1
|
Li J, Liu J, Tang Y, Zhang H, Zhang Y, Zha X, Zhao X. Role of C/EBP Homologous Protein (CHOP) and Nupr1 Interaction in Endoplasmic Reticulum Stress-Induced Apoptosis of Lens Epithelial Cells. Mol Biotechnol 2025; 67:1628-1640. [PMID: 38771421 PMCID: PMC11928426 DOI: 10.1007/s12033-024-01148-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 05/22/2024]
Abstract
Our study mainly analyzed the mechanism of C/EBP homologous protein (CHOP) and its interacting protein Nupr1 on endoplasmic reticulum stress (ERS) induced lens epithelial cells (LEC) apoptosis. Cell proliferation was detected by CCK-8. Apoptosis was detected by flow cytometry and TUNEL. Nupr1 expression was detected by RT-qPCR. The expressions of CHOP, Nupr1, apoptosis-related protein, and ERS-related protein were detected by Western blot. DCFH-DA probe was used to detect cell ROS. The SOD, GSH-PX, and MDA contents were detected by the kit. Co-IP was used to detect the interaction between CHOP and Nupr1. The morphology of the lens was detected by HE staining. The result shows that Tunicamycin (TU) can induce endoplasmic reticulum stress and apoptosis in LEC in a concentration-dependent manner. TU induction leads to the occurrence of CHOP nuclear translocation. Overexpression of CHOP can further enhance the inhibitory effect of TU on LEC proliferation and the promotion of apoptosis, while knockdown of CHOP has the opposite effect. CHOP and Nupr1 are interacting proteins, and knockdown of Nupr1 or addition of Nupr1 inhibitor ZZW-115 can reverse the effects of TU and overexpression of CHOP, respectively. It has been observed in animal experiments that treatment with oe-CHOP can further aggravate the pathological lesions of the rat lens, while ZZW-115 can reverse the effect of oe-CHOP to a certain extent and improve the lesions of the rat lens. Overall, CHOP interacts with Nupr1 to regulate apoptosis caused by ERS and mediate cataract progression in rats, and this study provides a new potential therapeutic target for the treatment of cataract.
Collapse
Affiliation(s)
- Jinghua Li
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Junyi Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Yongying Tang
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Hong Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Yuanping Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Xu Zha
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China.
| | - Xueying Zhao
- Department of Ophthalmology, The Second Affiliated Hospital of Kunming Medical University, No.374 Yunnan-Burma Avenue, Wuhua District, Kunming, 650000, Yunnan, China.
| |
Collapse
|
2
|
Sellitto C, White TW. Combinatorial genetic manipulation of Cx50, PI3K and PTEN alters postnatal mouse lens growth and homeostasis. FRONTIERS IN OPHTHALMOLOGY 2025; 5:1502836. [PMID: 40046897 PMCID: PMC11879993 DOI: 10.3389/fopht.2025.1502836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
Introduction Phosphoinositide 3-kinase (PI3K), Phosphatase and tensin homolog (PTEN) and connexin50 (Cx50) have individually been shown to play critical roles in the growth, development and maintenance of the lens and to functionally interact in vitro. To elucidate how gap junctional coupling mediated by Cx50 and intracellular signaling mediated by PI3K and PTEN synergistically interact to regulate lens homeostasis in vivo, we generated and characterized double knockout animal models lacking the p110α subunit of PI3K and Cx50, or PTEN and Cx50. Methods We interbred lens specific p110α and PTEN conditional knockout animals with Cx50 deficient mice to generate double knockouts. Animals and eyes were weighed, lenses were dissected, photographed, measured, fixed and sectioned for histological analysis. Lens epithelial cell proliferation was determined using 5-ethynyl-2'-deoxyuridine (EdU) labeling. Results Double knockout of p110α and Cx50 led to a significant reduction in lens and eye size, and a high rate of lens rupture. The individual cell proliferation defects of the Cx50 and p110α single knockout lenses both persisted in the double KO. Double deletion of Cx50 and PTEN produced severe lens defects, including cataract, aberrant cell migration, altered cell proliferation, vacuole formation and lens rupture. Conclusion The severe phenotypes in p110α/Cx50 and PTEN/Cx50 double deficient lenses suggest that PI3K, PTEN and Cx50 participate in both distinct and common regulatory pathways that are necessary to maintain normal lens growth and homeostasis.
Collapse
Affiliation(s)
| | - Thomas W. White
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, United States
| |
Collapse
|
3
|
Zhang K, Chen L, Qu L, Yan H. A comprehensive investigation of identifying miRNA biomarkers and their potential role in age-related cataract by meta-analysis and bioinformatics analysis. Graefes Arch Clin Exp Ophthalmol 2025:10.1007/s00417-024-06723-3. [PMID: 39760860 DOI: 10.1007/s00417-024-06723-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 10/19/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025] Open
Abstract
PURPOSE Age-related cataract (ARC) remains one of the leading causes of blindness globally. Despite the satisfactory outcomes of surgical interventions, significant disparities in access to medical care prevent many patients from receiving effective treatment. Thus, identifying reliable biomarkers and therapeutic targets to expand treatment options for ARC is essential. Recent evidence indicates that microRNAs (miRNAs) play a role in the development of cataracts and may serve as promising biomarkers. Consequently, this study aims to investigate miRNAs' levels and potential functions in ARC. METHODS We conducted a meta-analysis following the PRISMA guidelines by searching three databases from inception to March 31, 2023. The quality of the articles was assessed using the NOS. Subsequently, the targets of the miRNAs identified in the meta-analysis were predicted using six databases, and their GO functions and KEGG pathway enrichment information were analyzed via DAVID. RESULTS An initial search yielded 225 publications, from which 22 miRNAs across 37 studies were selected for our meta-analysis. We identified eight differentially expressed miRNAs (DEmiRNAs) in ARC, comprising two up-regulated miRNAs (miR-124 and miR-125a) and six down-regulated miRNAs (miR-15a, miR-23b, miR-34a, miR-221, miR-222, and miR-378a). A total of 972 targets for these miRNAs have been confirmed, and subsequent bioinformatics analysis has revealed their potential functions and pathways in various ARC-related processes. CONCLUSIONS This study indicates that eight differentially expressed miRNAs (miRNA-15a, miRNA-23b, miRNA-34a, miRNA-124, miRNA-125a, miRNA-221, miRNA-222, and miRNA-378a) may serve as biomarkers for ARC. Bioinformatics analyses suggest varied potential roles for each miRNA, providing a framework for future research in ARC. This systematic evaluation represents the initial depiction of the miRNA-biomarker landscape in ARC. KEY MESSAGES What is known MicroRNAs(miRNAs) could serve as biomarkers for age-related cataract(ARC) since their abundances are associated with ARC and can play a role in cataractogenesis. However, existing studies have reported inconsistent results regarding the miRNA level in ARC. Therefore, achieving a consensus on the role of miRNAs in ARC is essential to clarify their involvement. What is new This study suggested that eight differentially expressed miRNAs (miRNA-15a, miRNA-23b, miRNA-34a, miRNA-124, miRNA-125a, miRNA-221, miRNA-222, and miRNA-378a) may serve as biomarkers for ARC. Our bioinformatics analysis identified various potential roles for each miRNA, which could guide future research on ARC.
Collapse
Affiliation(s)
- Kaiyun Zhang
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, No. 21 Jiefang Road, Xi'an, Shaanxi Province, 710004, China
| | - Li Chen
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, No. 21 Jiefang Road, Xi'an, Shaanxi Province, 710004, China
| | - Laiqiang Qu
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, No. 21 Jiefang Road, Xi'an, Shaanxi Province, 710004, China
| | - Hong Yan
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, No. 21 Jiefang Road, Xi'an, Shaanxi Province, 710004, China.
| |
Collapse
|
4
|
Xie P, He J, Ou Y. RBM15 Promotes High Glucose-Induced Lens Epithelial Cell Injury by Inducing PRNP N6-Methyladenine Modification During Diabetic Cataract. Curr Eye Res 2024; 49:1145-1153. [PMID: 39206850 DOI: 10.1080/02713683.2024.2362855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/15/2024] [Accepted: 05/28/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Diabetic cataract (DC) is a major cause of blindness worldwide. Prion protein (PRNP) was proved to be up-regulated and hypomethylated in DC samples. Here, we investigated whether PRNP was involved in DC progression in N6-methyladenosine (m6A)-dependent manner, and its potential mechanisms. METHODS Levels of genes and proteins were assayed using qRT-PCR and western blotting. Cell proliferation and apoptosis were determined using Cell Counting Kit-8 assay, 5-thynyl-2'-deoxyuridine (EdU) assay, and flow cytometry, respectively. Oxidative stress was analyzed by measuring the production of glutathione peroxidase (GSH-PX), superoxide dismutase (SOD), and malondialdehyde (MDA). The m6A modification was determined by RNA immunoprecipitation (Me-RIP) assay. The interaction between RBM15 (RNA binding motif protein 15) and PRNP was probed using RIP assay. RESULTS PRNP was highly expressed in DC patients and HG-induced HLECs. Functionally, PRNP deficiency reversed HG-induced apoptosis and oxidative stress in HLECs. Mechanistically, RBM15 induced PRNP m6A modification and directly bound to PRNP. Knockdown of RBM15 abolished HG-induced apoptotic and oxidative injury in HLECs, while these effects were rescued after PRNP overexpression. CONCLUSION RBM15 silencing suppressed HG-induced lens epithelial cell injury by regulating PRNP in an m6A-mediated manner, hinting a novel therapeutic strategy for DC patients.
Collapse
Affiliation(s)
- Ping Xie
- Department of Ophthalmology, Jiujiang City Key Laboratory of Cell Therapy, JiuJiang NO.1 People's Hospital, Jiujiang, Jiangxi, China
| | - Jing He
- Department of Ophthalmology, Jiujiang City Key Laboratory of Cell Therapy, JiuJiang NO.1 People's Hospital, Jiujiang, Jiangxi, China
| | - Yangjun Ou
- Department of Ophthalmology, Jiujiang City Key Laboratory of Cell Therapy, JiuJiang NO.1 People's Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
5
|
Perez RC, Yang X, Familari M, Martinez G, Lovicu FJ, Hime GR, de Iongh RU. TOB1 and TOB2 mark distinct RNA processing granules in differentiating lens fiber cells. J Mol Histol 2024; 55:121-138. [PMID: 38165569 DOI: 10.1007/s10735-023-10177-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/12/2023] [Indexed: 01/04/2024]
Abstract
Differentiation of lens fiber cells involves a complex interplay of signals from growth factors together with tightly regulated gene expression via transcriptional and post-transcriptional regulators. Various studies have demonstrated that RNA-binding proteins, functioning in ribonucleoprotein granules, have important roles in regulating post-transcriptional expression during lens development. In this study, we examined the expression and localization of two members of the BTG/TOB family of RNA-binding proteins, TOB1 and TOB2, in the developing lens and examined the phenotype of mice that lack Tob1. By RT-PCR, both Tob1 and Tob2 mRNA were detected in epithelial and fiber cells of embryonic and postnatal murine lenses. In situ hybridization showed Tob1 and Tob2 mRNA were most intensely expressed in the early differentiating fibers, with weaker expression in anterior epithelial cells, and both appeared to be downregulated in the germinative zone of E15.5 lenses. TOB1 protein was detected from E11.5 to E16.5 and was predominantly detected in large cytoplasmic puncta in early differentiating fiber cells, often co-localizing with the P-body marker, DCP2. Occasional nuclear puncta were also observed. By contrast, TOB2 was detected in a series of interconnected peri-nuclear granules, in later differentiating fiber cells of the inner cortex. TOB2 did not appear to co-localize with DCP2 but did partially co-localize with an early stress granule marker (EIF3B). These data suggest that TOB1 and TOB2 are involved with different aspects of the mRNA processing cycle in lens fiber cells. In vitro experiments using rat lens epithelial explants treated with or without a fiber differentiating dose of FGF2 showed that both TOB1 and TOB2 were up-regulated during FGF-induced differentiation. In differentiating explants, TOB1 also co-localized with DCP2 in large cytoplasmic granules. Analyses of Tob1-/- mice revealed relatively normal lens morphology but a subtle defect in cell cycle arrest of some cells at the equator and in the lens fiber mass of E13.5 embryos. Overall, these findings suggest that TOB proteins play distinct regulatory roles in RNA processing during lens fiber differentiation.
Collapse
Affiliation(s)
- Rafaela C Perez
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Xenia Yang
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Mary Familari
- School of Biosciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Gemma Martinez
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Frank J Lovicu
- Molecular and Cellular Biomedicine, School of Medical Sciences and Save Sight Institute, University of Sydney, Sydney, NSW, 2006, Australia
| | - Gary R Hime
- Stem Cell Genetics Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Robb U de Iongh
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
6
|
Giannone AA, Sellitto C, Rosati B, McKinnon D, White TW. Single-Cell RNA Sequencing Analysis of the Early Postnatal Mouse Lens Epithelium. Invest Ophthalmol Vis Sci 2023; 64:37. [PMID: 37870847 PMCID: PMC10599162 DOI: 10.1167/iovs.64.13.37] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
Purpose The lens epithelium maintains the overall health of the organ. We used single-cell RNA sequencing (scRNA-seq) technology to assess transcriptional heterogeneity between cells in the postnatal day 2 (P2) epithelium and identify distinct epithelial cell subtypes. Analysis of these data was used to better understand lens growth, differentiation, and homeostasis on P2. Methods scRNA-seq on P2 mouse lenses was performed using the 10x Genomics Chromium Single Cell 3' Kit (v3.1) and short-read Illumina sequencing. Sequence alignment and preprocessing of data were conducted using 10x Genomics Cell Ranger software. Seurat was employed for preprocessing, quality control, dimensionality reduction, and cell clustering, and Monocle was utilized for trajectory analysis to understand the developmental progression of the lens cells. CellChat and GO analyses were used to explore cell-cell communication networks and signaling interactions. Results Lens epithelial cells (LECs) were divided into seven subclusters, classified by specific gene markers. The expression of crystallin, cell-cycle, and metabolic genes was not uniform, indicating distinct functional roles of LECs. Trajectory analysis predicted a bifurcation of differentiating and cycling cells from an Igfbp5+ progenitor pool. We also identified heterogeneity in signaling molecules and pathways, suggesting that cycling and progenitor subclusters have prominent roles in coordinating crosstalk. Conclusions scRNA-seq corroborated many known markers of epithelial differentiation and proliferation while providing further insight into the pathways and genes directing these processes. Interestingly, we demonstrated that the developing epithelium can be divided into distinct subpopulations. These clusters reflect the transcriptionally diverse roles of the epithelium in proliferation, signaling, and maintenance.
Collapse
Affiliation(s)
- Adrienne A. Giannone
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Caterina Sellitto
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Barbara Rosati
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
- Veterans Affairs Medical Center, Northport, New York, United States
| | - David McKinnon
- Department of Neurobiology and Behavior, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Thomas W. White
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| |
Collapse
|
7
|
Feng L, Wei Y, Sun Y, Zhou L, Bi S, Chen W, Xiang W. MIR34A modulates lens epithelial cell apoptosis and cataract development via the HK1/caspase 3 signaling pathway. Aging (Albany NY) 2023; 15:6331-6345. [PMID: 37414399 PMCID: PMC10373963 DOI: 10.18632/aging.204854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/15/2023] [Indexed: 07/08/2023]
Abstract
Cataracts are the leading cause of blindness in the world. Age is a major risk factor for cataracts, and with increasing aging, the burden of cataracts will grow, but the exact details of cataractogenesis remain unclear. A recent study showed that microRNA-34a (MIR34A) is involved in the development of cataracts, but the underlying pathogenesis remains obscure. Here, our results of microRNA target prediction showed that hexokinase 1 (HK1) is one of the genes targeted by MIR34A. Based on this finding, we focused on the function of MIR34A and HK1 in the progress of cataracts, whereby the human lens epithelial cell line SRA01/04 and mouse lens were treated with MIR34A mimics and HK1 siRNA. We found that HK1 mRNA is a direct target of MIR34A, whereby the high expression of MIR34A in the cataract lens suppresses the expression of HK1. In vitro, the upregulation of MIR34A together with the downregulation of HK1 inhibits the proliferation, induces the apoptosis of SRA01/04 cells, and accelerates the opacification of mouse lenses via the HK1/caspase 3 signaling pathway. In summary, our study demonstrates that MIR34A modulates lens epithelial cell (LEC) apoptosis and cataract development through the HK1/caspase 3 signaling pathway.
Collapse
Affiliation(s)
- Lujia Feng
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science; Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, China
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518040, China
| | - Yantao Wei
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science; Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, China
| | - Yimeng Sun
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science; Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, China
| | - Linbin Zhou
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science; Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, China
| | - Shaowei Bi
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science; Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, China
| | - Weirong Chen
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science; Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, China
| | - Wu Xiang
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science; Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, China
| |
Collapse
|
8
|
Zhu J, Chen J, Amjadian M, Liang S, Qu Z, Wang Y, Zhang Y, Wang L. Simultaneous dual-modal photoacoustic and harmonic ultrasound microscopy with an optimized acoustic combiner. BIOMEDICAL OPTICS EXPRESS 2023; 14:1626-1635. [PMID: 37078044 PMCID: PMC10110316 DOI: 10.1364/boe.484038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 05/03/2023]
Abstract
Simultaneous photoacoustic (PA) and ultrasound (US) imaging provides rich optical and acoustic contrasts with high sensitivity, specificity, and resolution, making it a promising tool for diagnosing and assessing various diseases. However, the resolution and penetration depth tend to be contradictory due to the increased attenuation of high-frequency ultrasound. To address this issue, we present simultaneous dual-modal PA/US microscopy with an optimized acoustic combiner that can maintain high resolution while improving the penetration of ultrasound imaging. A low-frequency ultrasound transducer is used for acoustic transmission, and a high-frequency transducer is used for PA and US detection. An acoustic beam combiner is utilized to merge the transmitting and receiving acoustic beams with a predetermined ratio. By combining the two different transducers, harmonic US imaging and high-frequency photoacoustic microscopy are implemented. In vivo experiments on the mouse brain demonstrate the simultaneous PA and US imaging ability. The harmonic US imaging of the mouse eye reveals finer iris and lens boundary structures than conventional US imaging, providing a high-resolution anatomical reference for co-registered PA imaging.
Collapse
Affiliation(s)
- Jingyi Zhu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
| | - Jiangbo Chen
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
| | - Mohammadreza Amjadian
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Siyi Liang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
| | - Zheng Qu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
| | - Yue Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
| | - Yachao Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
| | - Lidai Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
- City University of Hong Kong Shenzhen Research Institute, Yuexing Yi Dao, Nanshan District, China
| |
Collapse
|
9
|
FGF-2 Differentially Regulates Lens Epithelial Cell Behaviour during TGF-β-Induced EMT. Cells 2023; 12:cells12060827. [PMID: 36980168 PMCID: PMC10046997 DOI: 10.3390/cells12060827] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Fibroblast growth factor (FGF) and transforming growth factor-beta (TGF-β) can regulate and/or dysregulate lens epithelial cell (LEC) behaviour, including proliferation, fibre differentiation, and epithelial–mesenchymal transition (EMT). Earlier studies have investigated the crosstalk between FGF and TGF-β in dictating lens cell fate, that appears to be dose dependent. Here, we tested the hypothesis that a fibre-differentiating dose of FGF differentially regulates the behaviour of lens epithelial cells undergoing TGF-β-induced EMT. Postnatal 21-day-old rat lens epithelial explants were treated with a fibre-differentiating dose of FGF-2 (200 ng/mL) and/or TGF-β2 (50 pg/mL) over a 7-day culture period. We compared central LECs (CLECs) and peripheral LECs (PLECs) using immunolabelling for changes in markers for EMT (α-SMA), lens fibre differentiation (β-crystallin), epithelial cell adhesion (β-catenin), and the cytoskeleton (alpha-tropomyosin), as well as Smad2/3- and MAPK/ERK1/2-signalling. Lens epithelial explants cotreated with FGF-2 and TGF-β2 exhibited a differential response, with CLECs undergoing EMT while PLECs favoured more of a lens fibre differentiation response, compared to the TGF-β-only-treated explants where all cells in the explants underwent EMT. The CLECs cotreated with FGF and TGF-β immunolabelled for α-SMA, with minimal β-crystallin, whereas the PLECs demonstrated strong β-crystallin reactivity and little α-SMA. Interestingly, compared to the TGF-β-only-treated explants, α-SMA was significantly decreased in the CLECs cotreated with FGF/TGF-β. Smad-dependent and independent signalling was increased in the FGF-2/TGF-β2 co-treated CLECs, that had a heightened number of cells with nuclear localisation of Smad2/3 compared to the PLECs, that in contrast had more pronounced ERK1/2-signalling over Smad2/3 activation. The current study has confirmed that FGF-2 is influential in differentially regulating the behaviour of LECs during TGF-β-induced EMT, leading to a heterogenous cell population, typical of that observed in the development of post-surgical, posterior capsular opacification (PCO). This highlights the cooperative relationship between FGF and TGF-β leading to lens pathology, providing a different perspective when considering preventative measures for controlling PCO.
Collapse
|
10
|
Yao Y, Wei L, Chen Z, Li H, Qi J, Wu Q, Zhou X, Lu Y, Zhu X. Single-cell RNA sequencing: Inhibited Notch2 signalling underlying the increased lens fibre cells differentiation in high myopia. Cell Prolif 2023:e13412. [PMID: 36717696 PMCID: PMC10392066 DOI: 10.1111/cpr.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/26/2022] [Accepted: 01/18/2023] [Indexed: 02/01/2023] Open
Abstract
High myopia is the leading cause of blindness worldwide. It promotes the overgrowth of lens, which is an important component of ocular refractive system, and increases the risks of lens surgery. While postnatal growth of lens is based on the addition of lens fibre cells (LFCs) supplemented by proliferation and differentiation of lens epithelial cells (LECs), it remains unknown how these cellular processes change in highly myopic eyes and what signalling pathways may be involved. Single-cell RNA sequencing was performed and a total of 50,375 single cells isolated from the lens epithelium of mouse highly myopic and control eyes were analysed to uncover their underlying transcriptome atlas. The proportion of LFCs was significantly higher in highly myopic eyes. Meanwhile, Notch2 signalling was inhibited during lineage differentiation trajectory towards LFCs, while Notch2 predominant LEC cluster was significantly reduced in highly myopic eyes. In consistence, Notch2 was the top down-regulated gene identified in highly myopic lens epithelium. Further validation experiments confirmed NOTCH2 downregulation in the lens epithelium of human and mouse highly myopic eyes. In addition, NOTCH2 knockdown in primary human and mouse LECs resulted in enhanced differentiation towards LFCs accompanied by up-regulation of MAF and CDKN1C. These findings indicated an essential role of NOTCH2 inhibition in lens overgrowth of highly myopic eyes, suggesting a therapeutic target for future interventions.
Collapse
Affiliation(s)
- Yunqian Yao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,National Health Center Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Research Center of Ophthalmology and Optometry, Shanghai, China
| | - Ling Wei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,National Health Center Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hao Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,National Health Center Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jiao Qi
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,National Health Center Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Qingfeng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China.,Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xingtao Zhou
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,National Health Center Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Research Center of Ophthalmology and Optometry, Shanghai, China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,National Health Center Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Xiangjia Zhu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,National Health Center Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Ma J, Ye W, Yang Y, Wu T, Wang Y, Li J, Pei R, He M, Zhang L, Zhou J. The interaction between autophagy and the epithelial-mesenchymal transition mediated by NICD/ULK1 is involved in the formation of diabetic cataracts. Mol Med 2022; 28:116. [PMID: 36104669 PMCID: PMC9476327 DOI: 10.1186/s10020-022-00540-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cataracts are the leading cause of blindness and a common ocular complication of diabetes. The epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) and altered autophagic activity occur during the development of diabetic cataracts. The disturbed interaction of autophagy with EMT in LECs stimulated by high glucose levels may participate in cataract formation.
Methods
A rat diabetic cataract model induced by streptozotocin (STZ) and human lens epithelial cells (HLE-B3) stimulated with a high glucose concentration were employed in the study. These models were treated with rapamycin (an inhibitor of mammalian target of rapamycin (mTOR)), and N-(N-[3,5-difluorophenacetyl]-1-alanyl)-S-phenylglycine t-butyl ester (DAPT, an inhibitor of γ-secretase) alone or in combination. Lens opacity was observed and photographed under a slit-lamp microscope. Histological changes in paraffin sections of lenses were detected under a light microscope after hematoxylin and eosin staining. Alterations of autophagosomes in LECs were counted and evaluated under a transmission electron microscope. The expression levels of proteins involved in the EMT, autophagy, and the signaling pathways in LECs were measured using Western blotting and immunofluorescence staining. Cell migration was determined by performing transwell and scratch wound assays. Coimmunoprecipitation (Co-IP) was performed to verify protein-protein interactions. Proteins were overexpressed in transfected cells to confirm their roles in the signaling pathways of interest.
Results
In LECs, a high glucose concentration induces the EMT by activating Jagged1/Notch1/Notch intracellular domain (NICD)/Snail signaling and inhibits autophagy through the AKT/mTOR/unc 51-like kinase 1 (ULK1) signaling pathway in vivo and in vitro, resulting in diabetic cataracts. Enhanced autophagic activity induced by rapamycin suppressed the EMT by inducing Notch1 degradation by SQSTM1/p62 and microtubule-associated protein light chain 3 (LC3) in LECs, while inhibition of the Notch signaling pathway with DAPT not only prevented the EMT but also activated autophagy by decreasing the levels of NICD, which bound to ULK1, phosphorylated it, and then inhibited the initiation of autophagy.
Conclusions
We describe a new interaction of autophagy and the EMT involving NICD/ULK1 signaling, which mediates crosstalk between these two important events in the formation of diabetic cataracts. Activating autophagy and suppressing the EMT mutually promote each other, revealing a potential target and strategy for the prevention of diabetic cataracts.
Collapse
|
12
|
Ma B, Ni N, Shao W, Xu J, Ji J, Luo M. Bit1 is involved in regulation between integrin and TGFβ signaling in lens epithelial cells. Cell Cycle 2022; 21:2283-2297. [PMID: 35737738 PMCID: PMC9586669 DOI: 10.1080/15384101.2022.2092818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 04/03/2022] [Accepted: 06/16/2022] [Indexed: 11/03/2022] Open
Abstract
Bit1, as an integrin-specific effector, is specifically expressed in lens epithelial cells (LECs) and may be essential to maintain the normal function of LECs. The present study investigated the function of Bit1 and its regulatory mechanism in LECs. Knockdown of Bit1 was mediated by a lentivirus with a specific short-hairpin RNA against Bit1 in SRA01/04 cells. Cell proliferation ability was measured by CCK-8 assay. Cell migration was examined by transwell and wound-healing assays. The effect of Bit1 knockdown on genome-wide expression patterns was studied via a GeneChip® PrimeView™ Human Gene Expression Array. Based on the ingenuity pathway analysis (IPA), Bit1's regulation of target pathways and genes was verified by real-time qPCR and Western blotting. Bit1 knockdown inhibited proliferation, migration, and regulated cell cycle and apoptosis of LECs. Microarray gene expression analysis and IPA assays revealed that integrin and TGFβ signaling pathways were remarkably impacted by Bit1 expression. FAK, PAK2, ITGA5, and ITGB1 were identified as core node molecules under the control of Bit1. Bit1 participates in integrin and TGFβ signaling via regulating downstream FAK and PAK2 and subsequently affecting EMT-related gene expression including ITGA5, ITGB1, and αSMA. In conclusion, Bit1 plays as an important role in the regulation between integrin and TGFβ signaling, which affects cell survival, migration, and EMT of LECs.
Collapse
Affiliation(s)
- Bo Ma
- Department of Ophthalmology, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ni Ni
- Department of Ophthalmology, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanyu Shao
- Department of Ophthalmology, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingying Xu
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, China
| | - Jiali Ji
- Department of Ophthalmology, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Luo
- Department of Ophthalmology, Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Gao L, Jin N, Ye Z, Ma T, Huang Y, Li H, Du J, Li Z. A possible connection between reactive oxygen species and the unfolded protein response in lens development: From insight to foresight. Front Cell Dev Biol 2022; 10:820949. [PMID: 36211466 PMCID: PMC9535091 DOI: 10.3389/fcell.2022.820949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/31/2022] [Indexed: 11/28/2022] Open
Abstract
The lens is a relatively special and simple organ. It has become an ideal model to study the common developmental characteristics among different organic systems. Lens development is a complex process influenced by numerous factors, including signals from the intracellular and extracellular environment. Reactive oxygen species (ROS) are a group of highly reactive and oxygen-containing molecules that can cause endoplasmic reticulum stress in lens cells. As an adaptive response to ER stress, lens cells initiate the unfolded protein response (UPR) to maintain normal protein synthesis by selectively increasing/decreasing protein synthesis and increasing the degradation of misfolded proteins. Generally, the UPR signaling pathways have been well characterized in the context of many pathological conditions. However, recent studies have also confirmed that all three UPR signaling pathways participate in a variety of developmental processes, including those of the lens. In this review, we first briefly summarize the three stages of lens development and present the basic profiles of ROS and the UPR. We then discuss the interconnections between lens development and these two mechanisms. Additionally, the potential adoption of human pluripotent stem-cell-based lentoids in lens development research is proposed to provide a novel perspective on future developmental studies.
Collapse
Affiliation(s)
- Lixiong Gao
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ni Jin
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Endocrinology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, The Chinese PLA General Hospital, Beijing, China
| | - Zi Ye
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tianju Ma
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yang Huang
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongyu Li
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jinlin Du
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhaohui Li
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhaohui Li,
| |
Collapse
|
14
|
Sellitto C, Li L, White TW. Double Deletion of PI3K and PTEN Modifies Lens Postnatal Growth and Homeostasis. Cells 2022; 11:cells11172708. [PMID: 36078116 PMCID: PMC9455000 DOI: 10.3390/cells11172708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
We have previously shown that the conditional deletion of either the p110α catalytic subunit of phosphatidylinositol 3-kinase (PI3K), or its opposing phosphatase, phosphatase and tensin homolog (PTEN), had distinct effects on lens growth and homeostasis. The deletion of p110α reduced the levels of phosphorylated Akt and equatorial epithelial cell proliferation, and resulted in smaller transparent lenses in adult mice. The deletion of PTEN increased levels of phosphorylated Akt, altered lens sodium transport, and caused lens rupture and cataract. Here, we have generated conditional p110α/PTEN double-knockout mice, and evaluated epithelial cell proliferation and lens homeostasis. The double deletion of p110α and PTEN rescued the defect in lens size seen after the single knockout of p110α, but accelerated the lens rupture phenotype seen in PTEN single-knockout mice. Levels of phosphorylated Akt in double-knockout lenses were significantly higher than in wild-type lenses, but not as elevated as those reported for PTEN single-knockout lenses. These results showed that the double deletion of the p110α catalytic subunit of PI3K and its opposing phosphatase, PTEN, exacerbated the rupture defect seen in the single PTEN knockout and alleviated the growth defect observed in the single p110α knockout. Thus, the integrity of the PI3K signaling pathway was absolutely essential for proper lens homeostasis, but not for lens growth.
Collapse
|
15
|
Wang C, Zhao B, Fang J, Shi Z. IGF-1 Promotes Epithelial-Mesenchymal Transition of Lens Epithelial Cells That Is Conferred by miR-3666 Loss. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:5383146. [PMID: 35909588 PMCID: PMC9303496 DOI: 10.1155/2022/5383146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/13/2022] [Accepted: 06/21/2022] [Indexed: 11/27/2022]
Abstract
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-β2-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.
Collapse
Affiliation(s)
- Chao Wang
- The First Affiliated Hospital of Harbin Medical University, Anesthesiolgy Department, Harbin, China
| | - Baowen Zhao
- Hongqi Hospital Affiliated to Mudanjiang Medical College, Ophthalmology Department, Mudanjiang, China
| | - Jiahui Fang
- The First Affiliated Hospital of Harbin Medical University, Ophthalmology Department, Harbin, China
| | - Zhan Shi
- The First Affiliated Hospital of Harbin Medical University, Ophthalmology Department, Harbin, China
| |
Collapse
|
16
|
Guo M, Su F, Chen Y, Su B. Interfering Hsa_circRNA_0060640 Suppresses TGF-β2-Induced Proliferation, Motility and EMT in Human Lens Epithelium Cells by Targeting miR-214-3p and Collagen Type I alpha2 Chain. Curr Eye Res 2022; 47:735-746. [PMID: 35392747 DOI: 10.1080/02713683.2022.2053724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
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-β2). 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-β2-stimulated hLECs cells line SRA01/04. RNA interference of circ_0060640 could prevent SRA01/04 cells from TGF-β2-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-β2-stimulated SRA01/04 cells. CONCLUSION Circ_0060640 is a novel cataract-related gene and its silencing could block TGF-β2-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.
Collapse
Affiliation(s)
- Ming Guo
- Department of Ophthalmology, Jingzhou Hospital, Yangtze University (Jingzhou Central Hospital), Jingzhou, China
| | - Fanfan Su
- Department of Ophthalmology, Jingzhou Hospital, Yangtze University (Jingzhou Central Hospital), Jingzhou, China
| | - Yao Chen
- Department of Ophthalmology, Jingzhou Hospital, Yangtze University (Jingzhou Central Hospital), Jingzhou, China
| | - Bo Su
- Department of Pathology, School of Medicine, Yangtze University, Jingzhou, China
| |
Collapse
|
17
|
Paensuwan P, Ngoenkam J, Wangteeraprasert A, Pongcharoen S. Essential function of adaptor protein Nck1 in platelet-derived growth factor receptor signaling in human lens epithelial cells. Sci Rep 2022; 12:1063. [PMID: 35058548 PMCID: PMC8776929 DOI: 10.1038/s41598-022-05183-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 01/06/2022] [Indexed: 11/24/2022] Open
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.
Collapse
Affiliation(s)
- Pussadee Paensuwan
- Department of Optometry, Faculty of Allied Health Sciences, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand.
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand
| | - Apirath Wangteeraprasert
- Department of Medicine, Faculty of Medicine, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of Medicine, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand.
| |
Collapse
|
18
|
Wang C, Zhao R, Zhang S. lncRNA XIST knockdown suppresses cell proliferation and promotes apoptosis in diabetic cataracts through the miR‑34a/SMAD2 axis. Mol Med Rep 2021; 25:7. [PMID: 34751414 PMCID: PMC8600409 DOI: 10.3892/mmr.2021.12523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/22/2021] [Indexed: 11/06/2022] Open
Abstract
According to emerging evidence, long non-coding RNAs (lncRNAs) play critical roles in diabetes. The aim of the present study was to investigate the role and mechanism of X-inactive specific transcript (XIST) in cell proliferation, migration and apoptosis in diabetic cataracts (DC). SRA01/04 lens epithelial cells were treated with high glucose (HG). The levels of XIST, microRNA (miR)-34a and SMAD family member 2 (SMAD2) were examined via reverse transcription-quantitative PCR. MTT, Transwell, wound healing and TUNEL assays were performed to examine cell proliferation, invasion, migration and apoptosis, respectively. The interaction between miR-34a and XIST or SMAD2 was verified by luciferase reporter assay. It was found that the expression of XIST was increased and that of miR-34a was decreased in DC tissues and HG-treated SRA01/04 cells. XIST knockdown or miR-34a overexpression attenuated cell proliferation and migration, and induced apoptosis in HG-treated SRA01/04 cells. XIST targeted miR-34a and regulated DC progression through miR-34a. SMAD2 was identified as a target gene of miR-34a and was positively modulated by XIST. XIST knockdown inhibited cell proliferation and migration, and accelerated apoptosis in HG-stimulated SRA01/04 cells, and these effects were abrogated by SMAD2 overexpression. In conclusion, XIST promoted cell proliferation, migration and invasion, and inhibited apoptosis, through the miR-34a/SMAD2 axis in DC.
Collapse
Affiliation(s)
- Chao Wang
- Department of Ophthalmology, Shandong Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Ruiling Zhao
- Department of Ophthalmology, Shandong Tengzhou Central People's Hospital, Zaozhuang, Shandong, 277599, P.R. China
| | - Suhong Zhang
- Department of Ophthalmology, Shandong Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, P.R. China
| |
Collapse
|
19
|
Hidalgo-Alvarez V, Dhowre HS, Kingston OA, Sheridan CM, Levis HJ. Biofabrication of Artificial Stem Cell Niches in the Anterior Ocular Segment. Bioengineering (Basel) 2021; 8:135. [PMID: 34677208 PMCID: PMC8533470 DOI: 10.3390/bioengineering8100135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
The anterior segment of the eye is a complex set of structures that collectively act to maintain the integrity of the globe and direct light towards the posteriorly located retina. The eye is exposed to numerous physical and environmental insults such as infection, UV radiation, physical or chemical injuries. Loss of transparency to the cornea or lens (cataract) and dysfunctional regulation of intra ocular pressure (glaucoma) are leading causes of worldwide blindness. Whilst traditional therapeutic approaches can improve vision, their effect often fails to control the multiple pathological events that lead to long-term vision loss. Regenerative medicine approaches in the eye have already had success with ocular stem cell therapy and ex vivo production of cornea and conjunctival tissue for transplant recovering patients' vision. However, advancements are required to increase the efficacy of these as well as develop other ocular cell therapies. One of the most important challenges that determines the success of regenerative approaches is the preservation of the stem cell properties during expansion culture in vitro. To achieve this, the environment must provide the physical, chemical and biological factors that ensure the maintenance of their undifferentiated state, as well as their proliferative capacity. This is likely to be accomplished by replicating the natural stem cell niche in vitro. Due to the complex nature of the cell microenvironment, the creation of such artificial niches requires the use of bioengineering techniques which can replicate the physico-chemical properties and the dynamic cell-extracellular matrix interactions that maintain the stem cell phenotype. This review discusses the progress made in the replication of stem cell niches from the anterior ocular segment by using bioengineering approaches and their therapeutic implications.
Collapse
Affiliation(s)
- Veronica Hidalgo-Alvarez
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Hala S. Dhowre
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Olivia A. Kingston
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Carl M. Sheridan
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Hannah J. Levis
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| |
Collapse
|
20
|
Latta L, Figueiredo FC, Ashery-Padan R, Collinson JM, Daniels J, Ferrari S, Szentmáry N, Solá S, Shalom-Feuerstein R, Lako M, Xapelli S, Aberdam D, Lagali N. Pathophysiology of aniridia-associated keratopathy: Developmental aspects and unanswered questions. Ocul Surf 2021; 22:245-266. [PMID: 34520870 DOI: 10.1016/j.jtos.2021.09.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/19/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022]
Abstract
Aniridia, a rare congenital disease, is often characterized by a progressive, pronounced limbal insufficiency and ocular surface pathology termed aniridia-associated keratopathy (AAK). Due to the characteristics of AAK and its bilateral nature, clinical management is challenging and complicated by the multiple coexisting ocular and systemic morbidities in aniridia. Although it is primarily assumed that AAK originates from a congenital limbal stem cell deficiency, in recent years AAK and its pathogenesis has been questioned in the light of new evidence and a refined understanding of ocular development and the biology of limbal stem cells (LSCs) and their niche. Here, by consolidating and comparing the latest clinical and preclinical evidence, we discuss key unanswered questions regarding ocular developmental aspects crucial to AAK. We also highlight hypotheses on the potential role of LSCs and the ocular surface microenvironment in AAK. The insights thus gained lead to a greater appreciation for the role of developmental and cellular processes in the emergence of AAK. They also highlight areas for future research to enable a deeper understanding of aniridia, and thereby the potential to develop new treatments for this rare but blinding ocular surface disease.
Collapse
Affiliation(s)
- L Latta
- Dr. Rolf. M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Homburg, Saar, Germany; Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany.
| | - F C Figueiredo
- Department of Ophthalmology, Royal Victoria Infirmary, Newcastle Upon Tyne, United Kingdom
| | - R Ashery-Padan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - J M Collinson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - J Daniels
- Cells for Sight, UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - S Ferrari
- The Veneto Eye Bank Foundation, Venice, Italy
| | - N Szentmáry
- Dr. Rolf. M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - S Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - R Shalom-Feuerstein
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - M Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - S Xapelli
- Instituto Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - D Aberdam
- Centre de Recherche des Cordeliers, INSERM U1138, Team 17, France; Université de Paris, 75006, Paris, France.
| | - N Lagali
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Ophthalmology, Sørlandet Hospital Arendal, Arendal, Norway.
| |
Collapse
|
21
|
Wishart TFL, Flokis M, Shu DY, Das SJ, Lovicu FJ. Hallmarks of lens aging and cataractogenesis. Exp Eye Res 2021; 210:108709. [PMID: 34339681 DOI: 10.1016/j.exer.2021.108709] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022]
Abstract
Lens homeostasis and transparency are dependent on the function and intercellular communication of its epithelia. While the lens epithelium is uniquely equipped with functional repair systems to withstand reactive oxygen species (ROS)-mediated oxidative insult, ROS are not necessarily detrimental to lens cells. Lens aging, and the onset of pathogenesis leading to cataract share an underlying theme; a progressive breakdown of oxidative stress repair systems driving a pro-oxidant shift in the intracellular environment, with cumulative ROS-induced damage to lens cell biomolecules leading to cellular dysfunction and pathology. Here we provide an overview of our current understanding of the sources and essential functions of lens ROS, antioxidative defenses, and changes in the major regulatory systems that serve to maintain the finely tuned balance of oxidative signaling vs. oxidative stress in lens cells. Age-related breakdown of these redox homeostasis systems in the lens leads to the onset of cataractogenesis. We propose eight candidate hallmarks that represent common denominators of aging and cataractogenesis in the mammalian lens: oxidative stress, altered cell signaling, loss of proteostasis, mitochondrial dysfunction, dysregulated ion homeostasis, cell senescence, genomic instability and intrinsic apoptotic cell death.
Collapse
Affiliation(s)
| | - Mary Flokis
- School of Medical Sciences, The University of Sydney, NSW, Australia
| | - Daisy Y Shu
- School of Medical Sciences, The University of Sydney, NSW, Australia; Save Sight Institute, The University of Sydney, NSW, Australia; Schepens Eye Research Institute of Mass Eye and Ear. Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shannon J Das
- School of Medical Sciences, The University of Sydney, NSW, Australia
| | - Frank J Lovicu
- School of Medical Sciences, The University of Sydney, NSW, Australia; Save Sight Institute, The University of Sydney, NSW, Australia.
| |
Collapse
|
22
|
Gosak M, Gojić D, Spasovska E, Hawlina M, Andjelic S. Cataract Progression Associated with Modifications in Calcium Signaling in Human Lens Epithelia as Studied by Mechanical Stimulation. Life (Basel) 2021; 11:life11050369. [PMID: 33919270 PMCID: PMC8143283 DOI: 10.3390/life11050369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
Ca2+ homeostasis and signaling disturbances are associated with lens pathophysiology and are involved in cataract formation. Here, we explored the spatiotemporal changes in Ca2+ signaling in lens epithelial cells (LECs) upon local mechanical stimulation, to better understand the LECs’ intercellular communication and its association with cataractogenesis. We were interested in if the progression of the cataract affects the Ca2+ signaling and if modifications of the Ca2+ homeostasis in LECs are associated with different cataract types. Experiments were done on the human postoperative anterior lens capsule (LC) preparations consisting of the monolayer of LECs on the basement membrane. Our findings revealed that the Ca2+ signal spreads radially from the stimulation point and that the amplitude of Ca2+ transients decreases with increasing distance. It is noteworthy that a comparison of signaling characteristics with respect to the degree of cataract progression revealed that, in LCs from more developed cataracts, the Ca2+ wave propagates faster and the amplitudes of Ca2+ signals are lower, while their durations are longer. No differences were identified when comparing LCs with regard to the cataract type. Moreover, experiments with Apyrase have revealed that the Ca2+ signals are not affected by ATP-dependent paracrine communication. Our results indicated that cataract progression is associated with modifications in Ca2+ signaling in LECs, suggesting the functional importance of altered Ca2+ signaling of LECs in cataractogenesis.
Collapse
Affiliation(s)
- Marko Gosak
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia;
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
| | - Dajana Gojić
- Eye Hospital, University Medical Centre, 1000 Ljubljana, Slovenia; (D.G.); (E.S.); (M.H.)
| | - Elena Spasovska
- Eye Hospital, University Medical Centre, 1000 Ljubljana, Slovenia; (D.G.); (E.S.); (M.H.)
| | - Marko Hawlina
- Eye Hospital, University Medical Centre, 1000 Ljubljana, Slovenia; (D.G.); (E.S.); (M.H.)
| | - Sofija Andjelic
- Eye Hospital, University Medical Centre, 1000 Ljubljana, Slovenia; (D.G.); (E.S.); (M.H.)
- Correspondence:
| |
Collapse
|
23
|
Gao JL, Weaver JD, Tuo J, Wang LQ, Siwicki M, Despres D, Lizak M, Schneider EH, Kovacs W, Maminishkis A, Chen K, Yoshimura T, Ming Wang J, Chao Chan C, Murphy PM. Leukocyte chemotactic receptor Fpr1 protects against aging-related posterior subcapsular cataract formation. FASEB J 2021; 35:e21315. [PMID: 33538366 PMCID: PMC11005932 DOI: 10.1096/fj.202002135r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 11/11/2022]
Abstract
Cataracts are a common consequence of aging; however, pathogenesis remains poorly understood. Here, we observed that after 3 months of age mice lacking the G protein-coupled leukocyte chemotactic receptor Fpr1 (N-formyl peptide receptor 1) began to develop bilateral posterior subcapsular cataracts that progressed to lens rupture and severe degeneration, without evidence of either systemic or local ocular infection or inflammation. Consistent with this, Fpr1 was detected in both mouse and human lens in primary lens epithelial cells (LECs), the only cell type present in the lens; however, expression was confined to subcapsular LECs located along the anterior hemispheric surface. To maximize translucency, LECs at the equator proliferate and migrate posteriorly, then differentiate into lens fiber cells by nonclassical apoptotic signaling, which results in loss of nuclei and other organelles, including mitochondria which are a rich source of endogenous N-formyl peptides. In this regard, denucleation and posterior migration of LECs were abnormal in lenses from Fpr1-/- mice, and direct stimulation of LECs with the prototypic N-formyl peptide agonist fMLF promoted apoptosis. Thus, Fpr1 is repurposed beyond its immunoregulatory role in leukocytes to protect against cataract formation and lens degeneration during aging.
Collapse
Affiliation(s)
- Ji-Liang Gao
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Joseph D. Weaver
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jingsheng Tuo
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Long Q. Wang
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Marie Siwicki
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Daryl Despres
- Mouse Imaging Facility, National Institutes of Health, Bethesda, MD 20892
| | - Martin Lizak
- Mouse Imaging Facility, National Institutes of Health, Bethesda, MD 20892
| | - Erich H. Schneider
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - William Kovacs
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Arvydas Maminishkis
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Keqiang Chen
- Laboratory of Cancer and ImmunoMetabolism, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Okayama University, Okayama 700-8558, Japan
| | - Ji Ming Wang
- Laboratory of Cancer and ImmunoMetabolism, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702
| | - Chi Chao Chan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Philip M. Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
24
|
Liu X, Gong Q, Yang L, Liu M, Niu L, Wang L. microRNA-199a-5p regulates epithelial-to-mesenchymal transition in diabetic cataract by targeting SP1 gene. Mol Med 2020; 26:122. [PMID: 33276722 PMCID: PMC7718685 DOI: 10.1186/s10020-020-00250-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/26/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND As a common ocular complication of diabetes mellitus, diabetic cataract is becoming a leading cause of visual impairment. The progression of diabetic cataract progression involves epithelial-to-mesenchymal transition (EMT), the precise role of which remains to be investigated. As microRNAs (miRNAs) are suggested to be involved in the pathogenesis of many diseases, identification of aberrantly expressed miRNAs in diabetic lens epithelial cells (LECs) and their targets may provide insights into our understanding of diabetic cataract and potential therapeutic targets. METHODS Diabetic cataract capsules and LECs exposed to high glucose (25 mmol/L, 1-5 days) were used to mimic the model. Quantitative RT-PCR was performed to evaluate the differential expression of miRNA. Dual luciferase reporter assay was used to identify the binding target of miR-199a-5p. The expression of EMT-associated proteins was determined by immunofluorescence and Western blot analysis. RESULTS Our results showed the differential expression of miR-9, -16, -22, -199a and -204. MiR-199a was downregulated in diabetic cataract capsule and hyperglycemia-conditioned human LECs. Specific protein 1 could be directly targeted and regulated by miR-199a in LECs and inhibit EMT in diabetic LECs. CONCLUSION Our findings implied miR-199a could be a therapeutic target by regulating SP1 directly to affect EMT in diabetic cataract and provided novel insights into the pathogenesis of diabetic cataract.
Collapse
Affiliation(s)
- Xin Liu
- Eye Center, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China
| | - Qiaoyun Gong
- Eye Center, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiaotong University Medical School, #100 Haining Road, Shanghai, China
| | - Longfei Yang
- Jilin Provincial Key Laboratory On Molecular and Chemical Genetics, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China
| | - Min Liu
- Eye Center, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China
| | - Lingzhi Niu
- Eye Center, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China
| | - Lufei Wang
- Eye Center, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China.
| |
Collapse
|
25
|
Long-term myofibroblast persistence in the capsular bag contributes to the late spontaneous in-the-bag intraocular lens dislocation. Sci Rep 2020; 10:20532. [PMID: 33239706 PMCID: PMC7689492 DOI: 10.1038/s41598-020-77207-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Late spontaneous in-the-bag intraocular lens (IOL) dislocation is a complication presenting 6 months or later after cataract surgery. We aimed to characterize the cells in the lens capsules (LCs) of 18 patients with spontaneous late in-the-bag IOL dislocation. Patients' average age was 82.6 ± 1.5 years (range 72-98), and most of them had pseudoexfoliation syndrome (PEX). Cells from the LCs were positive for myofibroblast (αSMA), proliferation (Ki-67, PCNA), early lens development/lens progenitor (SOX2, PAX6), chemokine receptor (CXCR4), and transmembrane (N-cadherin) markers, while negative for epithelial (E-cadherin) marker. Moreover, the cells produced abundant fibronectin, type I and type V collagen in the nearby extracellular matrix (ECM). During ex vivo cultivation of dislocated IOL-LCs in toto, the cells proliferated and likely migrated onto the IOL's anterior side. EdU proliferation assay confirmed the proliferation potential of the myofibroblasts (MFBs) in dislocated IOL-LCs. Primary cultured lens epithelial cells/MFBs isolated from the LC of dislocated IOLs could induce collagen matrix contraction and continuously proliferated, migrated, and induced ECM remodeling. Taken together, this indicates that long-lived MFBs of dislocated IOLs might contribute to the pathogenic mechanisms in late in-the-bag IOL dislocation.
Collapse
|
26
|
LncRNA KCNQ1OT1 knockdown inhibits viability, migration and epithelial-mesenchymal transition in human lens epithelial cells via miR-26a-5p/ITGAV/TGF-beta/Smad3 axis. Exp Eye Res 2020; 200:108251. [PMID: 32950535 DOI: 10.1016/j.exer.2020.108251] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Long noncoding RNA potassium voltage-gated channel subfamily Q member 1 opposite strand/antisense transcript 1 (KCNQ1OT1) takes part in diabetic cataract progression. This research aims to analyze the function and mechanism of KCNQ1OT1 on viability, migration and epithelial-mesenchymal transition (EMT) in lens epithelial cells. METHODS 20 diabetic cataract posterior lens capsule tissues and normal samples were collected. Lens epithelial cells (SRA01/04) were stimulated via high glucose (HG). The levels of KCNQ1OT1, miR-26a-5p, integrin αV (ITGAV), TGF-β, Smad3 and phosphorylated (p)-Smad3 were measured via quantitative real-time polymerase chain reaction or Western blot. Cell viability, migration and EMT were analyzed via MTT, wound healing, transwell and Western blot assays. The target relationship between miR-26a-5p and KCNQ1OT1 or ITGAV was determined via luciferase reporter assay. RESULTS KCNQ1OT1 was up-regulated and miR-26a-5p level was reduced in diabetic cataract tissues and HG-treated SRA01/04 cells. Silence of KCNQ1OT1 or miR-26a-5p up-regulation repressed cell viability, migration and EMT in SRA01/04 cells stimulated via HG. KCNQ1OT1 could target miR-26a-5p and controlled cell viability, migration and EMT via regulating miR-26a-5p. ITGAV was targeted via miR-26a-5p and positively regulated via KCNQ1OT1. ITGAV overexpression promoted cell viability, migration and EMT in HG-treated SRA01/04 cells, which were mitigated by KCNQ1OT1 silence. KCNQ1OT1 knockdown mitigated HG-induced the activation of TGF-β/Smad3 signaling by regulating miR-26a-5p. CONCLUSION KCNQ1OT1 knockdown represses cell viability, migration and EMT through miR-26a-5p/ITGAV/TGF-β/Smad3 axis in SRA01/04 cells under HG condition, providing a new target for the treatment of diabetic cataract.
Collapse
|
27
|
Li J, Ye W, Xu W, Chang T, Zhang L, Ma J, Pei R, He M, Zhou J. Activation of autophagy inhibits epithelial to mesenchymal transition process of human lens epithelial cells induced by high glucose conditions. Cell Signal 2020; 75:109768. [PMID: 32896607 DOI: 10.1016/j.cellsig.2020.109768] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 01/11/2023]
Abstract
Subcapsular cataracts are common phenotype of diabetic cataracts, and abnormal lens epithelial cells (LECs) under the lens capsules have been considered to involve in the pathogenesis. Our previous studies have shown that the epithelial to mesenchymal transition (EMT), which is responsible for the LECs to lose their original polarity and tight junctions, occurs in a diabetic cataract mouse model. Autophagy is known to function in the EMT process in multiple tissues. However, the relationship between autophagy and EMT process in LECs has not yet been fully demonstrated. We found that high glucose retreatment reducing expression level of E-cadherin, an epithelial marker, but increasing that of α-smooth muscle actin (α-SMA), a mesenchymal marker, by Western blot and immunoflurence staining assays, and increased the cell migration by Transwell assay in human lens epithelial cell line HLE-B3. High glucose retreatment also led to impairment of autophagy, representing by downregulation of Beclin, LC3II/LC3I, and reducing the number of autophagosomes. Activation of autophagy by rapamycin could prevent high glucose-induced EMT. In addition, the levels of p62 and Snail were increased in high glucose-treated HLE-B3 cells, and their interactions were demonstrated by co-immunoprecipitation and immunoflurence staining, but all these changes were attenuated by application of rapamycin. These findings delineated a novel autophagy-mediated mechanism, p62 might mediate Snail underlying high glucose-induced EMT in LECs, suggesting a potential therapeutic approach for diabetic cataract by regulating autophagy.
Collapse
Affiliation(s)
- Ji Li
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China
| | - Wei Ye
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China
| | - Wenqin Xu
- The Orbital Disease Institute of the Third Medical Center of PLA General Hospital, 69 Yongding Road, Haidian District, 100039, Beijing, PR China
| | - Tianfang Chang
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China
| | - Luning Zhang
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China
| | - Jiyuan Ma
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China
| | - Rui Pei
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China
| | - Mengmei He
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China
| | - Jian Zhou
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, The Fourth Military Medical University, No.127 Changle West Road, Xi'an, 710032, Shaanxi, PR China.
| |
Collapse
|
28
|
Mei L, Yan H, Wang S, Guo C, Zheng X, Yan B, Zhao J, Yang A. Upregulation of miR-630 Induced by Oxidative Damage Resists Cell Migration Through Targeting ALCAM in Human Lens Epithelium Cells. Curr Eye Res 2019; 45:153-161. [PMID: 31869263 DOI: 10.1080/02713683.2019.1656748] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Purpose: To investigate the role of miRNAs in regulating oxidative damage during cataract formation.Methods: Microarray analysis and gene expression profiling assay were used to separately evaluate the miRNAs and mRNAs profiles in normal human lens epithelium cell line HLE-B3 treated by H2O2. The expression level of miR-630 was detected by RT-qPCR and the gene expression profiles were performed with gene ontology analysis using Bio Informatical database. The targets of miR-630 were predicted using miRecords and the results were used for screening targets of miR-630 combined with the GO analysis above. The mRNA levels of ALCAM, PCDH7, COL12A2, and EDIL3 in HLE-B3 cells after oxidative stimulation or miR-630 mimics transfection were measured by RT-qPCR, and the expression of ALCAM regulated by miR-630 was confirmed by Western blot and dual-luciferase reporter gene assay. The level of cell migration was measured by transwell assay and scratching test after transfection of miR-630 mimics and ALCAM siRNAs.Results: The microarray analysis demonstrated that miR-630 was significantly increased in HLE-B3 cells after oxidative stimulation. ALCAM, PCDH7, COL12A2, and EDIL3 were screened to be the possible targets of miR-630 by miRecords combined with GO analysis, but the results of RT-qPCR, Western blot and dual-luciferase reporter gene assay showed that only the expression of ALCAM was repressed by miR-630 transfection. Cell migration was inhibited through transfection of miR-630 mimics or ALCAM siRNAs and the upregulation of miR-630 partly reduced the cell migration increased by oxidative stimulation.Conclusion: miR-630 is one of the miRNAs increased by oxidative stimulation in human lens epithelium cells. Its upregulation may inhibit cell migration by targeting on ALCAM, which is important for HLECs to resist behavioral changes induced by oxidative damage and may delay the progression of cataract.
Collapse
Affiliation(s)
- Lin Mei
- Department of Ophthalmology, Affiliated Guangren Hospital School of Medicine, Xi'an Jiaotong University, Xi'an No. 4 Hospital, Shaanxi Eye Hospital, Xi'an, Shaanxi Province, China.,Department of Ophthalmology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Hong Yan
- Department of Ophthalmology, Affiliated Guangren Hospital School of Medicine, Xi'an Jiaotong University, Xi'an No. 4 Hospital, Shaanxi Eye Hospital, Xi'an, Shaanxi Province, China.,Department of Ophthalmology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Song Wang
- Department of Ophthalmology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Chenjun Guo
- Department of Ophthalmology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiaoliang Zheng
- Department of Ophthalmology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Bo Yan
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jing Zhao
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Angang Yang
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
29
|
Taler K, Weiss O, Rotem-Bamberger S, Rubinstein AM, Seritrakul P, Gross JM, Inbal A. Lysyl hydroxylase 3 is required for normal lens capsule formation and maintenance of lens epithelium integrity and fate. Dev Biol 2019; 458:177-188. [PMID: 31669351 DOI: 10.1016/j.ydbio.2019.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/03/2019] [Accepted: 10/20/2019] [Indexed: 02/06/2023]
Abstract
Lens abnormalities are a major cause of reduced vision and blindness. One mechanism that can lead to reduced lens transparency, i.e. cataract, is abnormal behavior of lens epithelial cells (LECs), the precursors of the transparent lens fiber cells. Here we describe a zebrafish mutation causing the embryonic lens epithelium to generate cellular masses comprising partially differentiated lens fiber cells. We identify the mutant gene as plod3, which encodes for Lysyl hydroxylase 3 (Lh3), an enzyme essential for modification of collagens, including Collagen IV, a main component of the lens capsule. We show that plod3-deficient lenses have abnormal lens epithelium from an early developmental stage, as well as abnormal lens capsules. Subsequently, upregulation of TGFβ signaling takes place, which drives the formation of lens epithelial cellular masses. We identify a similar phenotype in Collagen IVα5-deficient embryos, suggesting a key role for the defective lens capsule in the pathogenesis. We propose that plod3 and col4a5 mutant zebrafish can serve as useful models for better understanding the biology of LECs during embryonic development and in formation of lens epithelium-derived cataract.
Collapse
Affiliation(s)
- Kineret Taler
- Department of Medical Neurobiology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Omri Weiss
- Department of Medical Neurobiology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shahar Rotem-Bamberger
- Department of Medical Neurobiology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ariel M Rubinstein
- Department of Medical Neurobiology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Pawat Seritrakul
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey M Gross
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Adi Inbal
- Department of Medical Neurobiology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
30
|
Srinivasarao DA, Reddy SS, Reddy GB, Katti DS. Spatio-temporal control on the delivery of triamcinolone acetonide using polymeric nanoparticles reduces steroid induced cataract. Int J Pharm 2019; 568:118474. [DOI: 10.1016/j.ijpharm.2019.118474] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 01/13/2023]
|
31
|
Turan G, Turan M. The Evaluation of TUNEL, PCNA and SOX2 Expressions in Lens Epithelial Cells of Cataract Patients with Pseudoexfoliation Syndrome. Curr Eye Res 2019; 45:12-16. [DOI: 10.1080/02713683.2019.1657463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Gulay Turan
- Faculty of Medicine, Department of Pathology, Balikesir University, Balikesir, Turkey
| | - Meydan Turan
- Department of Ophthalmology, Balikesir Ataturk City Hospital, Balikesir, Turkey
| |
Collapse
|
32
|
Liu W, Yang Y, Yan J, Wang L. MicroRNA-23b-3p promotes the proliferation, migration, and epithelial-mesenchymal transition of lens epithelial cells by targeting Sprouty2. Acta Histochem 2019; 121:704-711. [PMID: 31235073 DOI: 10.1016/j.acthis.2019.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/18/2019] [Accepted: 05/20/2019] [Indexed: 12/30/2022]
Abstract
Cataract, opacification of the lens, is one of the most important reasons of visual impairment and blindness. Though microRNAs (miRNAs) have been demonstrated to play important roles in cataractogenesis, the underlying molecular mechanisms in this progress remain obscure. In the present study, microRNA-23b-3p (miR-23b) overexpression promoted the proliferation, migration and epithelial-mesenchymal transition (EMT), whereas miR-23b knockdown markedly inhibited the proliferation, migration and TGF-β-induced EMT of lens epithelial cells (LECs). In TGF-β-induced LECs, the expression of miR-23b was markedly upregulated and the expression of Sprouty2 (SPRY2) was markedly downregulated, furthermore the mRNA and protein levels of SPRY2 were markedly decreased in miR-23b inhibitor-transfected LECs. We then performed a Dual-luciferase reporter assay to confirm that miR-23b directly targeted SPRY2. The promoted migration and EMT of LECs by enforced expression of miR-23b were suppressed by SPRY2 overexpression. The findings present the first evidence indicating that miR-23b can promote the proliferation, migration, and EMT of LECs by targeting SPRY2 and the inhibition of miR-23b may possess the therapeutic potential for cataract.
Collapse
|
33
|
Wernecke L, Keckeis S, Reichhart N, Strauß O, Salchow DJ. Epithelial-Mesenchymal Transdifferentiation in Pediatric Lens Epithelial Cells. Invest Ophthalmol Vis Sci 2019; 59:5785-5794. [PMID: 30521667 DOI: 10.1167/iovs.18-23789] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Posterior capsule opacification (PCO) is a complication after cataract surgery, particularly in children. Epithelial-mesenchymal transition (EMT) of lens epithelial cells, mediated by transforming growth factor beta (TGFβ), contributes to PCO. However, its pathogenesis in children is poorly understood. We correlated cell growth in culture with patient characteristics, studied gene expression of pediatric lens epithelial cells (pLEC), and examined the effects of TGFβ-2 on these cells in vitro. Methods Clinical characteristics of children with cataracts correlated with growth behavior of pLEC in vitro. mRNA expression of epithelial (αB-crystallin, connexin-43) and mesenchymal (αV-integrin, α-smooth muscle actin, collagen-Iα2, fibronectin-1) markers was quantified in pLEC and in cell line HLE-B3 in the presence and absence of TGFβ-2. Results Fifty-four anterior lens capsules from 40 children aged 1 to 180 months were obtained. Cell outgrowth occurred in 44% of the capsules from patients ≤ 12 months and in 33% of capsules from children aged 13 to 60 months, but in only 6% of capsules from children over 60 months. TGFβ-2 significantly upregulated expression of αB-crystallin (HLE-B3), αV-integrin (HLE-B3), collagen-Iα2, and fibronectin-1 (in pLEC and HLE-B3 cells). Conclusions Patient characteristics correlated with growth behavior of pLEC in vitro, paralleling a higher clinical incidence of PCO in younger children. Gene expression profiles of pLEC and HLE-B3 suggest that upregulation of αV-integrin, collagen-Iα2, and fibronectin-1 are involved in EMT.
Collapse
Affiliation(s)
- Laura Wernecke
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susanne Keckeis
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nadine Reichhart
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Olaf Strauß
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Daniel J Salchow
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
34
|
Armao D, Bouldin TW, Bailey RM, Hooper JE, Bharucha DX, Gray SJ. Advancing the pathologic phenotype of giant axonal neuropathy: early involvement of the ocular lens. Orphanet J Rare Dis 2019; 14:27. [PMID: 30709364 PMCID: PMC6359799 DOI: 10.1186/s13023-018-0957-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/18/2018] [Indexed: 12/12/2022] Open
Abstract
Giant axonal neuropathy (GAN; ORPHA: 643; OMIM# 256850) is a rare, hereditary, pediatric neurodegenerative disorder associated with intracellular accumulations of intermediate filaments (IFs). GAN knockout (KO) mouse models mirror the IF dysregulation and widespread nervous system pathology seen in human GAN. Validation of therapeutic efficacy and viral vector delivery systems with these GAN KO models has provided the springboard for the development of a viral vector being delivered intrathecally in an ongoing Phase I gene therapy clinical trial for the treatment of children with GAN (https://clinicaltrials.gov/ct2/show/NCT02362438). During the course of a comprehensive pathologic characterization of the GAN KO mouse, we discovered the very early and unexpected involvement of the ocular lens. Light microscopy revealed the presence of intracytoplasmic inclusion bodies within lens epithelial cells. The inclusion bodies showed strong immunohistochemical positivity for glial fibrillary acidic protein (GFAP). We confirmed that intracytoplasmic inclusion bodies are also present within lens epithelial cells in human GAN. These IF inclusion bodies in lens epithelial cells are unique to GAN. Similar IF inclusion bodies in lens epithelial cells have not been reported previously in experimental animal models or human diseases. Since current paradigms in drug discovery and drug repurposing for IF-associated disorders are often hindered by lack of validated targets, our findings suggest that lens epithelial cells in the GAN KO mouse may provide a potential target, in vivo and in vitro, for evaluating drug efficacy and alternative therapeutic approaches in promoting the clearance of IF inclusions in GAN and other diseases characterized by intracellular IF accumulations.
Collapse
Affiliation(s)
- Diane Armao
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Department of Radiology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Thomas W Bouldin
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Rachel M Bailey
- Gene Therapy Center, University of North Carolina at Chapel Hill Chapel Hill, Chapel Hill, NC, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jody E Hooper
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Diana X Bharucha
- Department of Neurology and Pediatrics, Children's National Health System, Washington, DC, USA.,National Institutes of Health NINDS/ Neurogenetics Branch, Bethesda, MD, USA
| | - Steven J Gray
- Gene Therapy Center, University of North Carolina at Chapel Hill Chapel Hill, Chapel Hill, NC, USA. .,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, NC, USA. .,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
35
|
Zhao Y, Zheng D, Cvekl A. A comprehensive spatial-temporal transcriptomic analysis of differentiating nascent mouse lens epithelial and fiber cells. Exp Eye Res 2018; 175:56-72. [PMID: 29883638 PMCID: PMC6167154 DOI: 10.1016/j.exer.2018.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/01/2018] [Accepted: 06/03/2018] [Indexed: 02/07/2023]
Abstract
Elucidation of both the molecular composition and organization of the ocular lens is a prerequisite to understand its development, function, pathology, regenerative capacity, as well as to model lens development and disease using in vitro differentiation of pluripotent stem cells. Lens is comprised of the anterior lens epithelium and posterior lens fibers, which form the bulk of the lens. Lens fibers differentiate from lens epithelial cells through cell cycle exit-coupled differentiation that includes cellular elongation, accumulation of crystallins, cytoskeleton and membrane remodeling, and degradation of organelles within the central region of the lens. Here, we profiled spatiotemporal expression dynamics of both mRNAs and non-coding RNAs from microdissected mouse nascent lens epithelium and lens fibers at four developmental time points (embryonic [E] day 14.5, E16.5, E18.5, and P0.5) by RNA-seq. During this critical time window, multiple complex biosynthetic and catabolic processes generate the molecular and structural foundation for lens transparency. Throughout this developmental window, 3544 and 3518 genes show consistently and significantly greater expression in the nascent lens epithelium and fibers, respectively. Comprehensive data analysis confirmed major roles of FGF-MAPK, Wnt/β-catenin, PI3K/AKT, TGF-β, and BMP signaling pathways and revealed significant novel contributions of mTOR, EIF2, EIF4, and p70S6K signaling in lens formation. Unbiased motif analysis within promoter regions of these genes with consistent expression changes between epithelium and fiber cells revealed an enrichment for both established (e.g. E2Fs, Etv5, Hsf4, c-Maf, MafG, MafK, N-Myc, and Pax6) transcription factors and a number of novel regulators of lens formation, such as Arntl2, Dmrta2, Stat5a, Stat5b, and Tulp3. In conclusion, the present RNA-seq data serves as a comprehensive reference resource for deciphering molecular principles of normal mammalian lens differentiation, mapping a full spectrum of signaling pathways and DNA-binding transcription factors operating in both lens compartments, and predicting novel pathways required to establish lens transparency.
Collapse
Affiliation(s)
- Yilin Zhao
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Ales Cvekl
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
36
|
Dalke C, Neff F, Bains SK, Bright S, Lord D, Reitmeir P, Rößler U, Samaga D, Unger K, Braselmann H, Wagner F, Greiter M, Gomolka M, Hornhardt S, Kunze S, Kempf SJ, Garrett L, Hölter SM, Wurst W, Rosemann M, Azimzadeh O, Tapio S, Aubele M, Theis F, Hoeschen C, Slijepcevic P, Kadhim M, Atkinson M, Zitzelsberger H, Kulka U, Graw J. Lifetime study in mice after acute low-dose ionizing radiation: a multifactorial study with special focus on cataract risk. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2018; 57:99-113. [PMID: 29327260 PMCID: PMC5902533 DOI: 10.1007/s00411-017-0728-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/21/2017] [Indexed: 05/28/2023]
Abstract
Because of the increasing application of ionizing radiation in medicine, quantitative data on effects of low-dose radiation are needed to optimize radiation protection, particularly with respect to cataract development. Using mice as mammalian animal model, we applied a single dose of 0, 0.063, 0.125 and 0.5 Gy at 10 weeks of age, determined lens opacities for up to 2 years and compared it with overall survival, cytogenetic alterations and cancer development. The highest dose was significantly associated with increased body weight and reduced survival rate. Chromosomal aberrations in bone marrow cells showed a dose-dependent increase 12 months after irradiation. Pathological screening indicated a dose-dependent risk for several types of tumors. Scheimpflug imaging of the lens revealed a significant dose-dependent effect of 1% of lens opacity. Comparison of different biological end points demonstrated long-term effects of low-dose irradiation for several biological end points.
Collapse
Affiliation(s)
- Claudia Dalke
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| | - Frauke Neff
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Pathology, Neuherberg, Germany
- Present Address: Municipal Clinical Center Munich, Munich, Germany
| | - Savneet Kaur Bains
- Department of Life Sciences, Brunel University London, Uxbridge, UK
- Present Address: Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Scott Bright
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Present Address: University of Texas, MD Anderson, Houston, TX USA
| | - Deborah Lord
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Peter Reitmeir
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Health Economics and Health Care Management, Neuherberg, Germany
| | - Ute Rößler
- Department Radiation Protection and Health, Federal Office for Radiation Protection, Oberschleissheim, Germany
| | - Daniel Samaga
- Department Radiation Protection and Health, Federal Office for Radiation Protection, Oberschleissheim, Germany
- Present Address: Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit of Radiation Cytogenetics, Neuherberg, Germany
| | - Kristian Unger
- Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit of Radiation Cytogenetics, Neuherberg, Germany
| | - Herbert Braselmann
- Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit of Radiation Cytogenetics, Neuherberg, Germany
| | - Florian Wagner
- Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit Medical Radiation Physics and Diagnostics, Neuherberg, Germany
- Present Address: Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Radiation Protection, Neuherberg, Germany
| | - Matthias Greiter
- Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit Medical Radiation Physics and Diagnostics, Neuherberg, Germany
- Present Address: Helmholtz Center Munich, German Research Center for Environmental Health, Individual Monitoring Service, Neuherberg, Germany
| | - Maria Gomolka
- Department Radiation Protection and Health, Federal Office for Radiation Protection, Oberschleissheim, Germany
| | - Sabine Hornhardt
- Department Radiation Protection and Health, Federal Office for Radiation Protection, Oberschleissheim, Germany
| | - Sarah Kunze
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| | - Stefan J. Kempf
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Radiation Biology, Neuherberg, Germany
- Present Address: Department of Bioanalytical Sciences, CSL Behring GmbH, Marburg, Germany
| | - Lillian Garrett
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| | - Sabine M. Hölter
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| | - Wolfgang Wurst
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| | - Michael Rosemann
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Radiation Biology, Neuherberg, Germany
| | - Omid Azimzadeh
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Radiation Biology, Neuherberg, Germany
| | - Soile Tapio
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Radiation Biology, Neuherberg, Germany
| | - Michaela Aubele
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Pathology, Neuherberg, Germany
| | - Fabian Theis
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
| | - Christoph Hoeschen
- Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit Medical Radiation Physics and Diagnostics, Neuherberg, Germany
- Present Address: Chair of Medical Systems Technology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | - Munira Kadhim
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Michael Atkinson
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Radiation Biology, Neuherberg, Germany
| | - Horst Zitzelsberger
- Helmholtz Center Munich, German Research Center for Environmental Health, Research Unit of Radiation Cytogenetics, Neuherberg, Germany
| | - Ulrike Kulka
- Department Radiation Protection and Health, Federal Office for Radiation Protection, Oberschleissheim, Germany
| | - Jochen Graw
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| |
Collapse
|
37
|
Yan Y, Qian H, Jiang H, Yu H, Sun L, Wei X, Sun Y, Ge H, Zhou H, Li X, Hashimoto T, Tang X, Liu P. Laminins in an in vitro anterior lens capsule model established using HLE B-3 cells. Mol Med Rep 2018; 17:5726-5733. [PMID: 29436687 PMCID: PMC5866015 DOI: 10.3892/mmr.2018.8581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/11/2018] [Indexed: 12/23/2022] Open
Abstract
Cataracts are the most common eye disease to cause blindness in patients. The abnormal deposition of laminins (LMs) in the lens capsule and the disruption of capsular epithelium contribute to cataract development, although the mechanism by which this occurs is currently unclear. The present study aimed to reproduce HLE B-3 basement membranes (BMs) using HLE B-3 cells and to analyze the similarities of LM expression between HLE B-3 BMs and human anterior lens capsule (ALC). Immunohistochemistry (IHC), ELISA, western blot analysis and immunoprecipitation (IP)-western blot analysis were used to detect total LMs, LM trimers and 11 LM subunits in HLE B-3 cells, HLE B-3 BMs and human ALCs. In IHC staining, HLE B-3 cells and human ALCs were positive for LMs. In LM ELISA, all samples analyzed were positive for LMs. Western blot analysis detected all LM subunits except for LMγ3 in HLE B-3 cell lysate, 4 subunits (LMα4, LMα2, LMα1 and LMγ1) in HLE B-3 cell culture supernatant, 5 subunits (LMα4, LMα2, LMα1, LMβ3 and LMγ1) in HLE B-3 BMs, and 3 subunits (LMα4, LMγ2 and LMγ1) in human ALCs. The results of IP-western blot analysis revealed that the LM411 trimer was detected in HLE B-3 cell culture supernatant. These results indicated that HLE B-3 BMs were similar to human ALCs in terms of LM expression. Therefore, HLE B-3 BMs could be used as an in vitro ALC model to determine the role of LMs in ALC in the pathogenesis of cataracts and to select potential anti-cataract drugs.
Collapse
Affiliation(s)
- Yu Yan
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hua Qian
- Academician Workstation, Harbin Medical University and Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150081, P.R. China
| | - Hongda Jiang
- Department of Laboratory Diagnosis, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Haiyang Yu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Liyao Sun
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xi Wei
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yunduan Sun
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hongyan Ge
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Haizhou Zhou
- Department of Laboratory Diagnosis, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiaoguang Li
- Academician Workstation, Harbin Medical University and Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150081, P.R. China
| | - Takashi Hashimoto
- Department of Dermatology, Osaka City University Graduate School of Medicine, Osaka 545‑8585, Japan
| | - Xianling Tang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ping Liu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
38
|
Oltulu P, Oltulu R. The Association of Cataract and Lens Epithelial Cell Apoptosis in Patients with Pseudoexfoliation Syndrome. Curr Eye Res 2017; 43:300-303. [PMID: 29182401 DOI: 10.1080/02713683.2017.1406524] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE The aim of this study was to measure the density of apoptotic lens epithelial cells (LECs) and to determine its association with cataract formation in eyes with pseudoexfoliation syndrome (PEX). MATERIALS AND METHODS Twenty-one eyes of 21 patients diagnosed with PEX without glaucoma (Group 1) and 22 eyes of 22 subjects without PEX (Group 2) were enrolled in this study. During cataract surgery, anterior capsule samples were obtained by the 5.5 mm continuous curvilinear capsulorhexis method. Apoptosis was determined with a TUNEL kit (Boster, Wuhan, China) according to the manufacturer's protocol. Four fields in each pathology preparation were chosen randomly under a microscope, and 100 cells were counted in each field to calculate the apoptosis rates. RESULTS Forty-three eyes of 43 subjects were enrolled in this study. There were no significant differences in age or sex between the two studied groups (p > 0.05). Under a microscope, the LECs were stained light blue and their nuclei were oval shaped. Positive stained cells were found occasionally in Group 2, while a significant amount of black-brown positively stained LECs with condensed nuclei was found in Group 1. The apoptosis rates were 35.2 ± 2.1% and 14.1 ± 1% in Groups 1 and 2, respectively. The proportion of positively stained LECs was higher in Group 1 (p < 0.001). CONCLUSION Our study suggests that apoptosis in LECs is the pathophysiological mechanism for the higher rate of cataracts in PEX patients, in addition to the ocular ischemia hypothesis.
Collapse
Affiliation(s)
- Pembe Oltulu
- a Department of Pathology , Necmettin Erbakan University Meram Faculty of Medicine , Konya , Turkey
| | - Refik Oltulu
- b Department of Ophthalmology , Necmettin Erbakan University Meram Faculty of Medicine , Konya , Turkey
| |
Collapse
|
39
|
Qin Y, Zhu Y, Luo F, Chen C, Chen X, Wu M. Killing two birds with one stone: dual blockade of integrin and FGF signaling through targeting syndecan-4 in postoperative capsular opacification. Cell Death Dis 2017; 8:e2920. [PMID: 28703800 PMCID: PMC5550862 DOI: 10.1038/cddis.2017.315] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 12/12/2022]
Abstract
The most common complication after cataract surgery is postoperative capsular opacification, which includes anterior capsular opacification (ACO) and posterior capsular opacification (PCO). Increased adhesion of lens epithelial cells (LECs) to the intraocular lens material surface promotes ACO formation, whereas proliferation and migration of LECs to the posterior capsule lead to the development of PCO. Cell adhesion is mainly mediated by the binding of integrin to extracellular matrix proteins, while cell proliferation and migration are regulated by fibroblast growth factor (FGF). Syndecan-4 (SDC-4) is a co-receptor for both integrin and FGF signaling pathways. Therefore, SDC-4 may be an ideal therapeutic target for the prevention and treatment of postoperative capsular opacification. However, how SDC-4 contributes to FGF-mediated proliferation, migration, and integrin-mediated adhesion of LECs is unclear. Here, we found that downregulation of SDC-4 inhibited FGF signaling through the blockade of ERK1/2 and PI3K/Akt/mTOR activation, thus suppressing cell proliferation and migration. In addition, downregulation of SDC-4 suppressed integrin-mediated cell adhesion through inhibiting focal adhesion kinase (FAK) phosphorylation. Moreover, SDC-4 knockout mice exhibited normal lens morphology, but had significantly reduced capsular opacification after injury. Finally, SDC-4 expression level was increased in the anterior capsule LECs of age-related cataract patients. Taken together, we for the first time characterized the key regulatory role of SDC-4 in FGF and integrin signaling in human LECs, and provided the basis for future pharmacological interventions of capsular opacification.
Collapse
Affiliation(s)
- Yingyan Qin
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yi Zhu
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Furong Luo
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Chuan Chen
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xiaoyun Chen
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Mingxing Wu
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
40
|
Sellitto C, Li L, Vaghefi E, Donaldson PJ, Lin RZ, White TW. The Phosphoinosotide 3-Kinase Catalytic Subunit p110α is Required for Normal Lens Growth. Invest Ophthalmol Vis Sci 2017; 57:3145-51. [PMID: 27304846 PMCID: PMC4928694 DOI: 10.1167/iovs.16-19607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Signal transduction pathways influence lens growth, but little is known about the role(s) of the class 1A phosphoinositide 3-kinases (PI3Ks). To further investigate how signaling regulates lens growth, we generated and characterized mice in which the p110α and p110β catalytic subunits of PI3K were conditionally deleted in the mouse lens. Methods Floxed alleles of the catalytic subunits of PI3K were conditionally deleted in the lens by using MLR10-cre transgenic mice. Lenses of age-matched animals were dissected and photographed. Postnatal lenses were fixed, paraffin embedded, sectioned, and stained with hematoxylin-eosin. Cell proliferation was quantified by labeling S-phase cells in intact lenses with 5-ethynyl-2′-deoxyuridine. Protein kinase B (AKT) activation was examined by Western blotting. Results Lens-specific deletion of p110α resulted in a significant reduction of eye and lens size, without compromising lens clarity. Conditional knockout of p110β had no effect on lens size or clarity, and deletion of both the p110α and p110β subunits resulted in a phenotype that resembled the p110α single-knockout phenotype. Levels of activated AKT were decreased more in p110α- than in p110β-deficient lenses. A significant reduction in proliferating cells in the germinative zone was observed on postnatal day 0 in p110α knockout mice, which was temporally correlated with decreased lens volume. Conclusions These data suggest that the class 1A PI3K signaling pathway plays an important role in the regulation of lens size by influencing the extent and spatial location of cell proliferation in the perinatal period.
Collapse
Affiliation(s)
- Caterina Sellitto
- Department of Physiology and Biophysics Stony Brook University, Stony Brook, New York, United States
| | - Leping Li
- Department of Physiology and Biophysics Stony Brook University, Stony Brook, New York, United States
| | - Ehsan Vaghefi
- School of Optometry and Vision Science, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Paul J Donaldson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Z Lin
- Department of Physiology and Biophysics Stony Brook University, Stony Brook, New York, United States 4Medical Service, Department of Veterans Affairs Medical Center, Northport, New York, United States
| | - Thomas W White
- Department of Physiology and Biophysics Stony Brook University, Stony Brook, New York, United States
| |
Collapse
|
41
|
Abstract
Objective: This paper aimed to review the current literature on the surface modification of intraocular lenses (IOLs). Data Sources: All articles about surface modification of IOLs published up to 2015 were identified through a literature search on both PubMed and ScienceDirect. Study Selection: The articles on the surface modification of IOLs were included, but those on design modification and surface coating were excluded. Results: Technology of surface modification included plasma, ion beam, layer-by-layer self-assembly, ultraviolet radiation, and ozone. The main molecules introduced into IOLs surface were poly (ethylene glycol), polyhedral oligomeric silsesquioxane, 2-methacryloyloxyethyl phosphorylcholine, TiO2, heparin, F-heparin, titanium, titanium nitride, vinyl pyrrolidone, and inhibitors of cytokines. The surface modification either resulted in a more hydrophobic lens, a more hydrophilic lens, or a lens with a hydrophilic anterior and hydrophobic posterior surface. Advances in research regarding surface modification of IOLs had led to a better biocompatibility in both in vitro and animal experiments. Conclusion: The surface modification is an efficient, convenient, economic and promising method to improve the biocompatibility of IOLs.
Collapse
Affiliation(s)
| | | | | | - Gui-Qin Wang
- Department of Ophthalmology, Navy General Hospital of PLA, Beijing 100048; Department of Ophthalmilogy, Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
42
|
Bao X, Hou M, Qin Y, Luo F, Shang F, Wu M. Effect of an MG132-Sustained Drug Delivery Capsular Ring on the Inhibition of Posterior Capsule Opacification in a Rabbit Model. J Ocul Pharmacol Ther 2017; 33:103-110. [PMID: 28106491 DOI: 10.1089/jop.2016.0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To design an MG132-sustained drug delivery capsular ring (SDDCR) and investigate its effect on the inhibition of posterior capsule opacification (PCO) in a rabbit model. METHODS The SDDCRs were prepared by forming a slice of film made by the mixture of poly lactic-co-glycolic acid (PLGA) and MG132 on the surface of capsular tension rings (CTRs). The drug-loading capacity, entrapment efficiency, and in vitro release of the drug-containing film were detected. Eighteen New Zealand white rabbits were operated with phacoemulsification and MG132-SDDCRs/PLGA-CTRs/CTRs implantation in the single eye. The images of the anterior segments were acquired at certain days, and the epithelial-mesenchymal transition (EMT) markers were detected by western blot and immunofluorescence. RESULTS The drug-loading capacity and entrapment efficiency of MG132-SDDCRs were 1.15% ± 0.04% and 66.16% ± 0.027%, respectively, and the drug released well within a month. The PCO degree of the MG132-SDDCR group was significantly lower than the other groups. The expression of alpha-smooth muscle actin, fibronectin, vimentin, and collagen-I was lower, and the expression of E-cadherin (E-cad) was higher in the MG132-SDDCR group than the other groups. CONCLUSIONS MG132-SDDCRs could be established successfully. The PCO process was prevented, and the expression of EMT markers was inhibited by the implantation of MG132-SDDCRs, indicating that this could be a potential treatment against PCO.
Collapse
Affiliation(s)
- Xuan Bao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| | - Min Hou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| | - Yingyan Qin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| | - Furong Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| | - Fu Shang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| | - Mingxing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| |
Collapse
|
43
|
Pfleger CM. The Hippo Pathway: A Master Regulatory Network Important in Development and Dysregulated in Disease. Curr Top Dev Biol 2017; 123:181-228. [PMID: 28236967 DOI: 10.1016/bs.ctdb.2016.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Hippo Pathway is a master regulatory network that regulates proliferation, cell growth, stemness, differentiation, and cell death. Coordination of these processes by the Hippo Pathway throughout development and in mature organisms in response to diverse external and internal cues plays a role in morphogenesis, in controlling organ size, and in maintaining organ homeostasis. Given the importance of these processes, the Hippo Pathway also plays an important role in organismal health and has been implicated in a variety of diseases including eye disease, cardiovascular disease, neurodegeneration, and cancer. This review will focus on Drosophila reports that identified the core components of the Hippo Pathway revealing specific downstream biological outputs of this complicated network. A brief description of mammalian reports will complement review of the Drosophila studies. This review will also survey upstream regulation of the core components with a focus on feedback mechanisms.
Collapse
Affiliation(s)
- Cathie M Pfleger
- The Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
44
|
Che D, Zhou T, Lan Y, Xie J, Gong H, Li C, Feng J, Hong H, Qi W, Ma C, Wu Q, Yang X, Gao G. High glucose-induced epithelial-mesenchymal transition contributes to the upregulation of fibrogenic factors in retinal pigment epithelial cells. Int J Mol Med 2016; 38:1815-1822. [DOI: 10.3892/ijmm.2016.2768] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/29/2016] [Indexed: 11/06/2022] Open
|
45
|
Ainsbury EA, Barnard S, Bright S, Dalke C, Jarrin M, Kunze S, Tanner R, Dynlacht JR, Quinlan RA, Graw J, Kadhim M, Hamada N. Ionizing radiation induced cataracts: Recent biological and mechanistic developments and perspectives for future research. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 770:238-261. [DOI: 10.1016/j.mrrev.2016.07.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 02/06/2023]
|
46
|
He S, Limi S, McGreal RS, Xie Q, Brennan LA, Kantorow WL, Kokavec J, Majumdar R, Hou H, Edelmann W, Liu W, Ashery-Padan R, Zavadil J, Kantorow M, Skoultchi AI, Stopka T, Cvekl A. Chromatin remodeling enzyme Snf2h regulates embryonic lens differentiation and denucleation. Development 2016; 143:1937-47. [PMID: 27246713 PMCID: PMC4920164 DOI: 10.1242/dev.135285] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/21/2016] [Indexed: 12/30/2022]
Abstract
Ocular lens morphogenesis is a model for investigating mechanisms of cellular differentiation, spatial and temporal gene expression control, and chromatin regulation. Brg1 (Smarca4) and Snf2h (Smarca5) are catalytic subunits of distinct ATP-dependent chromatin remodeling complexes implicated in transcriptional regulation. Previous studies have shown that Brg1 regulates both lens fiber cell differentiation and organized degradation of their nuclei (denucleation). Here, we employed a conditional Snf2h(flox) mouse model to probe the cellular and molecular mechanisms of lens formation. Depletion of Snf2h induces premature and expanded differentiation of lens precursor cells forming the lens vesicle, implicating Snf2h as a key regulator of lens vesicle polarity through spatial control of Prox1, Jag1, p27(Kip1) (Cdkn1b) and p57(Kip2) (Cdkn1c) gene expression. The abnormal Snf2h(-/-) fiber cells also retain their nuclei. RNA profiling of Snf2h(-/) (-) and Brg1(-/-) eyes revealed differences in multiple transcripts, including prominent downregulation of those encoding Hsf4 and DNase IIβ, which are implicated in the denucleation process. In summary, our data suggest that Snf2h is essential for the establishment of lens vesicle polarity, partitioning of prospective lens epithelial and fiber cell compartments, lens fiber cell differentiation, and lens fiber cell nuclear degradation.
Collapse
Grants
- R01 EY012200 NEI NIH HHS
- R01 CA079057 NCI NIH HHS
- R01 DK096266 NIDDK NIH HHS
- R01 GM116143 NIGMS NIH HHS
- R01 EY013022 NEI NIH HHS
- R01 CA076329 NCI NIH HHS
- T32 GM007491 NIGMS NIH HHS
- R56 CA079057 NCI NIH HHS
- R01 EY014237 NEI NIH HHS
- 001 World Health Organization
- R01 EY022645 NEI NIH HHS
- Grant support: R01 EY012200 (AC), EY014237 (AC), EY014237-7S1 (AC), EY013022 (MK), CA079057 (AIS), EY022645 (WL), T32 GM007491 (SL), GACR: P305/12/1033 (TS, JK), UNCE: 204021 (TS, JK), and an unrestricted grant from Research to Prevent Blindness to the Department of Ophthalmology and Visual Sciences. TS is member of the BIOCEV ? Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (CZ.1.05/1.1.00/02.0109) supported by the European Regional Development Fund. The Israel Science Foundation 610/10, the Israel Ministry of Science 36494, the Ziegler Foundation and the Binational Science Foundation (2013016) to RAP.
Collapse
Affiliation(s)
- Shuying He
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Saima Limi
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rebecca S McGreal
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qing Xie
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lisa A Brennan
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Wanda Lee Kantorow
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Juraj Kokavec
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Romit Majumdar
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Harry Hou
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wei Liu
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Jiri Zavadil
- Department of Pathology and NYU Center for Health Informatics and Bioinformatics, New York University Langone Medical Center, New York, NY 10016, USA Mechanisms of Carcinogenesis Section, International Agency for Research on Cancer, Lyon Cedex 08 69372, France
| | - Marc Kantorow
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Arthur I Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tomas Stopka
- First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Ales Cvekl
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
47
|
Yang J, Zhou S, Guo M, Li Y, Gu J. Different alpha crystallin expression in human age-related and congenital cataract lens epithelium. BMC Ophthalmol 2016; 16:67. [PMID: 27234311 PMCID: PMC4884376 DOI: 10.1186/s12886-016-0241-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 05/19/2016] [Indexed: 11/24/2022] Open
Abstract
Background The purpose of this study was to investigate the different expressions of αA-crystallin and αB-crystallin in human lens epithelium of age-related and congenital cataracts. Methods The central part of the human anterior lens capsule approximately 5 mm in diameter together with the adhering epithelial cells, were harvested and processed within 6 hours after cataract surgery from age-related and congenital cataract patients or from normal eyes of fresh cadavers. The mRNA and soluble protein levels of αA-crystallin and αB-crystallin in the human lens epithelium were detected by real-time PCR and western blots, respectively. Results The mRNA and soluble protein expressions of αA-crystallin and αB-crystallin in the lens epithelium were both reduced in age-related and congenital cataract groups when compared with the normal control group. However, the degree of α-crystallin loss in the lens epithelium was highly correlated with different cataract types. The α-crystallin expression of the lens epithelium was greatly reduced in the congenital cataract group but only moderately decreased in the age-related cataract group. The reduction of αA-crystallin soluble protein levels in the congenital cataract group was approximately 2.4 fold decrease compared with that of the age-related cataract group, while an mRNA fold change of 1.67 decrease was observed for the age-related cataract group. Similarly, the reduction of soluble protein levels of αB-crystallin in the congenital cataract group was approximately a 1.57 fold change compared with that of the age-related cataract group. A 1.75 fold change for mRNA levels compared with that of the age-related cataract group was observed. Conclusions The results suggest that the differential loss of α-crystallin in the human lens epithelium could be associated with the different mechanisms of cataractogenesis in age-related versus congenital cataracts, subsequently resulting in different clinical presentations.
Collapse
Affiliation(s)
- Jing Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54S Xianlie, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Sheng Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54S Xianlie, Guangzhou, Guangdong, 510060, People's Republic of China.,Department of ophthalmology, The First People's Hospital of Foshan, Guangdong Province, China
| | - Minfei Guo
- Department of Ophthalmology, Huichang County People's Hospital, Jiangxi, China
| | - Yuting Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54S Xianlie, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Jianjun Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54S Xianlie, Guangzhou, Guangdong, 510060, People's Republic of China.
| |
Collapse
|
48
|
Andjelic S, Drašlar K, Hvala A, Lopic N, Strancar J, Hawlina M. Anterior lens epithelial cells attachment to the basal lamina. Acta Ophthalmol 2016; 94:e183-8. [PMID: 26581784 DOI: 10.1111/aos.12902] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/01/2015] [Indexed: 01/19/2023]
Abstract
PURPOSE To study the structure of the anterior lens epithelial cells (aLECs) and the contacts of the aLECs with the basal lamina (BL) in order to understand their role in the lens epithelium's function. METHODS The aLCs (BL and associated aLECs) were obtained from routine uneventful cataract surgery, prepared for and studied by scanning electron microscopy (SEM), transmission electron microscopy (TEM) and confocal microscopy. RESULTS SEM shows that the basal surface of the aLECs (~10-15 μm) is with aLECs foldings (~1-3 μm) and extensions (~0.5-3 μm) attached to the BL. Confocal microscopy images of the basal sections of the aLECs after membrane staining also suggest that the basal part of aLECs has foldings (~1-3 μm). TEM shows in the aLECs basal parts, towards BL, the structures that look like entanglement (~1-4 μm). In cases where there is a swelling of the cytoplasm and offset of the aLECs from the BL, individual extensions (~0.5-2 μm) that extend to the BL are visible by TEM. CONCLUSIONS We provide detail evidence about the structural organization of the aLECs, in particular about their basal side which is in contact with the BL. This is supported by the complementary use of three techniques, SEM, TEM and confocal microscopy, each of them showing the same morphological features, the extensions and the entanglements of the aLECs cytoplasmic membrane at the border with the BL. The basal surface of the aLECs is increased. It suggests the functional importance of the contact between aLECs and BL.
Collapse
Affiliation(s)
| | - Kazimir Drašlar
- Department of Biology; Biotechnical Faculty; University of Ljubljana; Ljubljana Slovenia
| | - Anastazija Hvala
- Department of Pathology; Medical Faculty; University of Ljubljana; Ljubljana Slovenia
| | - Nina Lopic
- Laboratory of Biophysics; Condensed Matter Physics Department; “Jožef Stefan” Institute; Ljubljana Slovenia
| | - Janes Strancar
- Laboratory of Biophysics; Condensed Matter Physics Department; “Jožef Stefan” Institute; Ljubljana Slovenia
| | - Marko Hawlina
- Eye Hospital; University Medical Centre; Ljubljana Slovenia
| |
Collapse
|
49
|
Zheng YP, Zhang SB, Wang F, Liu H, Zhang W, Song B, Liu ZY, Xiong L, Fan YZ, Liao DY. Effects of lentiviral RNA interference-mediated downregulation of integrin-linked kinase on biological behaviors of human lens epithelial cells. Int J Ophthalmol 2016; 9:21-8. [PMID: 26949605 DOI: 10.18240/ijo.2016.01.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 06/09/2015] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the effects of lentivirus (LV) mediated integrin-linked kinase (ILK) RNA interference (RNAi) on biological behaviors of human lens epithelial cells (LECs). METHODS Human cataract LECs and immortalized human LEC line, human lens epithelial (HLE) B-3 cells were transfected by lentiviral vector expressing ILK-specific short hairpin RNA (shRNA) and then stimulated by transforming growth factor-β (TGF-β), the silencing of ILK gene and protein was identified by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot methods; biological behaviors including cell cycle and apoptosis, cell morphology, α-smooth muscle actin (SMA) stress fiber formation and cell migration were examined. RESULTS Remarkable decreases of ILK protein expression were detected in LECs carrying lentiviral ILK-shRNA vector; flow cytometry revealed arresting of cell cycle progression through the G1/S transition and higher apoptosis rate in ILK-RNAi-LV transfected cells. Less α-SMA stress fiber formation and migration was observed in ILK-RNAi-LV transfected LECs. CONCLUSION The present study demonstrated that ILK was an important regulator for LECs proliferation and migration. LV mediated ILK RNAi is an effective way to decrease ILK-regulated cell growth by arresting cell cycle progression and increasing cell apoptosis, as well as, to prevent cell migration by inhibiting TGF-β induced α-SMA stress fiber formation. Thus, LV mediated ILK RNAi might be useful to prevent posterior capsular opacification.
Collapse
Affiliation(s)
- Yu-Ping Zheng
- Department of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Shao-Bo Zhang
- Department of Ophthalmology, Tangdu Hospital, the Fourth Military Medical University, Xi'an 710038, Shaanxi Province, China
| | - Feng Wang
- Department of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hui Liu
- Department of Otolaryngology, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Wen Zhang
- Department of Otolaryngology, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Bin Song
- Department of Otolaryngology, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Zi-Yao Liu
- Department of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Lei Xiong
- Department of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ya-Zhi Fan
- Department of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ding-Ying Liao
- Department of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
50
|
Zheng HH, Xu GX, Guo J, Fu LC, Yao Y. Aquaporin-1 down regulation associated with inhibiting cell viability and inducing apoptosis of human lens epithelial cells. Int J Ophthalmol 2016; 9:15-20. [PMID: 26949604 DOI: 10.18240/ijo.2016.01.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/18/2015] [Indexed: 01/19/2023] Open
Abstract
AIM To investigate the role of Aquaporin-1 (AQP-1) in lens epithelial cells (LECs) and its potential target genes. AQP-1 is specifically expressed in LECs of eyes and is significant for lens homeostasis and transparency maintenance. Herein, AQP-1 expression in LECs was investigated to evaluate its influence on cell survival in association with its potential role in cataract formation. METHODS LECs were transfected with lentivirus carrying AQP-1 small interfering RNA (siRNA). Real-time polymerase chain reaction (PCR) and Western blotting were conducted to detect AQP-1 expression in LECs from different groups. Meanwhile, cell counting kit-8 (CCK-8) assay and flow cytometry were performed to measure LEC proliferation and apoptosis, respectively. RESULTS AQP-1 expression was significantly reduced in LECs, both at mRNA and protein levels (P<0.05), after siRNA treatment. Decreased cell viability was detected by CCK-8 assay in LECs with siRNA interference, compared to control cells (P<0.05). The apoptosis rate significantly increased in cells after siRNA interference (P<0.05). CONCLUSION The decreased cell viability following AQP-1 down regulation is largely due to its induction of apoptosis of LECs. AQP-1 reduction might lead to changes of physiological functions in LECs, which might be associated with the occurrence and development of cataracts.
Collapse
Affiliation(s)
- Hong-Hua Zheng
- Fujian Institute of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Guo-Xing Xu
- Fujian Institute of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Jian Guo
- Fujian Institute of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Li-Cheng Fu
- Fujian Institute of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Yao Yao
- Fujian Institute of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| |
Collapse
|