogen peroxide, and the depletion of GSH (using buthionine sulfoximine (BSO)) in 21EM15 lens epithelial cells induced cytokine expression, an effect that was prevented by co-treatment of the cells with N-acetyl-l-cysteine (NAC), a antioxidant. The in vivo and ex vivo induction of cytokine expression by oxidative stress was associated with the expression of markers of epithelial-to-mesenchymal transition (EMT), α-SMA, in lens cells. Given that EMT of lens epithelial cells causes posterior capsule opacification (PCO), we propose that oxidative stress induces cytokine expression, EMT and the development of PCO in a positive feedback loop. Collectively these data indicate that oxidative stress induces inflammation of lens cells which promotes immune surveillance of ocular structures.


Subject(s)
Eye/anatomy & histology , Immunity, Innate , Lens, Crystalline/metabolism , Oxidative Stress , Acetylcysteine/pharmacology , Animals , Buthionine Sulfoximine/pharmacology , Cell Line , Chemokine CCL7/genetics , Chemokine CCL7/metabolism , Cytokines/genetics , Cytokines/metabolism , Down-Regulation/drug effects , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Eye/metabolism , Glutamate-Cysteine Ligase/deficiency , Glutamate-Cysteine Ligase/genetics , Lens, Crystalline/cytology , Leukocytes/cytology , Leukocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
11.
Curr Eye Res ; 47(4): 547-554, 2022 04.
Article in English | MEDLINE | ID: mdl-35179404

ABSTRACT

PURPOSE: The purpose of the study is to explore the mRNA and protein expression of FUNDC1 in cataract cells and tissues, and clarify the function and mechanism of FUNDC1 in cataract cells under oxidative stress. METHODS: We used bioinformatic analysis to screen differentially expressed genes in cataract cells from GSE153933. The expression of FUNDC1 in cataract specimens and cells was measured by reverse transcription quantitative polymerase chain reaction and western blotting. MethPrimer was used to predict CpG island of FUNDC1 promoter. The methylation of FUNDC1 in cataract specimens and cells was determined by methylation-specific polymerase chain reaction assay. Flow cytometry assay was used to measure cell apoptosis in FUNDC1-knockdown and -overexpression SRA01/04 cells. The expression of LC3 was analyzed by immunofluorescence assay. The expression of apoptosis-related proteins, autophagy, and PI3K/Akt/mTOR-related proteins was determined by western blotting. RESULTS: The results of bioinformatic analysis revealed that FUNDC1 was upregulated in cataract. FUNDC1 was high expression in SRA01/04 cells with H2O2 treatment, whereas hypomethylation of FUNDC1 in cataract lens cells under oxidative stress. The knockdown of FUNDC1 decreased cell apoptosis and autophagy in comparison with the negative control of SRA01/04 cells. While the overexpression of FUNDC1 elevated cell apoptosis and autophagy compared to the empty vector group in SRA01/04 cells. Mechanically, FUNDC1 reduced the phosphorylation of PI3K/Akt/mTOR pathway under oxidative stress in SRA01/04 cells. CONCLUSION: Our study suggested that FUNDC1 deficiency restrains cell apoptosis and autophagy by inhibiting PI3K/Akt/mTOR signal pathway.


Subject(s)
Cataract , Lens, Crystalline , Membrane Proteins , Mitochondrial Proteins , Oxidative Stress , Apoptosis , Autophagy , Cataract/genetics , Cell Line , Humans , Hydrogen Peroxide/pharmacology , Lens, Crystalline/cytology , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
12.
Mol Med Rep ; 25(3)2022 Mar.
Article in English | MEDLINE | ID: mdl-35039875

ABSTRACT

The aim of the present study was to explore the mechanism underlying the ultraviolet B (UVB) irradiation­induced apoptosis of human lens epithelial cells (HLECs), and to investigate the protective effect of epigallocatechin gallate (EGCG) against the UVB­induced apoptosis of HLECs. HLECs were exposed to different concentrations of EGCG plus UVB (30 mJ/cm2). Cell viability was determined using the MTT assay. Furthermore, mitochondrial membrane potential (Δψm) and apoptosis were assessed by flow cytometry with JC­1 and Annexin V/PI staining, respectively. Moreover, the activities of catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GSH­Px), as well as the levels of GSH, hydrogen peroxide (H2O2) and hydroxyl free radicals were determined using biochemical assay techniques. Reverse transcription­quantitative PCR and western blotting were used to detect the mRNA and protein expression levels of Bcl­2, Bax, cytochrome c, caspase­9 and caspase­3, respectively. The results revealed that UVB irradiation reduced the Δψm of HLECs and induced apoptosis. Notably, EGCG significantly attenuated the generation of H2O2 and hydroxyl free radicals caused by UVB irradiation in HLECs, and significantly increased CAT, SOD and GSH­Px activities, however, the GSH levels were not significantly increased. EGCG also reduced UVB­stimulated Bax, cytochrome c, caspase­9 and caspase­3 expression, and elevated Bcl­2 expression, suggesting that EGCG may possess free radical­scavenging properties, thus increasing cell viability. In conclusion, EGCG may be able to protect against UVB­induced HLECs apoptosis through the mitochondria­mediated apoptotic signaling pathway, indicating its potential application in clinical practice.


Subject(s)
Catechin/analogs & derivatives , Epithelial Cells/drug effects , Lens, Crystalline/cytology , Mitochondria/drug effects , Signal Transduction/drug effects , Ultraviolet Rays , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Blotting, Western , Caspases/genetics , Caspases/metabolism , Catalase/metabolism , Catechin/chemistry , Catechin/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Humans , Mitochondria/metabolism , Mitochondria/radiation effects , Molecular Structure , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/radiation effects , Superoxide Dismutase/metabolism
13.
Sci Rep ; 12(1): 1063, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058548

ABSTRACT

Binding of platelet-derived growth factor-BB (PDGF-BB) to its cognate receptor (PDGFR) promotes lens epithelial cell (LEC) proliferation and migration. After cataract surgery, these LEC behaviors have been proposed as an influential cause of posterior capsule opacification (PCO). Stimulated PDFGR undergoes dimerization and tyrosine phosphorylation providing docking sites for a SH2-domain-containing noncatalytic region of tyrosine kinase (Nck). Nck is an adaptor protein acting as a linker of the proximal and downstream signaling events. However, the functions of Nck1 protein in LEC have not been investigated so far. We reported here a crucial role of Nck1 protein in regulating PDGFR-mediated LEC activation using LEC with a silenced expression of Nck1 protein. The knockdown of Nck1 suppressed PDGF-BB-stimulated LEC proliferation and migration and disrupted the cell cycle progression especially G1/S transition. LEC lacking Nck1 protein failed to exhibit actin polymerization and membrane protrusions. The downregulation of Nck1 protein in LEC impaired PDGFR-induced phosphorylation of intracellular signaling proteins, including Erk1/2, Akt, CREB and ATF1, which resulted in inhibition of LEC responses. Therefore, these data suggest that the loss of Nck1 expression may disturb LEC activation and Nck1 may potentially be a drug target to prevent PCO and lens-related disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Becaplermin , Epithelial Cells/metabolism , Oncogene Proteins/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Capsule Opacification/etiology , Cell Line , Cell Proliferation , Epithelial Cells/cytology , Gene Silencing , Humans , Lens, Crystalline/cytology , Oncogene Proteins/genetics , Phosphorylation , Signal Transduction
14.
J Ethnopharmacol ; 284: 114783, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-34715300

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Capsella bursa-pastoris (L.) Medic. (CBP) is a cruciferous plant valuable in reducing fever, improving eyesight and calming the liver. This herb was recorded in the Compendium of Materia Medica for cataract treatment. AIM OF THE STUDY: To determine the effects and mechanism of CBP on cataract prevention and treatment using a selenite cataract model. MATERIALS AND METHODS: The main compounds in CBP extract were analyzed by UPLC, 1H-NMR and 13C-NMR spectroscopic techniques. Flavonoids formed a significant proportion of its compounds, thus necessitating an evaluation of their inhibitory effects on the development of cataract using a selenite cataract model. The protective effects of CBP flavonoids (CBPF) against oxidative damage and the modulation of mitochondrial apoptotic pathway were subsequently verified on H2O2-treated SRA01/04 lens epithelial cells. RESULTS: CBPF significantly alleviated the development of cataract by decreasing the MDA level and increasing the GSH-Px and SOD levels in the lens. It also inhibited H2O2-induced apoptosis in SRA01/04 cells, increased the expression of Bcl-2 protein and decreased the expressions of Caspase-3 and Bax proteins. CONCLUSION: CBPF exerts a significant preventive effect on cataract development by regulating the mitochondrial apoptotic pathway of the lens epithelial cells. It is thus a potent traditional Chinese medicine (TCM) whose application should be further developed for the clinical treatment of cataract.


Subject(s)
Capsella/chemistry , Cataract/prevention & control , Epithelial Cells/drug effects , Lens, Crystalline/cytology , Phytotherapy , Plant Extracts/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/genetics , Caspase 3/metabolism , Gene Expression Regulation/drug effects , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Hydrogen Peroxide , Malondialdehyde/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Plant Extracts/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
15.
Science ; 374(6573): eabk0410, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34882480

ABSTRACT

Cytokinetic membrane abscission is a spatially and temporally regulated process that requires ESCRT (endosomal sorting complexes required for transport)­dependent control of membrane remodeling at the midbody, a subcellular organelle that defines the cleavage site. Alteration of ESCRT function can lead to cataract, but the underlying mechanism and its relation to cytokinesis are unclear. We found a lens-specific cytokinetic process that required PI3K-C2α (phosphatidylinositol-4-phosphate 3-kinase catalytic subunit type 2α), its lipid product PI(3,4)P2 (phosphatidylinositol 3,4-bisphosphate), and the PI(3,4)P2­binding ESCRT-II subunit VPS36 (vacuolar protein-sorting-associated protein 36). Loss of each of these components led to impaired cytokinesis, triggering premature senescence in the lens of fish, mice, and humans. Thus, an evolutionarily conserved pathway underlies the cell type­specific control of cytokinesis that helps to prevent early onset cataract by protecting from senescence.


Subject(s)
Cataract/pathology , Cellular Senescence , Cytokinesis , Endosomal Sorting Complexes Required for Transport/metabolism , Lens, Crystalline/cytology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Aging, Premature , Animals , Biological Evolution , Calcium-Binding Proteins/metabolism , Cataract/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Humans , Lens, Crystalline/growth & development , Lens, Crystalline/metabolism , Mice , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Tubulin/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
16.
Invest Ophthalmol Vis Sci ; 62(15): 2, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34854886

ABSTRACT

Purpose: Sulforaphane (SFN) is a therapeutic phytochemical agent for many health conditions. SFN-induced cytotoxicity is shown to have promise in preventing posterior capsule opacification (PCO). In the current study, we aimed to elucidate key processes and mechanisms linking SFN treatment to lens cell death. Methods: The human lens epithelial cell line FHL124 and central anterior epithelium were used as experimental models. Cell death was assessed by microscopic observation and cell damage/viability assays. Gene or protein levels were assessed by TaqMan RT-PCR or immunoblotting. Mitochondrial networks and DNA damage were assessed by immunofluorescence. Mitochondrial membrane potential, activating transcription factor 6 (ATF6) activity, ratio of reduced glutathione (GSH) to oxidized glutathione (GSSG), and glutathione reductase (GR) activity were measured using different light reporter assays. SFN metabolites were analyzed by LC-MS/MS. Results: Treatment with N-acetylcysteine (NAC), a reactive oxygen species scavenger, prevented SFN-induced cell death in both models. NAC also significantly protected FHL124 cells from SFN-induced mitochondrial dysfunctions, endoplasmic reticulum stress (ERS), DNA damage and autophagy. SFN significantly depleted GSH, the major antioxidant in the eye, and reduced GR activity, despite doubling its protein levels. The most abundant SFN conjugate detected in lens cells following SFN application was SFN-GSH. The addition of GSH protected lens cells from all SFN-induced cellular events. Conclusions: SFN depletes GSH levels in lens cells through conjugation and inhibition of GR activity. This leads to increased reactive oxygen species and oxidative stress that trigger mitochondrial dysfunction, ERS, autophagy, and DNA damage, leading to cell death. In summary, the work presented provides a mechanistic understanding to support the therapeutic application of SFN for PCO and other disorders.


Subject(s)
Anticarcinogenic Agents/pharmacology , Biomarkers/metabolism , Epithelial Cells/drug effects , Glutathione/metabolism , Isothiocyanates/pharmacology , Lens, Crystalline/cytology , Sulfoxides/pharmacology , Acetylcysteine/pharmacology , Activating Transcription Factor 6/metabolism , Aged , Aged, 80 and over , Apoptosis/drug effects , Cell Line , Cell Survival , Chromatography, Liquid , Epithelial Cells/metabolism , Epithelial Cells/pathology , Free Radical Scavengers/pharmacology , Glutathione Disulfide/metabolism , Glutathione Reductase/metabolism , Humans , Immunoblotting , Membrane Potential, Mitochondrial/physiology , Middle Aged , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry
17.
Exp Eye Res ; 213: 108840, 2021 12.
Article in English | MEDLINE | ID: mdl-34798144

ABSTRACT

Posterior capsule opacification (PCO), the most common complication of cataract surgery occurring in 20-50% of patients after 2-5 years of cataract surgery, is a major problem in the aging society. The epithelial-mesenchymal transition (EMT) of lens epithelial cells after cataract surgery has been proposed as a major cause of PCO. Capsaicin, widely used as a food additive and analgesic agent, is a major pungent ingredient in red pepper. Although the effect of capsaicin on EMT has been reported in cancer cells, the biological reaction of capsaicin was unique in each cell type, and there have been no reports describing its effects on EMT earlier. In this study, we demonstrated that treatment with capsaicin inhibited TGFß2-induced EMT in vitro lens epithelial cells and ex vivo explant lens epithelial cells. Furthermore, eye drops of capsaicin inhibited the PCO model mice in vivo. Finally, we showed that capsaicin inhibited non-canonically induced Smad2/3 activation via suppression of EGFR activation and ERK phosphorylation. Our findings indicate that capsaicin and its derivatives are good candidate compounds for preventing PCO after cataract surgery.


Subject(s)
Capsaicin/pharmacology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lens, Crystalline/cytology , Sensory System Agents/pharmacology , Transforming Growth Factor beta2/antagonists & inhibitors , Actins/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Epithelial Cells/metabolism , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Smad2 Protein/metabolism , Transforming Growth Factor beta2/pharmacology , Wound Healing/drug effects
18.
Bioengineered ; 12(1): 7213-7222, 2021 12.
Article in English | MEDLINE | ID: mdl-34558381

ABSTRACT

Cataract is a disease that causes severe visual impairment in patients. Recent studies have found that lens epithelial cell apoptosis caused by oxidative damage is the critical cause of cataract. Moreover, TRIM22 could alleviate the ubiquitination of TRAF6. The expression of TRAF6 could activate the p38/MAPK pathway and aggravate the oxidative stress induced damage of lens epithelial cells. However, whether the TRIM22 could alleviate the oxidative stress induced damage of lens epithelial cells by regulating the expression of TRAF6 and p38/MAPK pathway is unclear. In this study, we stimulated the lens epithelial cells with the H2O2 and established the TRIM22 knockdown cells. Next, proliferation of these cells was determined by CCK-8 and EdU assays. Apoptosis of these cells was detected with the TUNEL assays. Levels of ROS was explored with the DCFH-DA staining. Finally, the expression levels of TRAF6, p-p38 and p-ERK were determined with the western blotting. According to the results, we found that knockdown of TRIM22 suppressed the proliferation and relieved the H2O2 induced DNA double-strand break and apoptosis of these cells. Inhibition of TRIM22 inhibited the production of ROS in these cells. Moreover, restriction of TRIM22 induced the decreased levels of TRAF6, p-p38 and p-ERK in lens epithelial cells. We concluded that inhibition of TRIM22 relieved the apoptosis of lens epithelial cells by suppressing the expression of TRAF6, p-p38 and p-ERK.


Subject(s)
Apoptosis/genetics , Epithelial Cells/cytology , Intracellular Signaling Peptides and Proteins/genetics , Lens, Crystalline/cytology , Minor Histocompatibility Antigens/genetics , Repressor Proteins/genetics , Tripartite Motif Proteins/genetics , Cataract , Cell Line , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Minor Histocompatibility Antigens/metabolism , Oxidative Stress/genetics , Repressor Proteins/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitination/genetics
19.
Exp Eye Res ; 212: 108768, 2021 11.
Article in English | MEDLINE | ID: mdl-34534541

ABSTRACT

The mouse lens is frequently used both in vivo and ex vivo in ophthalmic research to model conditions affecting the human lens, such as presbyopia. The mouse lens has a delicate structure which is prone to damage and biomechanical changes both before and after extraction from the whole globe. When not properly controlled for, these changes can confound the biomechanical analysis of mouse lenses. In this study, atomic force microscopy microindentation was used to assess changes in the Young's Modulus of Elasticity of the mouse lens as a function of mouse age and postmortem time. Old mouse lenses measured immediately postmortem were significantly stiffer than young mouse lenses (p = 0.028). However, after 18 h of incubation, there was no measurable difference in lens stiffness between old and young mouse lenses (p = 0.997). This demonstrates the need for careful experimental control in experiments using the mouse lens, especially regarding postmortem time.


Subject(s)
Aging , Lens Capsule, Crystalline/physiology , Lens, Crystalline/physiology , Microscopy, Atomic Force/methods , Animals , Elasticity , Female , Lens Capsule, Crystalline/cytology , Lens, Crystalline/cytology , Mice , Models, Animal
20.
Arch Biochem Biophys ; 711: 109024, 2021 10 30.
Article in English | MEDLINE | ID: mdl-34487720

ABSTRACT

This study attempted to determine the effect of EphA2 on H2O2-treated lens epithelial cells (SRA01/04) and the underlying mechanisms. MTT assay and flow cytometry were performed to assess cell viability and cell apoptosis. Western blot was carried out to examine the levels of proteins associated with apoptosis and autophagy. Our results revealed that EphA2 significantly elevated the reduced cell viability, and inhibited the increased cell apoptosis in H2O2-treated SRA01/04 cells, along with the significant up-regulated Bcl-2 and down-regulated Cleaved-caspase-3 and Bax protein levels, but which were all abolished by Rapa (autophagy activator). We also found that EphA2 significantly suppressed cell autophagy in H2O2-treated SRA01/04 cells. Additionally, EphA2 significantly up-regulated the protein levels of p-Akt and p-mTOR in H2O2-treated SRA01/04 cells, and the inhibition of Akt by MK-2206 and inhibition of mTOR by Rapa both obviously reversed EphA2-mediated the inhibition of autophagy in H2O2-treated SRA01/04 cells. In summary, these data demonstrated that EphA2 inhibited the apoptosis of SRA01/04 cells by inhibiting autophagy via activating PI3K/Akt/mTOR pathway.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Receptor, EphA2/metabolism , Signal Transduction/physiology , Apoptosis/drug effects , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , Lens, Crystalline/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...