1
|
Chen X, Li C, Li J, Guo Z, Zhang S, Guo C, Yan H. LncRNA HOTAIR Interaction With WTAP Promotes m6A Methyltransferase Complex Assembly and Posterior Capsule Opacification Formation by Increasing THBS1. Invest Ophthalmol Vis Sci 2025; 66:20. [PMID: 40341312 PMCID: PMC12068528 DOI: 10.1167/iovs.66.5.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
Purpose To explore the role of long non-coding RNAs (lncRNAs) and N6-methyladenosine (m6A) in posterior capsule opacification (PCO) and their underlying mechanisms. Methods The localization of lncRNAs and proteins was analyzed using fluorescence in situ hybridization and immunofluorescence staining. RNA m6A quantification, RNA immunoprecipitation, co-immunoprecipitation, MeRIP-seq, MeRIP-qPCR, western blotting, wound healing, and Transwell assays were applied to elucidate the underlying mechanisms. Results The levels of lncRNA HOX transcript antisense intergenic RNA (HOTAIR) and m6A methylation increased significantly during epithelial-mesenchymal transition (EMT) in lens epithelial cells (LECs). HOTAIR promoted EMT and m6A methyltransferase activity but had no effect on methyltransferase activity and was not modified by m6A. Nevertheless, HOTAIR interacted with WT1-associated protein (WTAP), a key m6A writer protein, facilitating WTAP-mediated recruitment of METTL3-METTL14 heterodimers and enhancing m6A modification. The HOTAIR/WTAP complex elevated m6A levels, thrombospondin 1 (THBS1) expression, and EMT in LECs. Conclusions LncRNA HOTAIR enhances the assembly of the WTAP/METTL3/METTL14 complex and promotes EMT in LECs by upregulating m6A modification and THBS1 expression. Targeting the HOTAIR/WTAP/THBS1 pathway may prevent or treat PCO.
Collapse
Affiliation(s)
- Xi Chen
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Chenshuang Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Jiankui Li
- Department of Gynecology & Obstetrics, The 960th Hospital of PLA, Jinan, Shandong, China
| | - Zaoxia Guo
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Siqi Zhang
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Chenjun Guo
- Department of Ophthalmology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Hong Yan
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
Chen S, Zhang C, Shen L, Hu J, Chen X, Yu Y. Noncoding RNAs in cataract formation: star molecules emerge in an endless stream. Pharmacol Res 2022; 184:106417. [PMID: 36038044 DOI: 10.1016/j.phrs.2022.106417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022]
Abstract
For decades, research on the pathological mechanism of cataracts has usually focused on the abnormal protein changes caused by a series of risk factors. However, an entire class of molecules, termed non-coding RNA (ncRNA), was discovered in recent years and proven to be heavily involved in cataract formation. Recent studies have recognized the key regulatory roles of ncRNAs in cataracts by shaping cellular activities such as proliferation, apoptosis, migration and epithelial-mesenchymal transition (EMT). This review summarizes our current insight into the biogenesis, properties and functions of ncRNAs and then discusses the development of research on ncRNAs in cataracts. Considering the significant role of ncRNA in cataract formation, research on novel associated regulatory mechanisms is urgently needed, and the development of therapeutic alternatives for the treatment of cataracts seems promising.
Collapse
Affiliation(s)
- Silong Chen
- Eye Center of the Second Affiliated Hospital, Medical College of Zhejiang University, 88 Jiefang Road, Hangzhou, China
| | - Chengshou Zhang
- Eye Center of the Second Affiliated Hospital, Medical College of Zhejiang University, 88 Jiefang Road, Hangzhou, China
| | - Lifang Shen
- Eye Center of the Second Affiliated Hospital, Medical College of Zhejiang University, 88 Jiefang Road, Hangzhou, China
| | - Jianghua Hu
- Eye Center of the Second Affiliated Hospital, Medical College of Zhejiang University, 88 Jiefang Road, Hangzhou, China; Department of Ophthalmology, Jiande Branch, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiangjun Chen
- Eye Center of the Second Affiliated Hospital, Medical College of Zhejiang University, 88 Jiefang Road, Hangzhou, China; Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kaixuan Road, China.
| | - Yibo Yu
- Eye Center of the Second Affiliated Hospital, Medical College of Zhejiang University, 88 Jiefang Road, Hangzhou, China.
| |
Collapse
|
3
|
Huai B, Huang C, Hu L. Curcumin suppresses TGF-β2-induced proliferation, migration, and invasion in lens epithelial cells by targeting KCNQ1OT1/miR-377-3p/COL1A2 axis in posterior capsule opacification. Curr Eye Res 2022; 47:715-726. [PMID: 35179079 DOI: 10.1080/02713683.2021.2021537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Posterior capsule opacification (PCO) is a common complication after cataract surgery, which can lead to secondary loss of vision. Curcumin has been reported to play a suppressive role in PCO progression, and the potential molecular mechanism was explored in this study. METHODS Cell viability and proliferation were analyzed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and 5-Ethynyl-2'-deoxyuridine (EdU) assay. Transwell assay and wound healing assay were performed to assess cell invasion and migration abilities. Western blot assay and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were conducted to measure the expression of proteins and RNAs. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted to confirm the interaction between microRNA-377-3p (miR-377-3p) and KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) or collagen type I alpha 2 chain (COL1A2). RESULTS Curcumin dose-dependently alleviated transforming growth factor-β2 (TGF-β2)-induced proliferation, migration, and invasion in SRA01/04 cells. KCNQ1OT1 was up-regulated in PCO patients and TGF-β2-induced SRA01/04 cells. Curcumin-induced protective effects in TGF-β2-induced SRA01/04 cells were largely overturned by KCNQ1OT1 overexpression. KCNQ1OT1 directly interacted with miR-377-3p and negatively regulated its expression. miR-377-3p silencing overturned Curcumin-mediated protective effects in SRA01/04 cells upon TGF-β2 treatment. miR-377-3p directly interacted with the 3' untranslated region (3'UTR) of COL1A2. COL1A2 overexpression largely counteracted KCNQ1OT1 silencing-induced effects in TGF-β2-stimulated SRA01/04 cells. KCNQ1OT1 could up-regulate COL1A2 expression by sponging miR-377-3p in SRA01/04 cells. CONCLUSION In conclusion, Curcumin suppressed TGF-β2-induced malignant changes in lens epithelial cells by targeting KCNQ1OT1/miR-377-3p/COL1A2 axis.
Collapse
Affiliation(s)
- Bin Huai
- Department of Pharmacy, Jinan Second People's Hospital (Jinan Eye Hospital), Jinan 250021, Shandong, China
| | - Chao Huang
- Department of Ophthalmology, Jinan Second People's Hospital (Jinan Eye Hospital), Jinan 250021, Shandong, China
| | - Lin Hu
- Department of Pharmacy, Jinan Maternal and Child Health Hospital, Jinan 250001, Shandong, China
| |
Collapse
|
4
|
Yu XH, Liu SY, Li CF. TGF-β2-induced NEAT1 regulates lens epithelial cell proliferation, migration and EMT by the miR-26a-5p/FANCE axis. Int J Ophthalmol 2021; 14:1674-1682. [PMID: 34804856 DOI: 10.18240/ijo.2021.11.05] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/05/2021] [Indexed: 01/15/2023] Open
Abstract
AIM To explore the regulatory mechanism of nuclear paraspeckle assembly transcript 1 (NEAT1) in the pathogenesis of posterior capsule opacification (PCO). METHODS Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was executed to analyze NEAT1 and microRNA (miR)-26a-5p expression in transforming growth factor-beta 2 (TGF-β2)-disposed lens epithelial cells (LECs). The proliferation, cell cycle progression, apoptosis, and migration of TGF-β2-disposed LECs were evaluated. The relationship between NEAT1 or fanconi anemia (FA) complementation group E (FANCE) and miR-26a-5p was verified by dual-luciferase reporter assay. RESULTS TGF-β2 induced NEAT1 expression in LECs. NEAT1 inhibition accelerated apoptosis, cell cycle arrest, decreased proliferation, epithelial-mesenchymal transition (EMT), and migration of TGF-β2-disposed LECs. NEAT1 sponged miR-26a-5p to further regulate FANCE expression. Rescue experiments presented that miR-26a-5p downregulation overturned NEAT1 silencing-mediated impacts on TGF-β2-disposed LEC biological behaviors. Additionally, FANCE overexpression reversed miR-26a-5p mimic-mediated impacts on TGF-β2-disposed LEC biological behaviors. CONCLUSION TGF-β2-induced NEAT1 facilitates LEC proliferation, migration, and EMT by upregulating FANCE via sequestering miR-26a-5p.
Collapse
Affiliation(s)
- Xiao-Hui Yu
- Department of Ophthalmology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong Province, China
| | - Shao-Yi Liu
- Department of Ophthalmology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong Province, China
| | - Cheng-Fang Li
- Department of Ophthalmology, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao 266033, Shandong Province, China
| |
Collapse
|
5
|
Meng K, Fang C. Knockdown of Tripartite motif-containing 22 (TRIM22)relieved the apoptosis of lens epithelial cells by suppressing the expression of TNF receptor-associated factor 6 (TRAF6). Bioengineered 2021; 12:7213-7222. [PMID: 34558381 PMCID: PMC8806417 DOI: 10.1080/21655979.2021.1980645] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cataract is a disease that causes severe visual impairment in patients. Recent studies have found that lens epithelial cell apoptosis caused by oxidative damage is the critical cause of cataract. Moreover, TRIM22 could alleviate the ubiquitination of TRAF6. The expression of TRAF6 could activate the p38/MAPK pathway and aggravate the oxidative stress induced damage of lens epithelial cells. However, whether the TRIM22 could alleviate the oxidative stress induced damage of lens epithelial cells by regulating the expression of TRAF6 and p38/MAPK pathway is unclear. In this study, we stimulated the lens epithelial cells with the H2O2 and established the TRIM22 knockdown cells. Next, proliferation of these cells was determined by CCK-8 and EdU assays. Apoptosis of these cells was detected with the TUNEL assays. Levels of ROS was explored with the DCFH-DA staining. Finally, the expression levels of TRAF6, p-p38 and p-ERK were determined with the western blotting. According to the results, we found that knockdown of TRIM22 suppressed the proliferation and relieved the H2O2 induced DNA double-strand break and apoptosis of these cells. Inhibition of TRIM22 inhibited the production of ROS in these cells. Moreover, restriction of TRIM22 induced the decreased levels of TRAF6, p-p38 and p-ERK in lens epithelial cells. We concluded that inhibition of TRIM22 relieved the apoptosis of lens epithelial cells by suppressing the expression of TRAF6, p-p38 and p-ERK.
Collapse
Affiliation(s)
- Kai Meng
- Department of Ophthalmology, Fuyang Futian Eye Hospital, Fuyang, Anhui Province, China
| | - Chengbo Fang
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
6
|
Wang H, Zheng G. LncRNA NEAT1 promotes proliferation, migration, invasion and epithelial-mesenchymal transition process in TGF-β2-stimulated lens epithelial cells through regulating the miR-486-5p/SMAD4 axis. Cancer Cell Int 2020; 20:529. [PMID: 33292220 PMCID: PMC7603719 DOI: 10.1186/s12935-020-01619-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/20/2020] [Indexed: 01/13/2023] Open
Abstract
Background Abnormal proliferation, metastasis and epithelial-mesenchymal transformation (EMT) of lens epithelial cells (LECs) are direct factors of posterior capsular opacification (PCO). Nuclear enriched abundant transcript 1 (NEAT1) has been shown to promote cell proliferation, metastasis and EMT, but whether it affects the progression of PCO is unclear. Methods The expression of NEAT1, microRNA-486-5p (miR-486-5p) and Drosophila mothers against decapentaplegic 4 (SMAD4) was determined using quantitative real-time polymerase chain reaction (qRT-PCR). The proliferation of cells was measured via 3-(4, 5-dimethyl-2 thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay. Transwell assay was employed to detect the migration and invasion of cells. The levels of EMT marker proteins, SMAD4 protein and transforming growth factor-β (TGF-β)/SMAD signaling pathway-related proteins were assessed by western blot (WB) analysis. Further, the relationship between miR-486-5p and NEAT1 or SMAD4 was confirmed by dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and biotin-labeled RNA pull-down assay. Results NEAT1 is upregulated and miR-486-5p is downregulated in the posterior capsular tissues of PCO patients and TGF-β2-induced LECs. Interference of NEAT1 reverses the promoting effect of TGF-β2 on the proliferation, migration, invasion and EMT of LECs. MiR-486-5p can be sponged by NEAT1, and its inhibitor reverses the suppression effect of NEAT1 silencing on the progression of TGF-β2-induced LECs. SMAD4 functions as a target of miR-486-5p, and its overexpression recovers the inhibition effect of miR-486-5p overexpression on the progression of TGF-β2-induced LECs. The activity of the TGF-β/SMAD signaling pathway is regulated by the NEAT1/miR-486-5p/SMAD4 axis. Conclusion Our study shows that NEAT1 has a positive effect on the progression of PCO and is expected to become a new target for PCO treatment.
Collapse
Affiliation(s)
- Huajun Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450000, Henan, China
| | - Guangying Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
7
|
Yao L, Yang L, Song H, Liu T, Yan H. MicroRNA miR-29c-3p modulates FOS expression to repress EMT and cell proliferation while induces apoptosis in TGF-β2-treated lens epithelial cells regulated by lncRNA KCNQ1OT1. Biomed Pharmacother 2020; 129:110290. [PMID: 32534225 DOI: 10.1016/j.biopha.2020.110290] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 05/13/2020] [Accepted: 05/16/2020] [Indexed: 10/24/2022] Open
Abstract
Age-related cataract (ARC) is a main cause of blindness for elderly people. MicroRNA hsa_miR-29c-3p (miR-29c-3p) was implicated in many biological processes in complicated diseases. However, the biological mechanism of miR-29c-3p in ARC is still undefined. Quantitative real-time polymerase chain reaction (qRT-PCR) showed that miR-29c-3p was lowly expressed, while FBJ murine osteosarcoma viral oncogene homolog (FOS) and KCNQ1 overlapping transcript 1 (KCNQ1OT1) were highly expressed in cataract tissues and in TGF-β2-treated SRA01/04 cells. Western blot assay indicated that TGF-β2 could promote epithelial-mesenchymal transition (EMT). Moreover, our data suggested that miR-29c-3p overexpression suppressed EMT, cell proliferation and promoted apoptosis in TGF-β2-treated SRA01/04 cells. The dual-luciferase reporter assay verified that FOS was a target of miR-29c-3p and miR-29c-3p was directly targeted by KCNQ1OT1. Furthermore, KCNQ1OT1 could regulate FOS expression by sponging miR-29c-3p. Functional assays revealed that miR-29c-3p regulated FOS to repress EMT, cell proliferation and facilitate apoptosis in TGF-β2-treated SRA01/04 cells mediated by KCNQ1OT1. In conclusion, KCNQ1OT1/miR-29c-3p/FOS axis played a vital role in the progression of ARC.
Collapse
Affiliation(s)
- Ling Yao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lei Yang
- Tianjin Key Laboratory of Tianjin Nankai Hospital, Tianjin, China
| | - Hui Song
- Department of Ophthalmology, Tianjin Eye Hospital, Tianjin, China
| | - Tiegang Liu
- Department of Ophthalmology, Beijing Capital International Airport Hospital, Beijing, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
8
|
Ling J, Tan K, Lu L, Yang F, Luan L. lncRNA MIAT increases cell viability, migration, EMT and ECM production in age-related cataracts by regulating the miR-181a/CTGF/ERK signaling pathway. Exp Ther Med 2020; 20:1053-1063. [PMID: 32742346 PMCID: PMC7388250 DOI: 10.3892/etm.2020.8749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 03/24/2020] [Indexed: 12/15/2022] Open
Abstract
Age-related cataract (ARC) is a common cause of blindness in elderly individuals. Long non-coding RNA (lncRNA) myocardial infarction associated transcript (MIAT) has been reported to participate in various biological processes in a number of diseases; however, the biological mechanism underlying MIAT during ARC is not completely understood. The expression levels of MIAT, microRNA (miR)-181a and connective tissue growth factor (CTGF) were measured by reverse transcription-quantitative PCR. The protein expression levels of CTGF, α-smooth muscle actin, fibronectin, collagen type I, ERK, phosphorylated (p)-ERK, mitogen-activated protein kinase (MEK), and p-MEK were detected by western blotting. Cell viability and migration were assessed using MTT and Transwell assays, respectively. Moreover, a dual-luciferase reporter assay was performed to investigate the interaction between miR-181a and MIAT or CTGF. MIAT and CTGF were upregulated, while miR-181a was significantly downregulated in ARC tissues compared with normal tissues. MIAT or CTGF knockdown decreased cell viability, migration, epithelial-mesenchymal transition and extracellular matrix production in TGF-β2-treated SRA01/04 cells. It was hypothesized that miR-181a may be sponged by MIAT and may target CTGF. Furthermore, the miR-181a inhibitor reversed the inhibitory effect of MIAT knockdown on the progression of TGF-β2-treated SRA01/04 cells. Moreover, CTGF knockdown also reversed MIAT overexpression-mediated progression of TGF-β2-treated SRA01/04 cells. In addition, MIAT and CTGF regulated the activity of the ERK signaling pathway. The results suggested that MIAT may regulate the progression of ARC via the miR-181a/CTGF/ERK signaling pathway, which may serve as a novel therapeutic target for ARC.
Collapse
Affiliation(s)
- Jiaojiao Ling
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Ke Tan
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Lu Lu
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Fang Yang
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Lan Luan
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|