1
|
Li X, Liu A, Zhou Y, Qi H, Wang J, Chen M, Sun T, Wu J, Huang Y, Wang L. Proliferator-Activated Receptor Alpha Inhibits Abnormal Extracellular Matrix Accumulation and Maintains Energy Metabolism in Late-Onset Fuchs Endothelial Corneal Dystrophy. Invest Ophthalmol Vis Sci 2025; 66:36. [PMID: 40232711 PMCID: PMC12007668 DOI: 10.1167/iovs.66.4.36] [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: 11/18/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Purpose Fuchs endothelial corneal dystrophy (FECD) is the most common corneal endothelial dystrophy and guttae are crucial in causing progressive loss of corneal endothelium. This study aimed to find a way to inhibit the formation of guttae in FECD. Methods and Results Mitochondria fatty acid β-oxidation (FAO) and tricarboxylic acid (TCA) cycle processes were negatively enriched in the FECD group according to gene set enrichment analysis in GSE171830. In vivo UV-A-induced late-onset FECD mouse model were established. After irradiation, aged proliferator-activated receptor alpha (PPARα-/-) mice manifested greater corneal opacity, cornea edema, and varied corneal endothelial cell morphology compared with wild-type mice. The total metabolites in cornea of aged PPARα-/- mice and wild-type mice were detected by mass spectrometry. Metabolites of the FAO pathway were decreased in corneas of PPARα-/- mice, coincident with enzymes of FAO decreased in GSE171830. The score for FAO energy metabolism was negatively related to that of the TGF-β pathway according to gene set variation analysis. The express of alpha smooth muscle actin (αSMA) and Col1a were increased in aged PPARα-/- mice and small interfering PPARα B4G12 cell lines. After irradiation, activation or overexpression of PPARα demonstrated reduced corneal endothelial damage and reversal of Descemet membrane thickening, along with downregulation of fibrosis-related genes such as αSMA and collagen type I alpha 1 (Col1a). In vitro experiments revealed that fenofibrate could reverse fibrosis and damage of cell-to-cell connections induced by TGF-β. Additionally, fenofibrate was found to alleviate mitochondrial damage in B4G12 and increase oxygen consumption rates after TGF-β treatment. Conclusions Overall, we suggested that the overexpression or activation of PPARα can inhibit FAO energy dysfunction of corneal endothelium and the abnormal extracellular matrix formation in Descemet's membrane, which is the primary pathology of FECD. Thus, PPARα may be a potential target for attenuating the progression of FECD.
Collapse
Affiliation(s)
- Xiaoqi Li
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Chinese PLA Navy No. 971 Hospital, Qingdao, China
| | - Anqi Liu
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yannan Zhou
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Haolan Qi
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Junyi Wang
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Mingxiong Chen
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Tunan Sun
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Wu
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yifei Huang
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Liqiang Wang
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Chi M, Zhao Y, Yuan B, Qiu Z, Peng R, Hong J. MiR-23a-3p targets PTEN as a novel anti-ferroptosis regulator in Fuchs endothelial corneal dystrophy. Exp Eye Res 2025; 250:110180. [PMID: 39581360 DOI: 10.1016/j.exer.2024.110180] [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: 08/28/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Fuchs endothelial corneal dystrophy (FECD) is the leading cause of keratoplasty without drug treatment. Research indicated that oxidative stress and lipid peroxidation play significant roles in FECD. However, the underlying pathogenesis and potential treatment remain poorly understood. We analyzed the mRNA expression of FECD using the GEO database (GSE171830). Utilizing the STRING database and Cytoscape's MCODE plugin, we identified hub genes that intersect with ferroptosis-related genes listed in FerrDb. FECD cell and animal models were developed, induced by Ultraviolet A exposure. We assessed ferroptosis by measuring GPX4 expression and ROS fluorescence intensity. MiR-23a-3p was compared between FECD model and normal control, and the target gene PTEN was confirmed through Western blot and dual-luciferase reporter assays. Treatment with PTEN, PI3K, Akt, and mTOR inhibitors provided insights into the role of the PTEN/PI3K/Akt/mTOR pathway in FECD model. Corneal endothelium and cellular structure were evaluated before and after delivery of miR-23a-3p. Bioinformatics analysis of the GSE171830 revealed the top five hub genes: TP53, PTEN, EGFR, EPAS1, and IL-1β. Ferroptosis is the predominant mechanism in FECD pathogenesis, distinct from apoptosis and necrosis. We uncovered a protective role for miR-23a-3p in corneal endothelial cells (CEnCs), mitigating ferroptosis by downregulating PTEN. Corroborating this, bpV (a PTEN inhibitor) was found to attenuate ferroptosis in CEnCs. Mechanistically, PTEN inhibition coupled with sustained PI3K/Akt/mTOR pathway activation emerged as a protective strategy against ferroptosis in CEnCs. Ferroptosis contributes to FECD pathogenesis, and targeted delivery of miR-23a-3p as a ferroptosis inhibitor may offer therapeutic potential by regulating PTEN/PI3K/Akt/mTOR signaling.
Collapse
Affiliation(s)
- Miaomiao Chi
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Yaning Zhao
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Bowei Yuan
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Zifeng Qiu
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Rongmei Peng
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China.
| | - Jing Hong
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China.
| |
Collapse
|
3
|
Leng T, Zhang L, Ma J, Qu X, Lei B. Intrinsically bioactive multifunctional Poly(citrate-curcumin) for rapid lung injury and MRSA infection therapy. Bioact Mater 2024; 41:158-173. [PMID: 39131630 PMCID: PMC11314446 DOI: 10.1016/j.bioactmat.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Dysregulated inflammation after trauma or infection could result in the further disease and delayed tissue reconstruction. The conventional anti-inflammatory drug treatment suffers to the poor bioavailability and side effects. Herein, we developed an amphiphilic multifunctional poly (citrate-polyglycol-curcumin) (PCGC) nano oligomer with the robust anti-inflammatory activity for treating acute lung injury (ALI) and Methicillin-resistant staphylococcus aureus (MRSA) infected wound. PCGC demonstrated the sustained curcumin release, inherent photoluminescence, good cellular compatibility, hemocompatibility, robust antioxidant activity and enhanced cellular uptake. PCGC could efficiently scavenge nitrogen-based free radicals, oxygen-based free radicals, and intracellular oxygen species, enhance the endothelial cell migration and reduce the expression of pro-inflammatory factors through the NF-κB signal pathway. Combined the anti-inflammation and antioxidant properties, PCGC can shortened the inflammatory process. In animal model of ALI, PCGC was able to reduce the pulmonary edema, bronchial cell infiltration, and lung inflammation, while exhibiting rapid metabolic behavior in vivo. The MRSA-infection wound model showed that PCGC significantly reduced the expression of pro-inflammatory factors, promoted the angiogenesis and accelerated the wound healing. The transcriptome sequencing and molecular mechanism studies further demonstrated that PCGC could inhibit multiple inflammatory related pathways including TNFAIP3, IL-15RA, NF-κB. This work demonstrates that PCGC is efficient in resolving inflammation and promotes the prospect of application in inflammatory diseases as the drug-loaded therapeutic system.
Collapse
Affiliation(s)
- Tongtong Leng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Long Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Junping Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Xiaoyan Qu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
4
|
Tratnig-Frankl M, Luft N, Magistro G, Priglinger S, Ohlmann A, Kassumeh S. Hepatocyte Growth Factor Modulates Corneal Endothelial Wound Healing In Vitro. Int J Mol Sci 2024; 25:9382. [PMID: 39273330 PMCID: PMC11395100 DOI: 10.3390/ijms25179382] [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: 08/05/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
In this study, we assessed the impact of hepatocyte growth factor (HGF) on corneal endothelial cells (CECs), finding that HGF concentrations of 100-250 ng/mL significantly increased CEC proliferation by 30%, migration by 32% and improved survival under oxidative stress by 28% compared to untreated controls (p < 0.05). The primary objective was to identify non-fibrotic pharmacological strategies to enhance corneal endothelial regeneration, addressing a critical need in conditions like Fuchs' endothelial dystrophy (FED), where donor tissue is scarce. To confirm the endothelial nature of the cultured CECs, Na+/K+-ATPase immunohistochemistry was performed. Proliferation rates were determined through BrdU incorporation assays, while cell migration was assessed via scratch assays. Cell viability was evaluated under normal and oxidative stress conditions using WST-1 assays. To ensure that HGF treatment did not trigger epithelial-mesenchymal transition, which could lead to undesirable fibrotic changes, α-SMA staining was conducted. These comprehensive methodologies provided robust data on the effects of HGF, confirming its potential as a therapeutic agent for corneal endothelial repair without inducing harmful EMT, as indicated by the absence of α-SMA expression. These findings suggest that HGF holds therapeutic promise for enhancing corneal endothelial repair, warranting further investigation in in vivo models to confirm its clinical applicability.
Collapse
Affiliation(s)
- Merle Tratnig-Frankl
- Department of Ophthalmology, LMU University Hospital, Ludwig-Maximilians University Munich, Mathildenstrasse 8, 80336 Munich, Germany
- Department of Ophthalmology and Optometry, Medical University Vienna, AKH Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Nikolaus Luft
- Department of Ophthalmology, LMU University Hospital, Ludwig-Maximilians University Munich, Mathildenstrasse 8, 80336 Munich, Germany
| | - Guiseppe Magistro
- Department of Urology, Asklepios Westklinikum Hamburg GmbH, Suurheid 20, 22559 Hamburg, Germany
| | - Siegfried Priglinger
- Department of Ophthalmology, LMU University Hospital, Ludwig-Maximilians University Munich, Mathildenstrasse 8, 80336 Munich, Germany
| | - Andreas Ohlmann
- Department of Ophthalmology, LMU University Hospital, Ludwig-Maximilians University Munich, Mathildenstrasse 8, 80336 Munich, Germany
| | - Stefan Kassumeh
- Department of Ophthalmology, LMU University Hospital, Ludwig-Maximilians University Munich, Mathildenstrasse 8, 80336 Munich, Germany
| |
Collapse
|
5
|
Ling X, Xu W, Tang J, Cao Q, Luo G, Chen X, Yang S, Reinach PS, Yan D. The Role of Ubiquitination and the E3 Ligase Nedd4 in Regulating Corneal Epithelial Wound Healing. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 38888282 PMCID: PMC11186577 DOI: 10.1167/iovs.65.6.29] [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: 11/25/2023] [Accepted: 05/25/2024] [Indexed: 06/20/2024] Open
Abstract
Purpose Ubiquitination serves as a fundamental post-translational modification in numerous cellular events. Yet, its role in regulating corneal epithelial wound healing (CEWH) remains elusive. This study endeavored to determine the function and mechanism of ubiquitination in CEWH. Methods Western blot and immunoprecipitation were used to discern ubiquitination alterations during CEWH in mice. Interventions, including neuronally expressed developmentally downregulated 4 (Nedd4) siRNA and proteasome/lysosome inhibitor, assessed their impact on CEWH. In vitro analyses, such as the scratch wound assay, MTS assay, and EdU staining, were conducted to gauge cell migration and proliferation in human corneal epithelial cells (HCECs). Moreover, transfection of miR-30/200 coupled with a luciferase activity assay ascertained their regulatory mechanism on Nedd4. Results Global ubiquitination levels were markedly increased during the mouse CEWH. Importantly, the application of either proteasomal or lysosomal inhibitors notably impeded the healing process both in vivo and in vitro. Furthermore, Nedd4 was identified as an essential E3 ligase for CEWH. Nedd4 expression was significantly upregulated during CEWH. In vivo studies revealed that downregulation of Nedd4 substantially delayed CEWH, whereas further investigations underscored its role in regulating cell proliferation and migration, through the Stat3 pathway by targeting phosphatase and tensin homolog (PTEN). Notably, our findings pinpointed miR-30/200 family members as direct regulators of Nedd4. Conclusions Ubiquitination holds pivotal significance in orchestrating CEWH. The critical E3 ligase Nedd4, under the regulatory purview of miR-30 and miR-200, facilitates CEWH through PTEN-mediated Stat3 signaling. This revelation sheds light on a prospective therapeutic target within the realm of CEWH.
Collapse
Affiliation(s)
- Xuemei Ling
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Weiwei Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Tang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiongjie Cao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Guangying Luo
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shuai Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peter Sol Reinach
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Yan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Yan J, Mehta S, Patel K, Dhupar N, Little N, Ong Tone S. Transcription factor 4 promotes increased corneal endothelial cellular migration by altering microtubules in Fuchs endothelial corneal dystrophy. Sci Rep 2024; 14:10276. [PMID: 38704483 PMCID: PMC11069521 DOI: 10.1038/s41598-024-61170-8] [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: 01/11/2024] [Accepted: 05/02/2024] [Indexed: 05/06/2024] Open
Abstract
Fuchs endothelial corneal dystrophy (FECD) is a complex corneal disease characterized by the progressive decline and morphological changes of corneal endothelial cells (CECs) that leads to corneal edema and vision loss. The most common mutation in FECD is an intronic CTG repeat expansion in transcription factor 4 (TCF4) that leads to its altered expression. Corneal endothelial wound healing occurs primarily through cell enlargement and migration, and FECD CECs have been shown to display increased migration speeds. In this study, we aim to determine whether TCF4 can promote cellular migration in FECD CECs. We generated stable CEC lines derived from FECD patients that overexpressed different TCF4 isoforms and investigated epithelial-to-mesenchymal (EMT) expression, morphological analysis and cellular migration speeds. We found that full length TCF4-B isoform overexpression promotes cellular migration in FECD CECs in an EMT-independent manner. RNA-sequencing identified several pathways including the negative regulation of microtubules, with TUBB4A (tubulin beta 4A class IVa) as the top upregulated gene. TUBB4A expression was increased in FECD ex vivo specimens, and there was altered expression of cytoskeleton proteins, tubulin and actin, compared to normal healthy donor ex vivo specimens. Additionally, there was increased acetylation and detyrosination of microtubules in FECD supporting that microtubule stability is altered in FECD and could promote cellular migration. Future studies could be aimed at investigating if targeting the cytoskeleton and microtubules would have therapeutic potential for FECD by promoting cellular migration and regeneration.
Collapse
Affiliation(s)
- Judy Yan
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
| | - Shanti Mehta
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Keya Patel
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Narisa Dhupar
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Ness Little
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Stephan Ong Tone
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Canada.
| |
Collapse
|
7
|
Lu Z, Lin H, Li J, Feng Y. Deciphering the molecular symphony: Unraveling endothelial-to-mesenchymal transition in corneal endothelial cells. Exp Eye Res 2024; 240:109795. [PMID: 38253308 DOI: 10.1016/j.exer.2024.109795] [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: 11/26/2023] [Revised: 01/04/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024]
Abstract
Understanding the molecular complexity of this phenomenon provides innovative targets for maintaining phenotypic integrity during in vitro expansion, thereby advancing corneal endothelial tissue engineering. In this study, we established an in vitro model to simulate endothelial-to-mesenchymal transition (EndMT) in corneal endothelial cells. Through RNA sequencing, we identified 452 upregulated and 163 downregulated genes, resulting in a total of 615 differentially expressed genes. Key pathways enriched by GO and KEGG analysis include extracellular matrix (ECM) regulation and the PI3K-Akt signaling pathway. Potential hub proteins such as THBS1, ITGA5, COL1A1, and SNAI1/2 were also identified, and their dynamic changes at different time points (0, 2, 12, 24 h) were monitored. Uncovering these key pathways and genes may deepen our understanding of the mechanisms underlying EndMT in corneal endothelial cells, providing valuable insights for optimizing in vitro cultivation strategies.
Collapse
Affiliation(s)
- Zhaoxiang Lu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China; Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Haimiao Lin
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Jinming Li
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Yun Feng
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China; Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
8
|
Vercammen H, Miron A, Oellerich S, Melles GRJ, Ní Dhubhghaill S, Koppen C, Van Den Bogerd B. Corneal endothelial wound healing: understanding the regenerative capacity of the innermost layer of the cornea. Transl Res 2022; 248:111-127. [PMID: 35609782 DOI: 10.1016/j.trsl.2022.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/14/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
Abstract
Currently, there are very few well-established treatments to stimulate corneal endothelial cell regeneration in vivo as a cure for corneal endothelial dysfunctions. The most frequently performed intervention for a damaged or dysfunctional corneal endothelium nowadays is corneal endothelial keratoplasty, also known as lamellar corneal transplantation surgery. Newer medical therapies are emerging and are targeting the regeneration of the corneal endothelium, helping the patients regain their vision without the need for donor tissue. Alternatives to donor tissues are needed as the aging population requiring transplants, has further exacerbated the pressure on the corneal eye banking system. Significant ongoing research efforts in the field of corneal regenerative medicine have been made to elucidate the underlying pathways and effector proteins involved in corneal endothelial regeneration. However, the literature offers little guidance and selective attention to the question of how to fully exploit these pathways. The purpose of this paper is to provide an overview of wound healing characteristics from a biochemical level in the lab to the regenerative features seen in the clinic. Studying the pathways involved in corneal wound healing together with their key effector proteins, can help explain the effect on the proliferation and migration capacity of the corneal endothelial cells.
Collapse
Affiliation(s)
- Hendrik Vercammen
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Alina Miron
- Netherlands Institute for Innovative Ocular Surgery (NIIOS), Rotterdam, The Netherlands
| | - Silke Oellerich
- Netherlands Institute for Innovative Ocular Surgery (NIIOS), Rotterdam, The Netherlands
| | - Gerrit R J Melles
- Netherlands Institute for Innovative Ocular Surgery (NIIOS), Rotterdam, The Netherlands; Melles Cornea Clinic Rotterdam, The Netherlands
| | - Sorcha Ní Dhubhghaill
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium; Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium; Netherlands Institute for Innovative Ocular Surgery (NIIOS), Rotterdam, The Netherlands
| | - Carina Koppen
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium; Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium
| | - Bert Van Den Bogerd
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
9
|
Zhang X, Li Z, Liu X, Qin X, Luo J, Zhang W, Liu B, Wei Y. ZPI prevents ox-LDL-mediated endothelial injury leading to inhibition of EndMT, inflammation, apoptosis, and oxidative stress through activating Pi3k/Akt signal pathway. Drug Dev Res 2022; 83:1212-1225. [PMID: 35656597 DOI: 10.1002/ddr.21952] [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: 11/24/2021] [Revised: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 11/09/2022]
Abstract
Oxidized low-density lipoprotein (ox-LDL)-mediated endothelial dysfunction exerts an essential role in the development of atherosclerosis. Protein Z-dependent protease inhibitor (ZPI), a member of the serine protease inhibitor superfamily, could inhibit the function of activated coagulation factor X (FXa) via interaction with protein Z (PZ). Studies have pointed out that ZPI was statistically related to atherosclerotic diseases, which may have a robust cardiovascular protective effect. However, the underlying mechanism of ZPI on ox-LDL-mediated endothelial injury requires further elucidation. Human umbilical vein endothelial cells (HUVECs) were treated with ox-LDL (100 μg/ml) and ZPI (10 μg/ml). Cell viability was measured by the Cell Counting Kit-8 (CCK-8) assay. Cell apoptosis, oxidative stress, and endothelial-to-mesenchymal transition (EndMT) were analyzed by immunofluorescence (IF). Cell migration was measured using a wound-healing assay. Quantitative real-time polymerase chain reaction and western blot analysis were performed to determine messenger RNA and protein expression. Ox-LDL (100 μg/ml, 48 h) significantly reduced cell viability and migration, increased EndMT, inflammation, apoptosis, and oxidative stress. The related protein expression of phosphatidylinositol 3 kinase/protein kinase B (Pi3k/Akt) signal pathway in HUVECs was also simultaneously decreased. We also discovered that ZPI treatment could prevent ox-LDL-mediated endothelial injury through the improvement of cell viability and alleviation of apoptosis, oxidative stress, EndMT, and inflammation. Thus, the protective effect of ZPI on HUVECs may be mediated by activation of the Pi3k/Akt signal pathway. ZPI may exert an important protective role in HUVECs dysfunction triggered by ox-LDL via activation of the Pi3k/Akt signal pathway. Therefore, ZPI may possess potential therapeutic effects on atherosclerotic endothelial injury-related diseases.
Collapse
Affiliation(s)
- Xingxu Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangdong Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoming Qin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiachen Luo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenming Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Li R, Qu Y, Li X, Tao Y, Yang Q, Wang J, Diao Y, Li Q, Fang Y, Huang Y, Wang L. Molecular Hydrogen Attenuated N-methyl-N-Nitrosourea Induced Corneal Endothelial Injury by Upregulating Anti-Apoptotic Pathway. Invest Ophthalmol Vis Sci 2021; 62:2. [PMID: 34196654 PMCID: PMC8267183 DOI: 10.1167/iovs.62.9.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Previous work by our group has demonstrated the value of N-methyl-N-nitrosourea (MNU)-induced corneal endothelial decompensation in animal models. The aim of this study was to investigate the effect of molecular hydrogen (H2) on MNU-induced corneal endothelial cell (CEC) injury and the underlying mechanism. Methods MNU-induced animal models of CEC injury were washed with hydrogen-rich saline (HRS) for 14 days. Immunofluorescence staining, immunohistochemical staining, and corneal endothelial assessment were applied to determine architectural and cellular changes on the corneal endothelium following HRS treatment. MNU-induced cell models of CEC injury were co-cultured with H2. The effect of H2 was examined using morphological and functional assays. Results It was shown that MNU could inhibit the proliferation and specific physiological functions of CECs by increasing apoptosis and decreasing the expression of ZO-1 and Na+/K+-ATPase, whereas H2 improved the proliferation and physiological function of CECs by anti-apoptosis. Cell experiments further confirmed that H2 could reverse MNU damage to CECs by decreasing oxidative stress injury, interfering with the NF-κB/NLRP3 pathway and the FOXO3a/p53/p21 pathway. Conclusions This study suggests that topical application of H2 could protect CECs against corneal damage factors through anti-apoptotic effect, reduce the incidence and severity of corneal endothelial decompensation, and maintain corneal transparency.
Collapse
Affiliation(s)
- Runpu Li
- Medical School of Chinese PLA, Beijing, China.,Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yingxin Qu
- Department of Ophthalmology, Chinese Aerospace 731 Hospital, Beijing, China
| | - Xiaoqi Li
- Medical School of Chinese PLA, Beijing, China.,Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ye Tao
- Department of Ophthalmology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Qinghua Yang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junyi Wang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yumei Diao
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qian Li
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yifan Fang
- Medical School of Chinese PLA, Beijing, China.,Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yifei Huang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Liqiang Wang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Yu F, Zhang W, Yan C, Yan D, Zhou M, Chen J, Zhao X, Zhu A, Zhou J, Liu H, Sun H, Fu Y. PAX6, modified by SUMOylation, plays a protective role in corneal endothelial injury. Cell Death Dis 2020; 11:683. [PMID: 32826860 PMCID: PMC7442823 DOI: 10.1038/s41419-020-02848-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 11/21/2022]
Abstract
Treating corneal endothelial diseases tends to be challenging as human corneal endothelial cells (CECs) do not proliferate in vivo. The pathogenesis or mechanisms underlying injured CECs need further studies. The abnormal expression of PAX6, which is an essential transcription factor for corneal homeostasis, exhibits corneal endothelial defects. However, the effects of PAX6 protein involved in corneal endothelial wound process are still unknown. Here, we found the upregulated protein levels of PAX6 in human corneal endothelial monolayer after injury; the expression of PAX6 also increased in murine and rat corneal endothelium injury models. Enforced PAX6 expression could alleviate the damages to CECs via regulating permeability by prompting cellular tight junction. In addition, SUMOylation mainly happened on both K53 and K89 residues of 48-kD PAX6 (the longest and main isoform expressed in cornea), and de-SUMOylation promoted the stability of PAX6 protein in vitro. In CECs of SENP1+/− mice, increased SUMOylation levels leading to instability and low expression of PAX6, delayed the repair of CECs after injury. Furthermore, overexpression of PAX6 accelerated the rate of corneal endothelial repair of SENP1+/− mice. Our findings indicate that SENP1-mediated de-SUMOylation improving the stability of PAX6, amplifies the protective effects of PAX6 on corneal endothelial injuries, highlighting potentials of PAX6 and/or SUMOylation to be used as a treatment target for corneal endothelial disorders.
Collapse
Affiliation(s)
- Fei Yu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Weijie Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Chenxi Yan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Dan Yan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Meng Zhou
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Junzhao Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Xiangteng Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Aoxue Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huiqing Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Department of Pediatric Neurosurgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hao Sun
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China.
| | - Yao Fu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China.
| |
Collapse
|