1
|
Lyu Y, Yin Q, Liao X, Xie Y, Yang H, Cui Y, Han Y, Yao K, Wang C, Shentu X. Aminated fullerene for comprehensive dry eye therapy: Promoting epithelial-barrier reconstruction and nerve regeneration by suppressing oxidation and inflammation. Biomaterials 2025; 321:123329. [PMID: 40273474 DOI: 10.1016/j.biomaterials.2025.123329] [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: 11/20/2024] [Revised: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
Dry eye disease (DED) affects up to 50 % of the global population, leading to serious discomforts that affect patients' quality of life. In the multifactorial etiology of DED, oxidative stress is at the core, initiating a sequence of inflammatory responses and surface damage via a vicious cycle. However, current therapies merely have a narrow focus on inflammation. In this study, we developed a novel antioxidative eye drop, ethylenediamine (EDA)-modified C70 fullerene derivatives (abbreviated as FN-EDA), to break this vicious cycle. FN-EDA was successfully synthesized by modifying C70 fullerene with multiple ethylenediamine (EDA) groups, resulting in enhanced water solubility and a positive charge. This modification significantly improved ocular surface retention time, cellular uptake, and lysosomal escape in vitro. Therapeutically, FN-EDA significantly alleviated dry eye disease (DED) in a mouse model. It reduced corneal epithelial damage by 3.8-fold compared to 0.05 % cyclosporine A (CsA) and restored tear secretion to approximately 65 % of the normal level. Mechanistically, both in vivo and in vitro results demonstrate that FN-EDA is endowed with superior biological activity in effectively scavenging excessive oxidative stress, down-regulating proinflammatory cytokines expression, and promoting epithelial barrier reconstruction, even recovering corneal innervation. Thus, our findings open an avenue to make this multi-functional eye drop a promising candidate for DED.
Collapse
Affiliation(s)
- Ying Lyu
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Qichuan Yin
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Xiaodan Liao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Youxuan Xie
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Hao Yang
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Yilei Cui
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Yuqi Han
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Ke Yao
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Xingchao Shentu
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| |
Collapse
|
2
|
Wang B, Liang L, Zeng H, Yang X, Zhang R, Deng W, Wang X, Yuan J. Single-Cell RNA Sequencing Revealed Functional Conjunctival Keratinocytes Loss via TGF-β-Wnt/β-Catenin Signaling in Sjögren's Syndrome Related Dry Eye. Invest Ophthalmol Vis Sci 2025; 66:43. [PMID: 40238113 PMCID: PMC12011132 DOI: 10.1167/iovs.66.4.43] [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: 02/03/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose The role of the conjunctiva in the pathophysiology of Sjögren's syndrome (SS) related dry eye disease (DED) remains obscure especially in the view of functional conjunctival epithelia. In order to illustrate effects of conjunctiva in SS, we investigated the interactions between parenchymal cells and immune cells in the conjunctiva with single-cell RNA sequencing technique. Methods Freshly collected conjunctiva from a canonical SS model was prepared for 10 × Genomics single-cell RNA sequencing and T cell receptor (TCR) sequencing. Conjunctiva was collected for Western blot, immunofluorescence, multiplex immunohistochemical (mIHC), and flow cytometry. Phenol red thread test, lissamine staining, and qRT-PCR were applied to evaluate signs of DED. Results DED phenotype was validated in the SS model. Loss of water-secreting keratinocyte was projected in scRNA-seq data and proved by mIHC test in SS mice. The proportion of Lgr4+ basal epithelial cells with poor ability to differentiate into mature keratinocyte increased, and Wnt/β-catenin signaling was upregulated in it under regulation of TGF-β derived from macrophages. Such macrophages promoted angiogenesis through secretion of VEGFA to activate endothelial cells. Immuno-fibroblasts had an increased population, which were implicated in specifically activated T cell chemotaxis. Conclusions In SS conjunctiva, a TGF-β-Wnt/β-catenin axis downregulated the formation of functional keratinocytes accompanied by infiltration of pro-angiogenetic and pro-fibrotic macrophage and pro-inflammatory T cell.
Collapse
Affiliation(s)
- Bowen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, China
| | - Lihong Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, China
| | - Hao Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, China
| | - Xue Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, China
| | - Runze Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, China
| | - Wenrui Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, China
| | - Xiaoran Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, China
| | - Jin Yuan
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| |
Collapse
|
3
|
Lian L, Ye X, Wang Z, Li J, Wang J, Chen L, Reinach PS, Ma X, Chen W, Zheng Q. Hyperosmotic stress-induced NLRP3 inflammasome activation via the mechanosensitive PIEZO1 channel in dry eye corneal epithelium. Ocul Surf 2025; 36:106-118. [PMID: 39832672 DOI: 10.1016/j.jtos.2025.01.005] [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: 10/01/2024] [Revised: 12/10/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The activation of the NLRP3 inflammasome by hyperosmotic stress is a critical pathophysiological response in dry eye disease (DED), driving the chronic cycle of inflammation on the ocular surface. The specific mechanism underlying hyperosmotic mechanical stimulation activates the NLRP3 inflammasome remains unclear. This study provides evidence that PIEZO1, a mechanosensitive ion channel, functions as the primary receptor for corneal epithelial cells in sensing mechanical stimulation induced by tear hyperosmolarity. Inhibition of PIEZO1 significantly reduces NLRP3 inflammasome-associated pyroptosis in corneal epithelial cells. These findings suggest a therapeutic strategy targeting mechanosensitive ion channels to manage chronic ocular surface inflammation in DED patients. Structured Abstract. PURPOSE PIEZO1 modulates the inflammatory response by translating mechanical signals from osmotic pressure into biological processes. This study investigates the functional role of PIEZO1 in activating the NLRP3 inflammasome in corneal epithelial cells under hyperosmotic stress and evaluates its contribution to the pathogenesis of dry eye disease (DED). METHODS In the in vitro experiments, immortalized human corneal epithelial cells (HCECs) were cultured under hyperosmotic conditions (450mOsm). For in vivo studies, a dry eye disease mouse model was established by subcutaneous injection of scopolamine (SCOP) in C57BL/6 mice. After successfully inducing the dry eye model, corneal epithelial cell damage was assessed through corneal fluorescein staining scores and TUNEL assays. Protein expression levels were examined via western blotting and immunofluorescence staining, while mRNA expression was analyzed using quantitative RT-PCR. Activation of the NLRP3 inflammasome was evaluated by measuring IL-1β protein cleavage and the formation of ASC speckles. RESULTS In the DED model, activation of the NLRP3 inflammasome was detected in corneal epithelial cells, along with increased expression of PIEZO1. The PIEZO1-specific agonist Yoda1 induced upregulation of NLRP3 inflammasome-related gene expression and triggered NLRP3 inflammasome activation. Conversely, silencing PIEZO1 using siRNA or inhibiting its activity suppressed hyperosmotic stress-induced changes in NLRP3 inflammasome-related gene expression and activation. In vivo, PIEZO1 inhibition effectively prevented NLRP3 inflammasome activation in corneal epithelial cells and restored the damaged phenotype associated with dry eye disease. CONCLUSION Hyperosmotic stress-induced activation of the NLRP3 inflammasome in corneal epithelial cells is mediated through PIEZO1 activation. The identification of PIEZO1's role in this DED-related pathophysiological response highlights its potential as a therapeutic target for mitigating inflammation in clinical settings.
Collapse
Affiliation(s)
- Lili Lian
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China
| | - Xuanqiao Ye
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China
| | - Zimo Wang
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China
| | - Jiuxiao Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China
| | - Jiahe Wang
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China
| | - Letong Chen
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China
| | - Peter S Reinach
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China
| | - Xiaoyin Ma
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China.
| | - Wei Chen
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China.
| | - Qinxiang Zheng
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, 325000, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Zhejiang, 325000, China.
| |
Collapse
|
4
|
Liu L, Chen Y, Duan Y, Wang X, Chen Q, Yang Y, Lu Q, Shi L, Lin Q, Shen L. Fluorinated multifunctional polymer vesicles for enhanced ocular surface penetration and synergistic treatment of dry eye disease. J Control Release 2025; 379:592-608. [PMID: 39837386 DOI: 10.1016/j.jconrel.2025.01.040] [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: 10/17/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Current pharmacotherapy for DED is limited by a vicious inflammatory cycle in which reactive oxygen species (ROS) play a critical role. Additionally, topical eye drop therapy for DED often suffers from poor ocular availability due to multiple ocular surface barriers. Considering the key role of the ROS-NLRP3-IL-1β signaling axis in DED, in this investigation, fluorinated multifunctional polymer vesicles were developed for enhanced ocular surface penetration and synergistic DED therapy by combining ROS scavenging and immunomodulation. MCC950, an NLRP3-IL-1β inhibitor, was loaded in situ during vesicle preparation. The results demonstrated that fluorocarbon units randomly distributed in the corona layer significantly enhanced ocular surface penetration. Furthermore, the vesicle membrane, composed of polyphenylborate ester blocks, efficiently scavenged excess ROS in inflamed corneal tissue. In response to excessive ROS, a hydrophobic-to-hydrophilic conversion of the vesicle membrane facilitated the efficient release of MCC950 to modulate the NLRP3-caspase-1-IL1β pathway. We believe that this work will provide insightful guidance to achieve effective treatment of DED by enhancing ocular surface penetration.
Collapse
Affiliation(s)
- Lu Liu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yifei Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yong Duan
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xin Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qiumeng Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yuxi Yang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qunzan Lu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Quankui Lin
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| | - Liangliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
5
|
Hu J, Zeng Y, Tang L, Ye L, Chen C, Ling Q, Wang X, He L, Chen X, Wang Y, Ge Q, Shao Y. Topical Application of VitB6 Ameliorates PM2.5-Induced Dry Eye via NFκB Pathway in a Murine Model. Biomedicines 2025; 13:541. [PMID: 40149519 PMCID: PMC11939930 DOI: 10.3390/biomedicines13030541] [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: 01/12/2025] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Dry eye (DE) is mainly characterized by dryness, foreign body sensation, eye pain and visual impairment. Their possible causes are mainly inflammation, tissue damage and neurosensory abnormalities, and vitamin B6 (VitB6) attenuates the inflammatory response by modulating the NF-κB pathway to quench reactive oxygen species (ROS). The aim of this experiment was to investigate the therapeutic effect of VitB6 eye drops on particulate matter 2.5 (PM2.5)-induced dry eye in mice. Methods: Mice induced with the dry eye group were first induced using PM2.5 eye drops in a standard environment for 14 days, and then treated with different concentrations of VitB6 eye drops for 14 consecutive days. The phenol red cotton test was used to measure tear production. Ocular inflammation index and tear film function were evaluated by slim microscopy. Hematoxylin-eosin (HE) staining was used to observe conjunctival and corneal structure. Periodate-Schiff (PAS) staining was used to quantify conjunctival goblet cells. Corneal cell apoptosis was determined by TUNEL assay. The expression of keratin 10 (K10) and p-NF-κB p65 was detected by immunofluorescent staining and Western blot analysis. Results: Mice using only the PM2.5 model all exhibited varying degrees of dry eye symptoms. VitB6 treatment increased tear secretion and reduced inflammatory indices in mice with increased nerve density and number of branches in the basement membrane of the corneal epithelium. Conclusions: We found that administering VitB6 eye drops has a therapeutic effect in PM2.5-induced DE. This observation suggests that VitB6 may be useful in the clinical therapy of DE.
Collapse
Affiliation(s)
- Jinyu Hu
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yanmei Zeng
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Liying Tang
- Department of Ophthalmology, Zhongshan Hospital of Xiamen University, Xiamen 361102, China;
| | - Lei Ye
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Cheng Chen
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qian Ling
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaoyu Wang
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Liangqi He
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xu Chen
- Ophthalmology Centre, Maastricht University, 6200 MS Maastricht, Limburg Provincie, The Netherlands
| | - Yixin Wang
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Qianmin Ge
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yi Shao
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.H.); (Y.Z.); (L.Y.); (C.C.); (X.W.)
| |
Collapse
|
6
|
Hu X, Feng J, Pan C, Sun Z, Liu J, Xie S, Xiao D, Ma X, Zheng Q, Chen W. HMGB1 Promotes Lysosome-Dependent Cell Death Induced Via Dry Eye by Disrupting Lysosomal Homeostasis. Invest Ophthalmol Vis Sci 2025; 66:5. [PMID: 39903181 PMCID: PMC11801392 DOI: 10.1167/iovs.66.2.5] [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: 07/18/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Purpose Hypertonic stress can induce apoptosis, inflammation, and dry eye disease (DED) through the upregulation of HMGB1 expression. However, its role in mediating and maintaining lysosomal homeostasis and suppressing DED development in living and in vitro models is unclear. Methods Immunofluorescence, nucleoplasmic separation, and electron microscopic analysis were used to compare the effects of hypertonic stress on lysosomal function, nuclear HGMB1 expression, and lysosomal localization in three different dry eye models. The live model was established by the subcutaneous injection of scopolamine (SCOP) into C57BL/6J female mice, and the in vitro model used human corneal epithelial cell (HCEC) cultures that were maintained at a hyperosmotic level of 450 milliosmolar (mOsm). RNAi technology was used to knockdown HMGB1 expression levels, altering hyperosmotic stress-induced changes in survival and autophagy of corneal epithelial cells in vitro. Lysosomal protease inhibitors were applied to suppress the increase of corneal tissue inflammation in the dry eye mouse model. Results This hypertrophic stress upregulated karyoplasmic HMGB1 expression in HCECs. This change disrupted lysosomal structural and functional integrity, which facilitated the release of HMGB1 into the cytoplasm. SiRNAs downregulated HMGB1 expression levels and reversed increases in HMGB1 transfer from the nucleus to the cytoplasm and lysosomal alkalinizing function. These changes promoted increases in cathepsin leakage from lysosomes and increased mouse corneal epithelial apoptosis, whereas autophagy decreased. In a mouse model, administration of a cathepsin inhibitor reduced both ocular inflammation and other aspects of dry eye symptomology. Conclusions Hyperosmotic-induced HMGB1 upregulation and increased HMGB1 transfer into the cytoplasm from the nucleus underlie the loss of lysosomal membrane integrity, which increases both cathepsin release and corneal epithelial apoptosis. HMGB1 downregulation instead ameliorated these pathogenic responses.
Collapse
Affiliation(s)
- Xiaojuan Hu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiayao Feng
- Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, China
| | - Chengjie Pan
- Ophthalmology Department, First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhenzhen Sun
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiechen Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shuang Xie
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Decheng Xiao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qinxiang Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Zhang R, Zou Y, Li H, Li D, Liu Y, Gong B, Yu M. NK1R Antagonist, CP-99,994 Ameliorates Dry Eye Disease via Inhibiting the Plk1-Cdc25c-Cdk1 Axis. J Biochem Mol Toxicol 2025; 39:e70177. [PMID: 39967330 DOI: 10.1002/jbt.70177] [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: 04/01/2024] [Revised: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Substance P/high-affinity neurokinin-1 receptor (SP/NK1R) system plays a crucial role in the pathogenesis of dry eye disease (DED). NK1R antagonist can improve DED, but the mechanism of NK1R antagonist treating DED remains unclear. We examined the role of NK1R antagonist, CP-99,994 in DED model by possessing the phenol red cotton thread test, corneal fluorescein staining, and hematoxylin and eosin staining. Enzyme linked immunosorbent assay was performed to determine the concentration of inflammatory factors. Additionally, RNA sequencing, enrichment analysis and protein-protein interaction network were employed to identify the key targets. Real-time quantitative PCR and western blot analysis were utilized to determine the expression of hub genes. Plk1 inhibitor, GSK461364 was applied to explore the treatment mechanism of CP-99,994. The NK1R antagonist CP-99,994 alleviated dry eye symptoms and the concentrations of IL-6, IL-1β, and TNF-α were significantly decreased after CP-99,994 treatment. We obtained 68 differentially expressed genes after CP-99,994 treatment by RNA sequencing and pyroptosis-related genes (Plk1, Cdc25c, Cdk1) were identified from protein-protein interaction network as key targets of CP-99,994 treating DED. The expression levels of the Plk1, Cdc25c, and Cdk1 were significantly upregulated in the DED group, and CP-99,994 downregulated the expression of Plk1, Cdc25c, and Cdk1. Moreover, Plk1 inhibitor considerably promoted the therapeutic effect of CP-99,994 on DED model by reducing the release of IL-6, IL-1β, and TNF-α. The NK1R antagonist, CP-99,994 mitigated DED symptoms via inhibiting Plk1-Cdc25c-Cdk1 axis, which served as a novel therapeutic target for DED treatment.
Collapse
Affiliation(s)
- Ruifan Zhang
- Department of Ophthalmology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
| | - Yuhao Zou
- Department of Ophthalmology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
| | - Huan Li
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
- Department of Health Management, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
| | - Dongfeng Li
- Department of Ophthalmology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
| | - Yi Liu
- Department of Ophthalmology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
| | - Bo Gong
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
| | - Man Yu
- Department of Ophthalmology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, China
| |
Collapse
|
8
|
Wang L, Tian Y, Zhang H, Dong Y, Hua X, Yuan X. Caspase 3/GSDME-Mediated Corneal Epithelial Pyroptosis Promotes Dry Eye Disease. Invest Ophthalmol Vis Sci 2025; 66:24. [PMID: 39792075 PMCID: PMC11731164 DOI: 10.1167/iovs.66.1.24] [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: 08/23/2024] [Accepted: 12/14/2024] [Indexed: 01/30/2025] Open
Abstract
Purpose Dry eye disease (DED) is a common ocular surface inflammatory disease with a complex pathogenesis. Herein, the role and effect of gasdermin E (GSDME) in DED pathogenesis were explored. Methods In vitro, flow cytometry, Cell Counting Kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assays were used to determine the effects of hyperosmotic stress on pyroptosis, apoptosis, and cell viability in human corneal epithelial cells (HCECs). Quantitative PCR (qPCR) and Western blot assays were used to detect GSDME expression in HCECs and in those transfected with si-GSDMD. In vivo, GSDMD-knockout (KO) mice were used to study the role of GSDME in DED pathogenesis. The qPCR, Western blotting, and immunofluorescence were used to explore the effects of GSDME on HCEC apoptosis, pyroptosis, and the expression of related genes and proteins in GSDMD-KO mice with scopolamine-induced dry eye. Results Pyroptosis and cell membrane rupture occurred, and caspase-3 and GSDME protein expression increased after HCECs were treated with 312 to 500 mOsm sodium chloride. GSDME gene and protein expression levels were increased in HCECs from both si-GSDMD- and GSDMD-KO mice. Although caspase-3 expression was increased in the dry eye group of GSDMD-KO mice, HCEC apoptosis and the apoptosis-related factors PARP were not detected. The gene and protein expression levels of the pyroptosis-related factors ASC and IL-1β were greater than those in GSDMD-KO mice without dry eye. Conclusions GSDME is involved in DED pathogenesis by mediating inflammation via the pyroptosis pathway, GSDME inhibition may be a therapeutic target for DED.
Collapse
Affiliation(s)
- Ling Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Ye Tian
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Hui Zhang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Yongxiao Dong
- Department of Ophthalmology, The First People's Hospital of Xianyang City, Xianyang, China
| | - Xia Hua
- Aier Eye Institute, Changsha, China
- Tianjin Aier Eye Hospital, Tianjin, China
| | - Xiaoyong Yuan
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| |
Collapse
|
9
|
Guo C, Yu M, Liu J, Jia Z, Liu H, Zhao S. Molecular mechanism of Wilms tumour 1-associated protein in diabetes-related dry eye disease by mediating m6A methylation modification of lncRNA NEAT1. J Drug Target 2024; 32:200-212. [PMID: 38153328 DOI: 10.1080/1061186x.2023.2300682] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/24/2023] [Indexed: 12/29/2023]
Abstract
Background: Dry eye disease (DED) is often secondary to diabetes mellitus (DM).Purpose: This study is to explore the action of Wilms tumor 1-associated protein (WTAP) in DM-DED via lncRNA NEAT1 m6A methylation.Methods: DM-DED mouse models were treated with sh-WTAP/sh-NEAT1, followed by assessment of corneal epithelial damage/histopathological changes. HCE-2 cells were exposed to hyperosmotic conditions to establish in vitro DED models and treated with oe-NEAT1/sh-NEAT1/sh-WTAP/nigericin (an NLRP3 inflammasome inducer). Cell viability/apoptosis were evaluated by CCK-8/TUNEL. Levels of WTAP/NEAT1/inflammatory factors/NLRP3 inflammasome- and apoptosis-related markers were determined. m6A modification was examined by MeRIP-qPCR and NEAT1 stability was also detected.Results: DM-DED mice exhibited up-regulated WTAP/NEAT1 expression and severe corneal damage, whereas WTAP/NEAT1 knockdown alleviated inflammation/corneal damage. In hyperosmolarity-induced HCE-2 cells, NEAT1 aggravated inflammation and apoptosis, while NEAT1 knockdown suppressed NLRP3 inflammasome activation and ameliorated cell injury. Hyperosmolarity-induced WTAP expression increased m6A modification and NEAT1 mRNA stability. WTAP mediated m6A methylation of NEAT1 and NLRP3 inflammasome activation in DM-DED mice.
Collapse
Affiliation(s)
- Chen Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Mingyi Yu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Jinghua Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Zhe Jia
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Hui Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Shaozhen Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
10
|
Huang D, Li Z. Multidimensional immunotherapy for dry eye disease: current status and future directions. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1449283. [PMID: 39554604 PMCID: PMC11564177 DOI: 10.3389/fopht.2024.1449283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024]
Abstract
Dry Eye Disease (DED) is a multifactorial condition driven by tear film hyperosmolarity, immune dysregulation, and neuro-immune interactions. The immune system plays a central role in its pathogenesis, influencing both inflammation and ocular surface damage. While traditional immunotherapies like anti-inflammatory agents and immunosuppressants offer symptom relief, their long-term use is limited by side effects. This review focuses on emerging immunotherapies, including biologics, stem cell therapy, gene therapy, nanotechnology, and exosome-based treatments, all of which hold promise in modulating immune responses and promoting tissue repair. The relationship between the ocular microbiome and DED is also explored, with an emphasis on personalized immunotherapy. Key challenges for future research include identifying novel therapeutic targets, optimizing clinical translation, and evaluating the long-term efficacy of these innovative treatments.
Collapse
Affiliation(s)
- Duliurui Huang
- Department of Ophthalmology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Henan University, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhijie Li
- Department of Ophthalmology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Henan University, People’s Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Yang M, Pan H, Chen T, Chen X, Ning R, Ye Q, Chen A, Li J, Li S, Zhao N, Wu Y, Fu X, Meek KM, Chen L, Wang X, Chen Z, Zhou X, Huang J. Customized Corneal Cross-Linking with Microneedle-Mediated Riboflavin Delivery for Keratoconus Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408136. [PMID: 39246198 DOI: 10.1002/adma.202408136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/18/2024] [Indexed: 09/10/2024]
Abstract
In this study, a novel customized corneal cross-linking (CXL) treatment is explored that utilizes microneedles (MNs) for targeted riboflavin (RF) administration prior to the CXL procedure. Unlike the conventional "one-size-fits-all" approach, this protocol offers an option for more precise and efficacious treatment. To simulate a customized corneal crosslinking technique, four distinct microneedle (MN) molds designs, including circular, semi-circular, annular and butterfly shaped, are crafted for loading an optimized RF-hyaluronic acid solution and for the subsequent fabrication of MN arrays with varying morphologies. These MNs can gently puncture the corneal epithelium while preserving the integrity of the underlying stromal layer. Following the application of these microneedles, RF solution is replenished to enhance the RF content within the stroma through the punctures created by the MNs, resulting in exceptional customized corneal cross-linking effects that are comparable to the conventional epi-off CXL protocol. Additionally, it flattened the corneal curvature within the treated zone and facilitated rapid postoperative recovery of corneal tissue. These findings suggest that the integration of customized microneedle RF delivery with corneal crosslinking technology represents a potential novel treatment modality, holding promise for the tailored treatment of corneal pathologies, and offering a more precise and efficient alternative to traditional methods.
Collapse
Affiliation(s)
- Mei Yang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Hongxian Pan
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Tingting Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Xin Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Rui Ning
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Qianfang Ye
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Aodong Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jiawei Li
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Siheng Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Nan Zhao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Yue Wu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Xueyu Fu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Keith M Meek
- School of Optometry and Vision Sciences, Cardiff Institute for Tissue Engineering and Repair School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Lingxin Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Xiaoying Wang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Zhongxing Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Xingtao Zhou
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| | - Jinhai Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai Research Center of Ophthalmology and Optometry, Fudan University, Shanghai, 200030, China
| |
Collapse
|
12
|
Li J, Bao X, Guo S, Huang Y, Huang C, Hu J, Liu Z. Cell death pathways in dry eye disease: Insights into ocular surface inflammation. Ocul Surf 2024; 34:535-544. [PMID: 39542089 DOI: 10.1016/j.jtos.2024.11.004] [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: 08/20/2024] [Revised: 10/11/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Dry eye disease (DED) is increasingly prevalent, with inflammation playing a crucial role in its pathogenesis. Severe cases of DED result in significant ocular discomfort and visual impairment due to damage and loss of ocular surface epithelial cells. The precise mechanisms underlying the loss of these epithelial cells remain a subject of ongoing research and debate. Programmed cell death (PCD) mechanisms, including pyroptosis, apoptosis, and necroptosis, are known to be critical in maintaining ocular surface homeostasis and responding to stressors in DED. The concept of PANoptosis, which integrates elements of various PCD pathways, has been implicated in the development of numerous systemic diseases, including infections, cancer, neurodegenerative, and inflammatory conditions. It also provides novel insights into the inflammatory processes underlying DED. This review highlights the crosstalk of PCD pathways in DED, particularly the significance of PANoptosis in ocular inflammation and its potential as a therapeutic target for more effective interventions.
Collapse
Affiliation(s)
- Jiani Li
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Xiaorui Bao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Shujia Guo
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yuhan Huang
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Caihong Huang
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jiaoyue Hu
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China.
| | - Zuguo Liu
- Xiamen University Affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
13
|
Rivera LA, Hernández PE, Vannan DT, Reyes JL, Rodríguez T, Sánchez-Barrera Á, González MI, Bustos J, Ramos OA, Juárez I, Rodriguez-Sosa M, Vázquez A. Macrophage Migration Inhibitory Factor (MIF) is a Key Player in Dry Eye Disease. Ocul Immunol Inflamm 2024; 32:1707-1721. [PMID: 38127798 DOI: 10.1080/09273948.2023.2290624] [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: 08/03/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE To explore the role of the proinflammatory cytokine, macrophage migration inhibitory factor (MIF), in a murine model of dry eye disease (DED). METHODS The role of MIF on DED was determined using genetically MIF deficient mice and pharmacological inhibition of MIF. DED was induced with 0.5 mg of scopolamine via subcutaneous injection in wild type (WT) and mice lacking MIF (Mif-/-), three times a day for 21 days. DED signs, tear volume, ferning pattern and cytology impression were evaluated. Also, eye tissues were collected to determine transcripts of key inflammatory mediators and histopathological damage. In a second set of experiments, we neutralized MIF with ISO-1, an isozaxiline-derivative MIF tautomerase activity-inhibiting small molecule in WT mice, following an acute DED model for 10 days. ISO-1 was given starting on day 3 after DED induction and signs were evaluated, including a recovery phase in both experimental approaches. RESULTS When compared to WT, Mif-/- mice showed attenuated signs of DED like preserved mucin pattern and increased tear volume. Also, Mif-/- mice maintained conjunctival epithelial cells and less corneal damage, associated with lower levels of TNFα and IL-1β. At recovery phase, Mif-/- mice presented improved signs. Interestingly, in cornea and conjunctiva the absence of MIF selectively downregulated the transcription of inflammatory enzymes like inos and nox4 whereas displayed enhanced transcripts of il-4, il-13, tgfβ and cox2. Finally, pharmacological inhibition of MIF using ISO-1, replicated the above findings in the mouse model. CONCLUSION MIF is a central positive mediator of the inflammatory process in experimental DED, thus, targeting MIF could be used as a novel therapy in ocular surface inflammatory pathologies.
Collapse
Affiliation(s)
- Luis A Rivera
- Laboratorio de Enfermedades Inflamatorias Oculares, Carrera de Optometría, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Pablo E Hernández
- Laboratorio de Enfermedades Inflamatorias Oculares, Carrera de Optometría, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Danielle T Vannan
- Boston Scientific, 300 Boston Scientific Way, Marlborough, Massachusetts, USA
| | - José L Reyes
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Tonathiu Rodríguez
- Laboratorio de Enfermedades Inflamatorias Oculares, Carrera de Optometría, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Ángel Sánchez-Barrera
- Laboratorio de Inmunoparasitología, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Marisol I González
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - José Bustos
- Laboratorio de Biología Molecular e Inmunología de Arbovirus, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, Estado de México
| | - Oscar A Ramos
- Laboratorio de Enfermedades Inflamatorias Oculares, Carrera de Optometría, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Imelda Juárez
- Laboratorio de Inmunidad Innata, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Miriam Rodriguez-Sosa
- Laboratorio de Inmunidad Innata, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| | - Alicia Vázquez
- Laboratorio de Enfermedades Inflamatorias Oculares, Carrera de Optometría, FES Iztacala, UNAM, Tlalnepantla de Baz, México
| |
Collapse
|
14
|
Ge J, Li X, Xia Y, Chen Z, Xie C, Zhao Y, Chen K, Shen Y, Tong J. Recent advances in NLRP3 inflammasome in corneal diseases: Preclinical insights and therapeutic implications. Ocul Surf 2024; 34:392-405. [PMID: 39357820 DOI: 10.1016/j.jtos.2024.09.007] [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: 04/17/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
NLRP3 inflammasome is a cytosolic multiprotein complex formed in response to exogenous environmental stress and cellular damage. The three major components of the NLRP3 inflammasome are the innate immunoreceptor protein NLRP3, the adapter protein apoptosis-associated speck-like protein containing a C-terminal caspase activation and recruitment domain, and the inflammatory protease enzyme caspase-1. The integrated NLRP3 inflammasome triggers the activation of caspase-1, leading to GSDMD-dependent pyroptosis and facilitating the maturation and release of inflammatory cytokines, namely interleukin (IL)-18 and IL-1β. However, the inflammatory responses mediated by the NLRP3 inflammasome exhibit dual functions in innate immune defense and cellular homeostasis. Aberrant activation of the NLRP3 inflammasome matters in the etiology and pathophysiology of various corneal diseases. Corneal alkali burn can induce pyroptosis, neutrophil infiltration, and corneal angiogenesis via the activation of NLRP3 inflammasome. When various pathogens invade the cornea, NLRP3 inflammasome recognizes pathogen-associated molecular patterns or damage-associated molecular patterns to engage in pro-inflammatory and anti-inflammatory mechanisms. Moreover, chronic inflammation and proinflammatory cascades mediated by the NLRP3 inflammasome contribute to the pathogenesis of diabetic keratopathy. Furthermore, overproduction of reactive oxygen species, mitochondrial dysfunction, and toll-like receptor-mediated activation of nuclear factor kappa B drive the stimulation of NLRP3 inflammasome and participate in the progression of dry eye disease. However, there still exist controversies regarding the regulatory pathways of the NLRP3 inflammasome. In this review, we provide a comprehensive overview of recent advancements in the function of NLRP3 inflammasome in corneal diseases and its regulatory pathways primarily through studies using animal models. Furthermore, we explore prospects for pharmacologically targeting pathways associated with NLRP3.
Collapse
Affiliation(s)
- Jiayun Ge
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiang Li
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yutong Xia
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhitong Chen
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chen Xie
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Zhao
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kuangqi Chen
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Jinan, Shandong, China; School of Ophthalmology, Shandong First Medical University, Jinan, Shandong, China.
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Huang Y, Ding X, Zhu L, Zhang X, Wang X, Ma F, Chen Y, Nan K. Anti-oxidative mesoporous polydopamine-based hypotensive nano-eyedrop for improved glaucoma management. Colloids Surf B Biointerfaces 2024; 245:114261. [PMID: 39317041 DOI: 10.1016/j.colsurfb.2024.114261] [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: 02/13/2024] [Revised: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
Conventional hypotensive eye drops remain suboptimal for glaucoma management, primarily due to their limited intraocular bioavailability and the growing concern regarding ocular surface side effects. Therefore, there is an urgent need to develop innovative intraocular pressure (IOP)-lowering formulations that not only possess enhanced corneal penetration ability but also provide ocular surface protection. Herein, anti-oxidative mesoporous polydopamine nanoparticles (MPDA NPs) were explored as a nano-carrier for Brimonidine to address the above issues. Nearly monodisperse MPDA NPs with obvious nanopores were successfully prepared by template-removal method and used for encapsulation of Brimonidine benefiting from their high specific surface area. Interestingly, the PEGylated and drug loaded MPDA-PEG@Brim NPs showed a near neutral surface charge, which is expected to enhance intraocular drug delivery. Consequently, much higher concentration of Brimonidine in the aqueous humor was found after topical administration of MPDA-PEG@Brim nano-dispersion as compared to free Brimonidine solution. Accordingly, superior IOP reduction effect was achieved for the nano-formulation in both hypertensive and normotensive rat eyes. Moreover, MPDA-PEG NPs showed good capability in scavenging diverse free radicals, alleviating intracellular oxidative stress, and mitigating ocular surface oxidative level in a mouse model of preservative-induced dry eye. In addition, the excellent biosafety of this novel Brimonidine nanodrug was confirmed both in vitro and in vivo. Therefore, the present work may shed light on the development of next generation hypotensive formulations for extended ocular surface protection and glaucoma management.
Collapse
Affiliation(s)
- Yate Huang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiaoxu Ding
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Li Zhu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xuehan Zhang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiaoxue Wang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Feiyan Ma
- The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050004, China
| | - Yangjun Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Institute of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Kaihui Nan
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Institute of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
16
|
Ouyang W, Yan D, Hu J, Liu Z. Multifaceted mitochondrial as a novel therapeutic target in dry eye: insights and interventions. Cell Death Discov 2024; 10:398. [PMID: 39242592 PMCID: PMC11379830 DOI: 10.1038/s41420-024-02159-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
Dry eye, recognized as the most prevalent ocular surface disorder, has risen to prominence as a significant public health issue, adversely impacting the quality of life for individuals across the globe. Despite decades of extensive research into the chronic inflammation that characterizes dry eye, the intricate mechanisms fueling this persistent inflammatory state remain incompletely understood. Among the various cellular components under investigation, mitochondria-essential for cellular energy production and homeostasis-have attracted increasing attention for their role in dry eye pathogenesis. This involvement points to mechanisms such as oxidative stress, apoptosis, and sustained inflammation, which are central to the progression of the disease. This review aims to provide a thorough exploration of mitochondrial dysfunction in dry eye, shedding light on the critical roles played by mitochondrial oxidative stress, apoptosis, and mitochondrial DNA damage. It delves into the mechanisms through which diverse pathogenic factors may trigger mitochondrial dysfunction, thereby contributing to the onset and exacerbation of dry eye. Furthermore, it lays the groundwork for an overview of current therapeutic strategies that specifically target mitochondrial dysfunction, underscoring their potential in managing this complex condition. By spotlighting this burgeoning area of research, our review seeks to catalyze the development of innovative drug discovery and therapeutic approaches. The ultimate goal is to unlock promising avenues for the future management of dry eye, potentially revolutionizing treatment paradigms and improving patient outcomes. Through this comprehensive examination, we endeavor to enrich the scientific community's understanding of dry eye and inspire novel interventions that address the underlying mitochondrial dysfunctions contributing to this widespread disorder.
Collapse
Affiliation(s)
- Weijie Ouyang
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Xiamen, Fujian, China; Department of Ophthalmology, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Dan Yan
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Xiamen, Fujian, China
| | - Jiaoyue Hu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Department of Ophthalmology of Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Zuguo Liu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Department of Ophthalmology of Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China; Department of Ophthalmology, the First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
17
|
Kumar P, Banik SP, Ohia SE, Moriyama H, Chakraborty S, Wang CK, Song YS, Goel A, Bagchi M, Bagchi D. Current Insights on the Photoprotective Mechanism of the Macular Carotenoids, Lutein and Zeaxanthin: Safety, Efficacy and Bio-Delivery. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024; 43:505-518. [PMID: 38393321 DOI: 10.1080/27697061.2024.2319090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/07/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024]
Abstract
Ocular health has emerged as one of the major issues of global health concern with a decline in quality of life in an aging population, in particular and rise in the number of associated morbidities and mortalities. One of the chief reasons for vision impairment is oxidative damage inflicted to photoreceptors in rods and cone cells by blue light as well as UV radiation. The scenario has been aggravated by unprecedented rise in screen-time during the COVID and post-COVID era. Lutein and Zeaxanthin are oxygenated carotenoids with proven roles in augmentation of ocular health largely by virtue of their antioxidant properties and protective effects against photobleaching of retinal pigments, age-linked macular degeneration, cataract, and retinitis pigmentosa. These molecules are characterized by their characteristic yellow-orange colored pigmentation and are found in significant amounts in vegetables such as corn, spinach, broccoli, carrots as well as fish and eggs. Unique structural signatures including tetraterpenoid skeleton with extensive conjugation and the presence of hydroxyl groups at the end rings have made these molecules evolutionarily adapted to localize in the membrane of the photoreceptor cells and prevent their free radical induced peroxidation. Apart from the benefits imparted to ocular health, lutein and zeaxanthin are also known to improve cognitive function, cardiovascular physiology, and arrest the development of malignancy. Although abundant in many natural sources, bioavailability of these compounds is low owing to their long aliphatic backbones. Under the circumstances, there has been a concerted effort to develop vegetable oil-based carriers such as lipid nano-emulsions for therapeutic administration of carotenoids. This review presents a comprehensive update of the therapeutic potential of the carotenoids along with the challenges in achieving an optimized delivery tool for maximizing their effectiveness inside the body.
Collapse
Affiliation(s)
- Pawan Kumar
- R&D Department, Chemical Resources (CHERESO), Panchkula, India
| | - Samudra P Banik
- Department of Microbiology, Maulana Azad College, Kolkata, India
| | - Sunny E Ohia
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Hiroyoshi Moriyama
- Department of Scientific Affairs, The Japanese Institute for Health Food Standards, Tokyo, Japan
| | - Sanjoy Chakraborty
- Department of Biological Sciences, New York City College of Technology/CUNY, Brooklyn, NY, USA
| | - Chin-Kun Wang
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Yong Sang Song
- Department of Obstetrics and Gynaecology, Seoul National University Hospital, Seoul, South Korea
| | - Apurva Goel
- Regulation Department, Chemical Resources (CHERESO), Panchkula, India
| | | | - Debasis Bagchi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
- Department of Biology, College of Arts and Sciences, and Department of Psychology, Gordon F. Derner School of Psychology, Adelphi University, Garden City, NY, USA
| |
Collapse
|
18
|
Zhao D, Zhao H, He Y, Zhang M. BMSC Alleviates Dry Eye by Inhibiting the ROS-NLRP3-IL-1β Signaling Axis by Reducing Inflammation Levels. Curr Eye Res 2024; 49:698-707. [PMID: 38450655 DOI: 10.1080/02713683.2024.2324434] [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: 08/17/2023] [Accepted: 02/24/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Bone marrow mesenchymal stem cells (BMSC) have multiple biological functions and are widely involved in regulating inflammatory diseases, tissue repair and regeneration. However, the mechanism of their action in dry eye disease (DED) is currently unclear. The purpose of this study was to investigate the effect of BMSCs in the treatment of dry eye mice and to explore its specific therapeutic mechanism. METHODS Mouse corneal epithelial cells (MCECs) were treated with 500 mOsM sodium chloride hypertonic solution to induce a DED cell model. The dry eye animal model was constructed by adding 5 μL 0.2% benzalkonium chloride solution to mouse eyes. Western blotting was used to detect the expression of related proteins, and flow cytometry, enzyme-linked immunosorbent assay (ELISA), terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining, hematoxylin-eosin (HE) staining, and periodic acid schiff (PAS) staining were used to detect cell and eye tissue damage. RESULTS The experimental results showed that BMSCs can reduce the levels of reactive oxygen species (ROS) and inflammatory factors in MCECs, promote cell proliferation, inhibit cell apoptosis, improve the integrity of the corneal epithelial layer in vivo, promote an increase in the number of goblet cells, and alleviate DED. Further exploration of the molecular mechanism of BMSCs treatment revealed that BMSCs alleviate the progression of DED by inhibiting the ROS-NLRP3-IL-1β signaling pathway. CONCLUSION BMSCs inhibit ROS-NLRP3-IL-1β signaling axis, reducing inflammation levels and alleviating dry eye symptoms. These findings provide new ideas and a basis for the treatment of DED and provide an experimental basis for further research on the application value of BMSCs in alleviating DED.
Collapse
Affiliation(s)
- Dandan Zhao
- Ophthalmology Department, Yan'An Hospital of Kunming City, Kunming, China
| | - Hongxia Zhao
- Ophthalmology Department, Yan'An Hospital of Kunming City, Kunming, China
| | - Yang He
- Ophthalmology Department, Yan'An Hospital of Kunming City, Kunming, China
| | - Meixia Zhang
- Ophthalmology Department, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Bu J, Liu Y, Zhang R, Lin S, Zhuang J, Sun L, Zhang L, He H, Zong R, Wu Y, Li W. Potential New Target for Dry Eye Disease-Oxidative Stress. Antioxidants (Basel) 2024; 13:422. [PMID: 38671870 PMCID: PMC11047456 DOI: 10.3390/antiox13040422] [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: 12/29/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Dry eye disease (DED) is a multifactorial condition affecting the ocular surface. It is characterized by loss of tear film homeostasis and accompanied by ocular symptoms that may potentially result in damage to the ocular surface and even vision loss. Unmodifiable risk factors for DED mainly include aging, hormonal changes, and lifestyle issues such as reduced sleep duration, increased screen exposure, smoking, and ethanol consumption. As its prevalence continues to rise, DED has garnered considerable attention, prompting the exploration of potential new therapeutic targets. Recent studies have found that when the production of ROS exceeds the capacity of the antioxidant defense system on the ocular surface, oxidative stress ensues, leading to cellular apoptosis and further oxidative damage. These events can exacerbate inflammation and cellular stress responses, further increasing ROS levels and promoting a vicious cycle of oxidative stress in DED. Therefore, given the central role of reactive oxygen species in the vicious cycle of inflammation in DED, strategies involving antioxidants have emerged as a novel approach for its treatment. This review aims to enhance our understanding of the intricate relationship between oxidative stress and DED, thereby providing directions to explore innovative therapeutic approaches for this complex ocular disorder.
Collapse
Affiliation(s)
- Jinghua Bu
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yanbo Liu
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Rongrong Zhang
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Sijie Lin
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Jingbin Zhuang
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Le Sun
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Lingyu Zhang
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Hui He
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Rongrong Zong
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yang Wu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| | - Wei Li
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
- Xiamen University Affiliated Xiamen Eye Center, Xiamen 361102, China
| |
Collapse
|
20
|
Liang C, Wang L, Wang X, Jia Y, Xie Q, Zhao L, Yuan H. Altered ocular surface microbiota in obesity: a case-control study. Front Cell Infect Microbiol 2024; 14:1356197. [PMID: 38533385 PMCID: PMC10963539 DOI: 10.3389/fcimb.2024.1356197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Purpose This study aimed to investigate the composition of ocular surface microbiota in patients with obesity. Methods This case-control study, spanning from November 2020 to March 2021 at Henan Provincial People's Hospital, involved 35 patients with obesity and an equivalent number of age and gender-matched healthy controls. By employing 16S rRNA sequencing, this study analyzed the differences in ocular surface microbiota between the two groups. The functional prediction analysis of the ocular surface microbiota was conducted using PICRUSt2. Results The alpha diversity showed no notable differences in the richness or evenness of the ocular surface microbiota when comparing patients with obesity to healthy controls (Shannon index, P=0.1003). However, beta diversity highlighted significant variances in the microbiota composition of these two groups (ANOSIM, P=0.005). LEfSe analysis revealed that the relative abundances of Delftia, Cutibacterium, Aquabacterium, Acidovorax, Caulobacteraceae unclassified, Comamonas and Porphyromonas in patients with obesity were significantly increased (P<0.05). Predictive analysis using PICRUSt2 highlighted a significant enhancement in certain metabolic pathways in patients with obesity, notably xenobiotics metabolism via cytochrome P450 (CYP450), lipid metabolism, and the oligomerization domain (NOD)-like receptor signaling pathway (P<0.05). Conclusions Patients with obesity exhibit a distinct ocular surface core microbiome. The observed variations in this microbiome may correlate with increased activity in CYP450, changes in lipid metabolism, and alterations in NOD-like receptor signaling pathways.
Collapse
Affiliation(s)
- Chenghong Liang
- Department of Endocrinology, Zhengzhou University People’s Hospital, Zhengzhou, China
- Department of Endocrinology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Limin Wang
- Department of Endocrinology, Zhengzhou University People’s Hospital, Zhengzhou, China
- Department of Endocrinology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiudan Wang
- Department of Endocrinology, Zhengzhou University People’s Hospital, Zhengzhou, China
- Department of Endocrinology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yifan Jia
- Department of Endocrinology, Zhengzhou University People’s Hospital, Zhengzhou, China
- Department of Endocrinology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Qinyuan Xie
- Department of Endocrinology, Zhengzhou University People’s Hospital, Zhengzhou, China
- Department of Endocrinology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lingyun Zhao
- Department of Endocrinology, Zhengzhou University People’s Hospital, Zhengzhou, China
- Department of Endocrinology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Huijuan Yuan
- Department of Endocrinology, Zhengzhou University People’s Hospital, Zhengzhou, China
- Department of Endocrinology, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
21
|
Ren Y, Wen H, Bai F, Huang B, Wang Z, Zhang S, Pu Y, Le Z, Gong X, Wang L, Chen W, Zheng Q. Comparison of deep learning-assisted blinking analysis system and Lipiview interferometer in dry eye patients: a cross-sectional study. EYE AND VISION (LONDON, ENGLAND) 2024; 11:7. [PMID: 38374153 PMCID: PMC10875838 DOI: 10.1186/s40662-024-00373-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 01/01/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Abnormal blinking pattern is associated with ocular surface diseases. However, blink is difficult to analyze due to the rapid movement of eyelids. Deep learning machine (DLM) has been proposed as an optional tool for blinking analysis, but its clinical practicability still needs to be proven. Therefore, the study aims to compare the DLM-assisted Keratograph 5M (K5M) as a novel method with the currently available Lipiview in the clinic and assess whether blinking parameters can be applied in the diagnosis of dry eye disease (DED). METHODS Thirty-five DED participants and 35 normal subjects were recruited in this cross-sectional study. DED questionnaire and ocular surface signs were evaluated. Blinking parameters including number of blinks, number of incomplete blinking (IB), and IB rate were collected from the blinking videos recorded by the K5M and Lipiview. Blinking parameters were individually collected from the DLM analyzed K5M videos and Lipiview generated results. The agreement and consistency of blinking parameters were compared between the two devices. The association of blinking parameters to DED symptoms and signs were evaluated via heatmap. RESULTS In total, 140 eyes of 70 participants were included in this study. Lipiview presented a higher number of IB and IB rate than those from DLM-assisted K5M (P ≤ 0.006). DLM-assisted K5M captured significant differences in number of blinks, number of IB and IB rate between DED and normal subjects (P ≤ 0.035). In all three parameters, DLM-assisted K5M also showed a better consistency in repeated measurements than Lipiview with higher intraclass correlation coefficients (number of blinks: 0.841 versus 0.665; number of IB: 0.750 versus 0.564; IB rate: 0.633 versus 0.589). More correlations between blinking parameters and DED symptoms and signs were found by DLM-assisted K5M. Moreover, the receiver operating characteristic analysis showed the number of IB from K5M exhibiting the highest area under curve of 0.773. CONCLUSIONS DLM-assisted K5M is a useful tool to analyze blinking videos and detect abnormal blinking patterns, especially in distinguishing DED patients from normal subjects. Large sample investigations are therefore warranted to assess its clinical utility before implementation.
Collapse
Affiliation(s)
- Yueping Ren
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Han Wen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Furong Bai
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Binge Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zhenzhen Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Shuwen Zhang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yaojia Pu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zhenmin Le
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xianhui Gong
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Lei Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Wei Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Qinxiang Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- The School of Ophthalmology and Optometry, Wenzhou Medical University, 270 Xueyuan West Road, Wenzhou, 325027, Zhejiang, People's Republic of China.
| |
Collapse
|
22
|
Korhonen E. Inflammasome activation in response to aberrations of cellular homeostasis in epithelial cells from human cornea and retina. Acta Ophthalmol 2024; 102 Suppl 281:3-68. [PMID: 38386419 DOI: 10.1111/aos.16646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 02/24/2024]
|
23
|
Huang B, Zhang N, Qiu X, Zeng R, Wang S, Hua M, Li Q, Nan K, Lin S. Mitochondria-targeted SkQ1 nanoparticles for dry eye disease: Inhibiting NLRP3 inflammasome activation by preventing mitochondrial DNA oxidation. J Control Release 2024; 365:1-15. [PMID: 37972763 DOI: 10.1016/j.jconrel.2023.11.021] [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: 08/21/2023] [Revised: 11/04/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023]
Abstract
Dry eye disease (DED) is a multifactorial ocular surface disorder mutually promoted by reactive oxygen species (ROS) and ocular surface inflammation. NLRP3 is the key regulator for inducing ocular surface inflammation in DED. However, the mechanism by which ROS influences the bio-effects of NLRP3, and the consequent development of DED, largely remains elusive. In the present study, we uncovered that robust ROS can oxidate mitochondrial DNA (ox-mtDNA) along with loss of mitochondria compaction causing the cytosolic release of ox-mtDNA and subsequent co-localization with cytosolic NLRP3, which can promote the activation of NLRP3 inflammasome and stimulate NLRP3-mediated inflammation. Visomitin (also known as SkQ1), a mitochondria-targeted anti-oxidant, could reverse such a process by in situ scavenging of mitochondrial ROS. To effectively deliver SkQ1, we further developed a novel mitochondria-targeted SkQ1 nanoparticle (SkQ1 NP) using a charge-driven self-assembly strategy. Compared with free SkQ1, SkQ1 NPs exhibited significantly higher cytosolic- and mitochondrial-ROS scavenging activity (1.7 and 1.9 times compared to levels of the free SkQ1 group), thus exerting a better in vitro protective effect against H2O2-induced cell death in human corneal epithelial cells (HCECs). After topical administration, SkQ1 NPs significantly reduced in vivo mtDNA oxidation, while suppressing the expressions of NLRP3, Caspase-1, and IL-1β, which consequently resulted in better therapeutic effects against DED. Results suggested that by efficiently scavenging mitochondrial ROS, SkQ1 NPs could in situ inhibit DED-induced mtDNA oxidation, thus blocking the interaction of ox-mtDNA and NLRP3; this, in turn, suppressed NLRP3 inflammasome activation and NLRP3-mediated inflammatory signaling. Results suggested that SkQ1 NPs have great potential as a new treatment for DED.
Collapse
Affiliation(s)
- Baoshan Huang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Na Zhang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; First Affiliated Hospital of Northwestern University, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Xi'an 710002, China
| | - Xinying Qiu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Rui Zeng
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Shuimiao Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Mengxia Hua
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Li
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| | - Kaihui Nan
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China.
| | - Sen Lin
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China; School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
24
|
Tian Y, Zhang Y, Zhao J, Luan F, Wang Y, Lai F, Ouyang D, Tao Y. Combining MSC Exosomes and Cerium Oxide Nanocrystals for Enhanced Dry Eye Syndrome Therapy. Pharmaceutics 2023; 15:2301. [PMID: 37765270 PMCID: PMC10536361 DOI: 10.3390/pharmaceutics15092301] [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: 07/22/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Dry eye syndrome (DES) is a prevalent ocular disorder involving diminishe·d tear production and increased tear evaporation, leading to ocular discomfort and potential surface damage. Inflammation and reactive oxygen species (ROS) have been implicated in the pathophysiology of DES. Inflammation is one core cause of the DES vicious cycle. Moreover, there are ROS that regulate inflammation in the cycle from the upstream, which leads to treatment failure in current therapies that merely target inflammation. In this study, we developed a novel therapeutic nanoparticle approach by growing cerium oxide (Ce) nanocrystals in situ on mesenchymal stem cell-derived exosomes (MSCExos), creating MSCExo-Ce. The combined properties of MSCExos and cerium oxide nanocrystals aim to target the "inflammation-ROS-injury" pathological mechanism in DES. We hypothesized that this approach would provide a new treatment option for patients with DES. Our analysis confirmed the successful in situ crystallization of cerium onto MSCExos, and MSCExo-Ce displayed excellent biocompatibility. In vitro and in vivo experiments have demonstrated that MSCExo-Ce promotes corneal cell growth, scavenges ROS, and more effectively suppresses inflammation compared with MSCExos alone. MSCExo-Ce also demonstrated the ability to alleviate DES symptoms and reverse pathological alterations at both the cellular and tissue levels. In conclusion, our findings highlight the potential of MSCExo-Ce as a promising therapeutic candidate for the treatment of DES.
Collapse
Affiliation(s)
- Ying Tian
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yiquan Zhang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiawei Zhao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Fuxiao Luan
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yingjie Wang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Fan Lai
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, Center for Life Science, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau 999078, China
| | - Yong Tao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
25
|
Das A, Sil A, Kumar P, Khan I. Blue light and skin: what is the intriguing link? Clin Exp Dermatol 2023; 48:968-977. [PMID: 37097168 DOI: 10.1093/ced/llad150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/20/2023] [Indexed: 04/26/2023]
Abstract
Blue light has garnered attention because of its ability to penetrate more deeply into the skin layers, and induce cellular dysfunction and DNA damage. Photoageing, hyperpigmentation and melasma are some of the cutaneous changes that develop on exposure to blue light. To date, the therapeutic roles of blue light have been evaluated in dermatological conditions like psoriasis, eczema, acne vulgaris, actinic keratosis and cutaneous malignancies, among others. In this review, we have attempted to present an evidence-based compilation of the effects of blue light on the skin.
Collapse
Affiliation(s)
- Anupam Das
- Department of Dermatology, KPC Medical College and Hospital, Kolkata, West Bengal, India
| | - Abheek Sil
- Department of Dermatology, Venereology and Leprosy, R.G. Kar Medical College and Hospital, Kolkata, West Bengal, India
| | - Piyush Kumar
- Department of Dermatology, Madhubani Medical College & Hospital, Madhubani, Bihar, India
| | - Ismat Khan
- Department of Dermatology, Medical College and Hospital Kolkata, Kolkata, West Bengal, India
| |
Collapse
|
26
|
Jiang D, Xu W, Peng F, Sun Y, Pan C, Yu J, Zheng Q, Chen W. Tunneling nanotubes-based intercellular mitochondrial trafficking as a novel therapeutic target in dry eye. Exp Eye Res 2023; 232:109497. [PMID: 37169281 DOI: 10.1016/j.exer.2023.109497] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/08/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
Cell-to-cell mitochondria transfer via tunneling nanotubes (TNTs) has recently been revealed as a spontaneous way to protect damaged cells. Previously, we have reported mesenchymal stem cells (MSCs) can rescue retinal ganglion cell and corneal epithelium through intercellular mitochondrial trafficking. Mitochondrial damage and oxidative stress in corneal epithelial cells are vital in dry eye disease (DED). However, whether intercellular mitochondrial transfer is involved in the pathological and repair process of DED is currently unknown. Therefore, in this study, we designed a coculture system to evaluate the role of intercellular mitochondrial transfer between human corneal epithelial cells (CEC) in DED. In addition, we successfully discovered the ROCK inhibitor, Y-27632 as an intensifier to improve the efficiency of intercellular mitochondrial transport. As expected, the enhanced mitochondrial transfer promotes the regeneration of CECs. Moreover, through further exploration of mechanisms, it was demonstrated that F-actin-mediated cell morphological changes and cytoskeletal remodeling may be potential mechanisms for Y-27632 to induce mitochondrial metastasis. In conclusion, we established a new method for cell repair in DED that healthy CEC offered mitochondria to damaged CEC, providing a new insight into the cellular mechanism of corneal epithelium homeostatic regenerative therapeutics in DED.
Collapse
Affiliation(s)
- Dan Jiang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wei Xu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Fangli Peng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yining Sun
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Chengjie Pan
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jinjie Yu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qinxiang Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Wei Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
27
|
Zhuang D, Misra SL, Mugisho OO, Rupenthal ID, Craig JP. NLRP3 Inflammasome as a Potential Therapeutic Target in Dry Eye Disease. Int J Mol Sci 2023; 24:10866. [PMID: 37446038 DOI: 10.3390/ijms241310866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Dry eye disease (DED) is a multifactorial ocular surface disorder arising from numerous interrelated underlying pathologies that trigger a self-perpetuating cycle of instability, hyperosmolarity, and ocular surface damage. Associated ocular discomfort and visual disturbance contribute negatively to quality of life. Ocular surface inflammation has been increasingly recognised as playing a key role in the pathophysiology of chronic DED. Current readily available anti-inflammatory agents successfully relieve symptoms, but often without addressing the underlying pathophysiological mechanism. The NOD-like receptor protein-3 (NLRP3) inflammasome pathway has recently been implicated as a key driver of ocular surface inflammation, as reported in pre-clinical and clinical studies of DED. This review discusses the intimate relationship between DED and inflammation, highlights the involvement of the inflammasome in the development of DED, describes existing anti-inflammatory therapies and their limitations, and evaluates the potential of the inflammasome in the context of the existing anti-inflammatory therapeutic landscape as a therapeutic target for effective treatment of the disease.
Collapse
Affiliation(s)
- Dian Zhuang
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Stuti L Misra
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Jennifer P Craig
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
28
|
Lepre CC, Russo M, Trotta MC, Petrillo F, D'Agostino FA, Gaudino G, D'Amico G, Campitiello MR, Crisci E, Nicoletti M, Gesualdo C, Simonelli F, D'Amico M, Hermenean A, Rossi S. Inhibition of Galectins and the P2X7 Purinergic Receptor as a Therapeutic Approach in the Neurovascular Inflammation of Diabetic Retinopathy. Int J Mol Sci 2023; 24:ijms24119721. [PMID: 37298672 DOI: 10.3390/ijms24119721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Diabetic retinopathy (DR) is the most frequent microvascular retinal complication of diabetic patients, contributing to loss of vision. Recently, retinal neuroinflammation and neurodegeneration have emerged as key players in DR progression, and therefore, this review examines the neuroinflammatory molecular basis of DR. We focus on four important aspects of retinal neuroinflammation: (i) the exacerbation of endoplasmic reticulum (ER) stress; (ii) the activation of the NLRP3 inflammasome; (iii) the role of galectins; and (iv) the activation of purinergic 2X7 receptor (P2X7R). Moreover, this review proposes the selective inhibition of galectins and the P2X7R as a potential pharmacological approach to prevent the progression of DR.
Collapse
Affiliation(s)
- Caterina Claudia Lepre
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania
| | - Marina Russo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Francesco Petrillo
- Ph.D. Course in Translational Medicine, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Fabiana Anna D'Agostino
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Gennaro Gaudino
- School of Anesthesia and Intensive Care, University of Foggia, 71122 Foggia, Italy
| | | | - Maria Rosaria Campitiello
- Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, 84124 Salerno, Italy
| | - Erminia Crisci
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Maddalena Nicoletti
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Carlo Gesualdo
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Francesca Simonelli
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Anca Hermenean
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania
| | - Settimio Rossi
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
29
|
Kang WS, Kim E, Choi H, Lee KH, Kim KJ, Lim D, Choi SY, Kim Y, Son SA, Kim JS, Kim S. Therapeutic Potential of Peucedanum japonicum Thunb. and Its Active Components in a Delayed Corneal Wound Healing Model Following Blue Light Irradiation-Induced Oxidative Stress. Antioxidants (Basel) 2023; 12:1171. [PMID: 37371901 DOI: 10.3390/antiox12061171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/22/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Blue light is reported to be harmful to eyes by inducing reactive oxygen species (ROS). Herein, the roles of Peucedanum japonicum Thunb. leaf extract (PJE) in corneal wound healing under blue light irradiation are investigated. Blue-light-irradiated human corneal epithelial cells (HCECs) show increased intracellular ROS levels and delayed wound healing without a change in survival, and these effects are reversed by PJE treatment. In acute toxicity tests, a single oral administration of PJE (5000 mg/kg) does not induce any signs of clinical toxicity or body weight changes for 15 days post-administration. Rats with OD (oculus dexter, right eye) corneal wounds are divided into seven treatment groups: NL (nonwounded OS (oculus sinister, left eye)), NR (wounded OD), BL (wounded OD + blue light (BL)), and PJE (BL + 25, 50, 100, 200 mg/kg). Blue-light-induced delayed wound healing is dose-dependently recovered by orally administering PJE once daily starting 5 days before wound generation. The reduced tear volume in both eyes in the BL group is also restored by PJE. Forty-eight hours after wound generation, the numbers of inflammatory and apoptotic cells and the expression levels of interleukin-6 (IL-6) largely increase in the BL group, but these values return to almost normal after PJE treatment. The key components of PJE, identified by high-performance liquid chromatography (HPLC) fractionation, are CA, neochlorogenic acid (NCA), and cryptochlorogenic acid (CCA). Each CA isomer effectively reverses the delayed wound healing and excessive ROS production, and their mixture synergistically enhances these effects. The expression of messenger RNAs (mRNAs) related to ROS, such as SOD1, CAT, GPX1, GSTM1, GSTP1, HO-1, and TRXR1, is significantly upregulated by PJE, its components, and the component mixture. Therefore, PJE protects against blue-light-induced delayed corneal wound healing via its antioxidative, anti-inflammatory, and antiapoptotic effects mechanistically related to ROS production.
Collapse
Affiliation(s)
- Wan Seok Kang
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Eun Kim
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Hakjoon Choi
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Ki Hoon Lee
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Kyeong Jo Kim
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Dosung Lim
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Su-Young Choi
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Youngbae Kim
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Seon Ah Son
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Jin Seok Kim
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| | - Sunoh Kim
- Central R&D Center, B&Tech Co., Ltd., Naju 58205, Republic of Korea
| |
Collapse
|
30
|
Li K, Gong Q, Lu B, Huang K, Tong Y, Mutsvene TE, Lin M, Xu Z, Lu F, Li X, Hu L. Anti-inflammatory and antioxidative effects of gallic acid on experimental dry eye: in vitro and in vivo studies. EYE AND VISION (LONDON, ENGLAND) 2023; 10:17. [PMID: 37122017 PMCID: PMC10150500 DOI: 10.1186/s40662-023-00334-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 02/22/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND To investigate the anti-inflammatory and antioxidative effects of gallic acid (GA) on human corneal epithelial cells (HCECs) and RAW264.7 macrophages as well as its therapeutic effects in an experimental dry eye (EDE) mouse model. METHODS A cell counting kit-8 (CCK-8) assay was used to test the cytotoxicity of GA. The effect of GA on cell migration was evaluated using a scratch wound healing assay. The anti-inflammatory and antioxidative effects of GA in vitro were tested using a hypertonic model (HCECs) and an inflammatory model (RAW264.7 cells). The in vivo biocompatibility of GA was detected by irritation tests in rabbits, whereas the preventive and therapeutic effect of GA in vivo was evaluated using a mouse model of EDE. RESULTS In the range of 0-100 μM, GA showed no cytotoxicity in RAW264.7 cells or HCECs and did not delay the HCECs monolayer wound healing within 24 h. Ocular tolerance to GA in the in vivo irritation test was good after seven days. In terms of antioxidative activity, GA significantly reduced the intracellular reactive oxygen species (ROS) in lipopolysaccharide (LPS) activated RAW264.7 macrophages and HCECs exposed to hyperosmotic stress. Furthermore, after pre-treatment with GA, the expression levels of nuclear factor E2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and NADPH quinone oxidoreductase-1 (NQO-1) were significantly upregulated in RAW264.7 macrophages. GA also exhibits excellent anti-inflammatory properties. This is mainly demonstrated by the ability of GA to effectively downregulate the nuclear transcription factor-κB (NF-κB) pathway in LPS-activated RAW264.7 macrophages and to reduce inflammatory factors, such as nitric oxide (NO), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α). In vivo efficacy testing results in a mouse model of EDE showed that GA can effectively prevent and inhibit the apoptosis of corneal epithelial cells (CECs), reduce inflammatory factors in the cornea and conjunctiva as well as protect goblet cells. CONCLUSION In vitro and in vivo results indicate that GA possesses potent anti-inflammatory and antioxidative properties with no apparent cytotoxicity within the range of 0-100 μM. It is a promising eye drop formulation for the effective prevention and treatment of dry eye disease (DED).
Collapse
Affiliation(s)
- Kexin Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Qianwen Gong
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Bin Lu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Kaiyan Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Yixuan Tong
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Tinashe Emmanuel Mutsvene
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Meng Lin
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Zhiqiang Xu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Fan Lu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
| | - Xingyi Li
- Institute of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, People's Republic of China.
| | - Liang Hu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
| |
Collapse
|
31
|
Zhang ZN, Yang DL, Liu H, Bi J, Bao YB, Ma JY, Zheng QX, Cui DL, Chen W, Xiang P. Effects of TCPP and TCEP exposure on human corneal epithelial cells: Oxidate damage, cell cycle arrest, and pyroptosis. CHEMOSPHERE 2023; 331:138817. [PMID: 37127200 DOI: 10.1016/j.chemosphere.2023.138817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/17/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
Tris(2-chloroisopropyl) phosphate (TCPP) and Tris(2-chloroethyl) phosphate (TCEP) are the widely used organophosphorus flame retardants indoors and easily accessible to the eyes as the common adhesive components of dust and particle matter, however, hardly any evidence has demonstrated their corneal toxicity. In this study, the adverse effects of TCPP, TCEP, and TCPP + TCEP exposure on human corneal epithelial cells (HCECs) were investigated. The cell viability and morphology, intracellular reactive oxygen species (ROS), cell cycle, and the expressions of cell cycle and pyroptosis-related genes were assessed to explain the underlying mechanisms. Compared to individual exposure, co-exposure to TCPP20+TCEP20 showed higher cytotoxicity with a sharp decrease of >30% in viability and more serious oxidative damage by increasing ROS production to 110.92% compared to the control group. Furthermore, the cell cycle arrested at the S phase (36.20%) was observed after combined treatment, evidenced by the upregulation of cyclin D1, CDK2, CDK4, CDK6, p21, and p27. Interestingly, pyroptosis-related genes GSDMD, Caspase-1, NLRP3, IL-1β, IL-18, NLRP1, and NLRC4 expressions were promoted with cell swelling and glowing morphology. Oxidative stress and cell cycle arrest probably acted as a key role in TCPP20+TCEP20-induced cytotoxicity and pyroptosis in HCECs. Our results suggested that TCPP20+TCEP20 co-exposure induced severer corneal damage, further illustrating its significance in estimating indoor health hazards to humans.
Collapse
Affiliation(s)
- Zhen-Ning Zhang
- Yunnan Province Innovative Research Team of Environmental Pollution, Food SafetyAnd Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, 650224, China
| | - Dan-Lei Yang
- Yunnan Province Innovative Research Team of Environmental Pollution, Food SafetyAnd Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, 650224, China
| | - Hai Liu
- Affiliated Hospital of Yunnan University, Eye Hospital of Yunnan Province, Kunming, 650224, China
| | - Jue Bi
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agriculture Sciences, Baoshan, 678000, China
| | - Ya-Bo Bao
- Yunnan Province Innovative Research Team of Environmental Pollution, Food SafetyAnd Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, 650224, China
| | - Jiao-Yang Ma
- Yunnan Province Innovative Research Team of Environmental Pollution, Food SafetyAnd Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, 650224, China
| | - Qin-Xiang Zheng
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, 315040, China
| | - Dao-Lei Cui
- Yunnan Province Innovative Research Team of Environmental Pollution, Food SafetyAnd Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, 650224, China
| | - Wei Chen
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, 315040, China.
| | - Ping Xiang
- Yunnan Province Innovative Research Team of Environmental Pollution, Food SafetyAnd Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, 650224, China.
| |
Collapse
|
32
|
Chen Y, Pu J, Li X, Lian L, Ge C, Liu Z, Wang W, Hou L, Chen W, Li J. Aim2 Deficiency Ameliorates Lacrimal Gland Destruction and Corneal Epithelium Defects in an Experimental Dry Eye Model. Invest Ophthalmol Vis Sci 2023; 64:26. [PMID: 36920364 PMCID: PMC10029764 DOI: 10.1167/iovs.64.3.26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Purpose Dry eye disease (DED) is a multifactorial disease that is associated with inflammation. Excessive DNA is present in the tear fluid of patients with DED. Absent in melanoma 2 (AIM2) is a key DNA sensor. This study aimed to investigate the role of AIM2 in the pathogenesis of DED. Methods DED was induced by injection of scopolamine (SCOP). Aberrant DNA was detected by cell-free DNA (cfDNA) ELISA and immunostaining. Corneal epithelial defects were assessed by corneal fluorescein staining, zonula occludens-1 immunostaining and TUNEL. Tear production was analyzed by phenol red thread test. Lacrimal gland (LG) histology was evaluated by hematoxylin and eosin staining, and transmission electron microscopy examination. Macrophage infiltration in LG was detected by immunohistochemistry for the macrophage marker F4/80. Gene expression was analyzed by RT-qPCR. Protein production was examined by immunoblot analysis or ELISA. Results Aim2-/- mice displayed a normal structure and function of LG and cornea under normal conditions. In SCOP-induced DED, wild type (WT) mice showed increased cfDNA in tear fluid, and aberrant accumulations of dsDNA accompanied by increased AIM2 expression in the LG. In SCOP-induced DED, WT mice displayed damaged structures of LG, reduced tear production, and severe corneal epithelium defects, whereas Aim2-/- mice had a better preserved LG structure, less decreased tear production, and improved clinical signs of dry eye. Furthermore, genetic deletion of Aim2 suppressed the increased infiltration of macrophages and inhibited N-GSDMD and IL18 production in the LG of SCOP-induced DED. Conclusions Aim2 deficiency alleviates ocular surface damage and LG inflammation in SCOP-induced DED.
Collapse
Affiliation(s)
- Yu Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiheng Pu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Ophthalmology, The East Beijing Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xinda Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lili Lian
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chaoxiang Ge
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zuimeng Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Weizhuo Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ling Hou
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jinyang Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
33
|
Martinez-Carrasco R, Fini ME. Dynasore Protects Corneal Epithelial Cells Subjected to Hyperosmolar Stress in an In Vitro Model of Dry Eye Epitheliopathy. Int J Mol Sci 2023; 24:ijms24054754. [PMID: 36902183 PMCID: PMC10003680 DOI: 10.3390/ijms24054754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Epitheliopathy at the ocular surface is a defining sign of dry eye disease, a common disorder that affects 10% to 30% of the world's population. Hyperosmolarity of the tear film is one of the main drivers of pathology, with subsequent endoplasmic reticulum (ER) stress, the resulting unfolded protein response (UPR), and caspase-3 activation implicated in the pathway to programmed cell death. Dynasore, is a small molecule inhibitor of dynamin GTPases that has shown therapeutic effects in a variety of disease models involving oxidative stress. Recently we showed that dynasore protects corneal epithelial cells exposed to the oxidant tBHP, by selective reduction in expression of CHOP, a marker of the UPR PERK branch. Here we investigated the capacity of dynasore to protect corneal epithelial cells subjected to hyperosmotic stress (HOS). Similar to dynasore's capacity to protect against tBHP exposure, dynasore inhibits the cell death pathway triggered by HOS, protecting against ER stress and maintaining a homeostatic level of UPR activity. However, unlike with tBHP exposure, UPR activation due to HOS is independent of PERK and mostly driven by the UPR IRE1 branch. Our results demonstrate the role of the UPR in HOS-driven damage, and the potential of dynasore as a treatment to prevent dry eye epitheliopathy.
Collapse
Affiliation(s)
- Rafael Martinez-Carrasco
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA
- Correspondence: (R.M.-C.); (M.E.F.)
| | - M. Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Correspondence: (R.M.-C.); (M.E.F.)
| |
Collapse
|
34
|
Lou Q, Pan L, Xiang S, Li Y, Jin J, Tan J, Huang B, Nan K, Lin S. Suppression of NLRP3/Caspase-1/GSDMD Mediated Corneal Epithelium Pyroptosis Using Melatonin-Loaded Liposomes to Inhibit Benzalkonium Chloride-Induced Dry Eye Disease. Int J Nanomedicine 2023; 18:2447-2463. [PMID: 37192892 PMCID: PMC10182801 DOI: 10.2147/ijn.s403337] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/03/2023] [Indexed: 05/18/2023] Open
Abstract
Introduction Benzalkonium chloride (BAC) is widely employed as a preservative in eye drops, which will cause the death of corneal epithelial cells due to ROS production, DNA strand breakage, and mitochondrial dysfunction, resulting in dry eye disease (DED)-like changes in ocular surface tissues. In this study, Melatonin (MT) liposomes (TAT-MT-LIPs) designed by loading MT into TAT-modified liposomes have been developed, characterized, and used for inhibiting BAC-induced DED (BAC-DED). Methods The TAT was chemically grafted onto the Mal-PEG2000-DSPE by Michael's addition between the sulfhydryl group in TAT and the maleimide group in Mal-PEG2000-DSPE. TAT-MT-LIPs were prepared using film dispersion followed by the extrusion method and topically treated in rats once a day. BAC-DED was induced in rats by topical administration with 0.2% BAC twice daily. Defects, edema, and inflammation of the corneas, as well as IOP, were examined. Histologic analyses of corneas were performed to assess the change of mitochondrial DNA oxidation and NLRP3/Caspase-1/GSDMD signaling transduction. Results After topical administration, TAT-MT-LIPs significantly alleviated DED-clinical symptoms of experimental animals by inhibiting tissue inflammation and preventing the loss of the corneal epithelium and conjunctival goblet cells. Our data suggested continuous ocular surface exposure of BAC-induced NLRP3/Caspase-1/GSDMD mediated corneal epithelium pyroptosis, which was not reported before. BAC caused substantial mt-DNA oxidation, which promoted the transduction of NLRP3/Caspase-1/GSDMD and consequent corneal epithelium pyroptosis. TAT-MT-LIPs could efficiently suppress the BAC-induced corneal epithelium pyroptosis and inflammation by inhibiting mt-DNA oxidation and the subsequent signal transmission. Conclusion NLRP3/Caspase-1/GSDMD mediated corneal epithelium pyroptosis is involved in the development of BAC-DED. The present study provided new insights into the adverse effects of BAC, which can serve as a new target for protecting corneal epithelium when applying BAC as a preservative in eye drops. The developed TAT-MT-LIPs can efficiently inhibit BAC-DED and give great potential to be developed as a new DED treatment.
Collapse
Affiliation(s)
- Qi Lou
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Lu Pan
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Shengjin Xiang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Yueting Li
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Jiahui Jin
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Jingyang Tan
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Baoshan Huang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Kaihui Nan
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
- Correspondence: Kaihui Nan; Sen Lin, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China, Tel +86-577-88067962, Email ;
| | - Sen Lin
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| |
Collapse
|
35
|
Serrano A, Osei KA, Huertas-Bello M, Sabater AL. The Potential of Stem Cells as Treatment for Ocular Surface Diseases. CURRENT OPHTHALMOLOGY REPORTS 2022. [DOI: 10.1007/s40135-022-00303-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
36
|
Peng F, Jiang D, Xu W, Sun Y, Zha Z, Tan X, Yu J, Pan C, Zheng Q, Chen W. AMPK/MFF Activation: Role in Mitochondrial Fission and Mitophagy in Dry Eye. Invest Ophthalmol Vis Sci 2022; 63:18. [PMID: 36374514 PMCID: PMC9669805 DOI: 10.1167/iovs.63.12.18] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose To assess the role of mitochondrial morphology and adenosine monophosphate-activated protein kinase (AMPK)/mitochondrial fission factor (MFF) in dry eye and the underlying mechanisms. Methods Immortalized human corneal epithelial cells (HCECs) and primary HCECs were cultured under high osmotic pressure (HOP). C57BL/6 female mice were injected subcutaneously with scopolamine. Quantitative real-time PCR was used to measure mRNA expression. Protein expression was assessed by western blot and immunofluorescence staining. Mitochondrial morphology was observed by confocal microscopy and transmission electron microscopy. Results First, HOP induced mitochondrial oxidative damage to HCECs, accompanied by mitochondrial fission and increased mitophagy. Then, AMPK/MFF pathway proteins were increased consequent to HOP-induced energy metabolism dysfunction. Interestingly, the AMPK pathway promoted mitochondrial fission and mitophagy by increasing the recruitment of dynamin-related protein 1 (DRP1) to the mitochondrial outer membrane in the HOP group. Moreover, AMPK knockdown attenuated mitochondrial fission and mitophagy due to HOP in HCECs. AMPK activation triggered mitochondrial fission and mitophagy. Mitochondrial fission of HCECs stressed by HOP was mediated via MFF phosphorylation. MFF knockdown reversed mitochondrial fragmentation and mitophagy in HCECs treated with HOP. Inhibition of MFF protected HCECs against oxidative damage, cell death, and inflammation in the presence of HOP. Finally, we detected mitochondrial fission and AMPK pathway activation in vivo. Conclusions The AMPK/MFF pathway mediates the development of dry eye by positively regulating mitochondrial fission and mitophagy. Inhibition of mitochondrial fission can alleviate oxidative damage and inflammation in dry eye and may provide experimental evidence for treating dry eye.
Collapse
Affiliation(s)
- Fangli Peng
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dan Jiang
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Xu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yining Sun
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiwei Zha
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiying Tan
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinjie Yu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengjie Pan
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qinxiang Zheng
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
37
|
Li X, Jin X, Wang J, Li X, Zhang H. Dexamethasone attenuates dry eye-induced pyroptosis by regulating the KCNQ1OT1/miR-214 cascade. Steroids 2022; 186:109073. [PMID: 35779698 DOI: 10.1016/j.steroids.2022.109073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 10/17/2022]
Abstract
Dry eye disease (DED) is an inflammatory disorder of the ocular surface seriously affecting the quality of life of patients. Topical dexamethasone (Dex) administration protects the cornea from the hyperosmotic stress (HS) induced by tears. Pyroptosis participates in the activation of epithelial inflammation during DED. However, it remains unclear whether Dex attenuates the progression of DED through pyroptosis. In this study, we aimed to investigate the effect of Dex on DED using both cell and animal models and its underlying mechanism. The inflammatory factors contained in tears were detected using a cytokine assay. The pyroptosis in DED mice and human corneal epithelial cells (HCECs) treated with hyperosmotic medium under various treatments was evaluated by immunohistochemical assays (IHC) or western blotting (WB). RNA expression was manipulated with siRNA or agomir microRNAs and measured using a polymerase chain reaction. The scratch assay was used to assess the migration rate of HCECs. Remaining corneal defects were evaluated using fluorescein staining and photographed using a digital camera. Dex could suppress the release of inflammatory factors and notably attenuate pyroptosis, KCNQ1OT1 expression, and NF-κB activation induced by HS injury in vivo and in vitro. KCNQ1OT1 upregulation could activate pyroptosis by sponging miR-214. Furthermore, KCNQ1OT1 knockdown and miR-214 overexpression reversed the effect of HS, promoted the migration of HCECs, and accelerated corneal wound healing. Dex effectively suppressed HS-induced pyroptosis through the KCNQ1OT1/miR-214/caspase-1 signaling axis by inhibiting the NF-κB activation. Our results provide a novel understanding of the mechanism of Dex as an anti-inflammatory drug in DED.
Collapse
Affiliation(s)
- Xuran Li
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Xin Jin
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Jingrao Wang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Xinyue Li
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China
| | - Hong Zhang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Basic and Clinical Research of Heilongjiang Province, Harbin, China.
| |
Collapse
|
38
|
Chen M, Rong R, Xia X. Spotlight on pyroptosis: role in pathogenesis and therapeutic potential of ocular diseases. J Neuroinflammation 2022; 19:183. [PMID: 35836195 PMCID: PMC9281180 DOI: 10.1186/s12974-022-02547-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
Pyroptosis is a programmed cell death characterized by swift plasma membrane disruption and subsequent release of cellular contents and pro-inflammatory mediators (cytokines), including IL‐1β and IL‐18. It differs from other types of programmed cell death such as apoptosis, autophagy, necroptosis, ferroptosis, and NETosis in terms of its morphology and mechanism. As a recently discovered form of cell death, pyroptosis has been demonstrated to be involved in the progression of multiple diseases. Recent studies have also suggested that pyroptosis is linked to various ocular diseases. In this review, we systematically summarized and discussed recent scientific discoveries of the involvement of pyroptosis in common ocular diseases, including diabetic retinopathy, age-related macular degeneration, AIDS-related human cytomegalovirus retinitis, glaucoma, dry eye disease, keratitis, uveitis, and cataract. We also organized new and emerging evidence suggesting that pyroptosis signaling pathways may be potential therapeutic targets in ocular diseases, hoping to provide a summary of overall intervention strategies and relevant multi-dimensional evaluations for various ocular diseases, as well as offer valuable ideas for further research and development from the perspective of pyroptosis.
Collapse
Affiliation(s)
- Meini Chen
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,Hunan Key Laboratory of Ophthalmology, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Changsha, 410008, Hunan, People's Republic of China
| | - Rong Rong
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,Hunan Key Laboratory of Ophthalmology, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Changsha, 410008, Hunan, People's Republic of China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China. .,Hunan Key Laboratory of Ophthalmology, Changsha, 410008, Hunan, People's Republic of China. .,National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
39
|
Zha Z, Chen Q, Xiao D, Pan C, Xu W, Shen L, Shen J, Chen W. Mussel-Inspired Microgel Encapsulated NLRP3 Inhibitor as a Synergistic Strategy Against Dry Eye. Front Bioeng Biotechnol 2022; 10:913648. [PMID: 35721850 PMCID: PMC9198461 DOI: 10.3389/fbioe.2022.913648] [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] [Received: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
The inflammatory response mediated by oxidative stress is the main pathogenesis of dry eye, but clinical observations have shown that scavenging oxygen-free radicals alone has limited therapeutic effect. Moreover, the unique anatomy and physiology of the ocular surface result in low bioavailability of drugs, and higher concentration is required to achieve the desired efficacy, which, however, may bring systemic side effects. These problems pose a challenge, but the revelation of the ROS-NLRP3-IL-1β signaling axis opens up new possibilities. In this investigation, an NLRP3 inhibitor was successfully encapsulated in polydopamine-based microgels and used for dry eye treatment. It was demonstrated that the well-designed microgels exhibited good biocompatibility, prolonged drug retention time on the ocular surface, and effective inhibition of corneal epithelial damage and cell apoptosis. In addition, due to the synergistic effect, the NLRP3 inhibitor–loaded microgels could exert enhanced oxygen radical scavenging and inflammation-inhibiting effects at a lower dose than monotherapy. These findings suggest that polydopamine-based microgels have advantages as ocular surface drug delivery platforms and have promising applications in oxidative damage–related inflammatory diseases in synergy with anti-inflammatory drugs.
Collapse
Affiliation(s)
- Zhiwei Zha
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Qiumeng Chen
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Decheng Xiao
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Chengjie Pan
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Wei Xu
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Liangliang Shen
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Liangliang Shen, ; Jianliang Shen, ; Wei Chen,
| | - Jianliang Shen
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China
- *Correspondence: Liangliang Shen, ; Jianliang Shen, ; Wei Chen,
| | - Wei Chen
- Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Liangliang Shen, ; Jianliang Shen, ; Wei Chen,
| |
Collapse
|
40
|
Li Q, Hua X, Li L, Zhou X, Tian Y, Deng Y, Zhang M, Yuan X, Chi W. AIP1 suppresses neovascularization by inhibiting the NOX4-induced NLRP3/NLRP6 imbalance in a murine corneal alkali burn model. Cell Commun Signal 2022; 20:59. [PMID: 35524333 PMCID: PMC9074213 DOI: 10.1186/s12964-022-00877-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/07/2022] [Indexed: 12/05/2022] Open
Abstract
Background Apoptosis signal-regulating kinase 1-interacting protein 1 (AIP1) participates in inflammatory neovascularization induction. NADPH oxidase 4 (NOX4) produces reactive oxygen species (ROS), leading to an imbalance in nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) and NLR family pyrin domain containing 6 (NLRP6) expression. The mechanisms of AIP1, NOX4, ROS and inflammasomes in corneal neovascularization were studied herein. Methods C57BL/6 and AIP1-knockout mice were used in this study. The alkali burn procedure was performed on the right eye. Adenovirus encoding AIP1 plus green fluorescence protein (GFP) (Ad-AIP1-GFP) or GFP alone was injected into the right anterior chamber, GLX351322 was applied as a NOX4 inhibitor, and then corneal neovascularization was scored. The expression of related genes was measured by quantitative real-time polymerase chain reaction, western blotting and immunofluorescence staining. 2′,7′-Dichlorofluorescin diacetate staining was used to determine the ROS levels. Results The expression of AIP1 was decreased, while that of cleaved interleukin-1β (clv-IL-1β) and vascular endothelial growth factor A (VEGFa) was increased after alkali burn injury. NOX4 expression was increased, the imbalance in NLRP3/NLRP6 was exacerbated, and corneal neovascularization was increased significantly in AIP1-knockout mice compared with those in C57BL/6 mice after alkali burns. These effects were reversed by AIP1 overexpression. NLRP3/NLRP6 expression was imbalanced after alkali burns. GLX351322 reversed the imbalance in NLRP3/NLRP6 by reducing the ROS levels. This treatment also reduced the expression of clv-IL-1β and VEGFa, suppressing neovascularization. Conclusions AIP1 and NOX4 can regulate corneal inflammation and neovascularization after alkali burn injury. Based on the pathogenesis of corneal neovascularization, these findings are expected to provide new therapeutic strategies for patients. Plain English summary Corneal alkali burn injury is a common type of ocular injury that is difficult to treat in the clinic. The cornea is a clear and avascular tissue. Corneal neovascularization after alkali burn injury is a serious complication; it not only seriously affects the patient’s vision but also is the main reason for failed corneal transplantation. Corneal neovascularization affects approximately 1.4 million patients a year. We show for the first time that AIP1 and NOX4 can regulate corneal inflammation and neovascularization after alkali burns. The expression of AIP1 was decreased, while that of clv-IL-1β and VEGFa was increased after alkali burns. We tried to elucidate the specific molecular mechanisms by which AIP1 regulates corneal neovascularization. NOX4 activation was due to decreased AIP1 expression in murine corneas with alkali burns. NOX4 expression was increased, the imbalance in NLRP3/NLRP6 was exacerbated, and corneal neovascularization was increased significantly in AIP1-knockout mice compared with those in C57BL/6 mice after alkali burns. These effects were reversed by AIP1 overexpression. Additionally, NLRP3/NLRP6 expression was unbalanced, with NLRP3 activation and NLRP6 suppression in the corneal alkali burn murine model. Eye drops containing GLX351322, a NOX4 inhibitor, reversed the imbalance in NLRP3/NLRP6 by reducing ROS expression. This treatment also reduced the expression of clv-IL-1β and VEGFa, reducing neovascularization. Therefore, we provide new gene therapeutic strategies for patients. With the development of neovascularization therapy, we believe that in addition to corneal transplantation, new drug or gene therapies can achieve better results. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00877-5.
Collapse
Affiliation(s)
- Qingyu Li
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Xia Hua
- Tianjin Aier Eye Hospital, Tianjin University, Tianjin, China
| | - Liangpin Li
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Xueyan Zhou
- School of Medicine, Nankai University, Tianjin, China
| | - Ye Tian
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Yang Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Min Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xiaoyong Yuan
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China. .,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China.
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
41
|
Stuard WL, Guner MK, Robertson DM. IGFBP-3 Regulates Mitochondrial Hyperfusion and Metabolic Activity in Ocular Surface Epithelia during Hyperosmolar Stress. Int J Mol Sci 2022; 23:4066. [PMID: 35409425 PMCID: PMC9000157 DOI: 10.3390/ijms23074066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
In the eye, hyperosmolarity of the precorneal tear film triggers inflammation and the development of dry eye disease (DED), a highly prevalent condition that causes depression and disability in severe forms. A member of the insulin-like growth factor (IGF) family, the IGF binding protein-3 (IGFBP-3), is a pleiotropic protein with known roles in growth downregulation and survival. IGFBP-3 exerts these effects by blocking IGF-1 activation of the type 1 IGF-receptor (IGF-1R). Here, we examined a new IGF-independent role for IGFBP-3 in the regulation of mitochondrial and metabolic activity in ocular surface epithelial cells subject to hyperosmolar stress and in a mouse model of DED. We found that hyperosmolar stress decreased IGFBP-3 expression in vitro and in vivo. Treatment with exogenous IGFBP-3 induced an early, transient shift in IGF-1R to mitochondria, followed by IGFBP-3 nuclear accumulation. IGFBP-3 nuclear accumulation increased protein translation, blocked the hyperosmolar-mediated decrease in oxidative phosphorylation through the induction of mitochondrial hyperfusion, and restored corneal health in vivo. These data indicate that IGFBP-3 acts a stress response protein in ocular surface epithelia subject to hyperosmolar stress. These findings may lead to the development of first-in-class therapeutics to treat eye diseases with underlying mitochondrial dysfunction.
Collapse
Affiliation(s)
- Whitney L Stuard
- Department of Ophthalmology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Melis K Guner
- Department of Ophthalmology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Danielle M Robertson
- Department of Ophthalmology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| |
Collapse
|
42
|
Ren Y, Feng J, Lin Y, Reinach PS, Liu Y, Xia X, Ma X, Chen W, Zheng Q. MiR-223 inhibits hyperosmolarity-induced inflammation through downregulating NLRP3 activation in human corneal epithelial cells and dry eye patients. Exp Eye Res 2022; 220:109096. [DOI: 10.1016/j.exer.2022.109096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/29/2022] [Accepted: 04/21/2022] [Indexed: 01/10/2023]
|
43
|
Xiao K, Shang J, Liu Y, Chen Z, Wang L, Long Q. Effect of NLRP3 repression on NLRP3 inflammasome activation in human corneal epithelial cells with black carbon exposure. Cutan Ocul Toxicol 2022; 41:107-112. [PMID: 35298317 DOI: 10.1080/15569527.2022.2050746] [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
PURPOSE To investigate the inhibitory effects of NLRP3 siRNA on NLRP3 inflammasome activation in human corneal epithelial cells (HCECs) with fresh black carbon (FBC) particles and ozone-oxidized BC (OBC) particles treatment. METHODS HCECs were transfected with NLRP3 siRNA or control siRNA for 48 h, followed by 200 μg/ml FBC or OBC suspension for an additional 72 h. Untreated controls were cells with no siRNA transfection or BC treatment. RT-qPCR and Western blot were used to measure mRNA and protein levels of components of the NLRP3 inflammasome (NLRP3, ASC, and Caspase-1) and downstream cytokine (IL-1β), respectively. RESULTS Compared with untreated control cells, mRNA levels of NLRP3, ASC, Caspase-1, and IL-1β were significantly higher (p < 0.05) in control siRNA transfected cells with BC treatments. Compared with the control siRNA transfected cells, NLRP3 siRNA transfection reduced the expression of NLRP3 and ASC, whereas it had a limited effect on the expression of Caspase-1 and IL-1β with FBC or OBC exposures. Under FBC treatment, the reductions of NLRP3 and Caspase-1 mRNA levels were 53.5% (p < 0.001) and 34.2% (p < 0. 01), respectively, and NLRP3 and ASC protein levels were lowered by 58.2% (p < 0.001) and 45.4% (p < 0.001), respectively. Under OBC treatment, the reductions of NLRP3 and Caspase-1 mRNA levels were 39.8% (p < 0.001) and 25.6% (p < 0.05), respectively, and NLRP3 and ASC protein levels were lowered by 44.8% (p < 0.001) and 41.7% (p < 0.001), respectively. Moreover, mRNA levels of ASC and IL-1β, the protein levels of Caspase-1 and IL-1β showed a tendency to decrease in NLRP3 siRNA transfected cells, it was statistically insignificant (p > 0.05). CONCLUSIONS NLRP3 siRNA transfection could partially reverse the increased mRNA levels of NLRP3 and Caspase-1, the protein levels of NLRP3 and ASC in HCECs with BC treatment, whereas the reductions of protein levels of Caspase-1 and IL-1β were not significant, indicating that NLRP3 siRNA has a limited inhibitory effect on the activation of NLRP3 inflammasome triggered by BC.
Collapse
Affiliation(s)
- Kang Xiao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Shang
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing, China
| | - Ying Liu
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhengyu Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Liqiang Wang
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Qin Long
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Zheng Q, Wang L, Wen H, Ren Y, Huang S, Bai F, Li N, Craig JP, Tong L, Chen W. Impact of Incomplete Blinking Analyzed Using a Deep Learning Model With the Keratograph 5M in Dry Eye Disease. Transl Vis Sci Technol 2022; 11:38. [PMID: 35357395 PMCID: PMC8976934 DOI: 10.1167/tvst.11.3.38] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To establish a deep learning model (DLM) for blink analysis, and investigate whether blink video frame sampling rate influences the accuracy of analysis. Methods This case-controlled study recruited 50 dry eye disease (DED) participants and 50 normal subjects. Blink videos recorded by a Keratograph 5M, symptom questionnaires, and ocular surface assessments were collected. After processing the blink images as datasets, further training and evaluation of DLM was performed. Blink videos of 30 frames per second (FPS) under white light, eight FPS extracted from white light videos, and eight FPS under infrared light were processed by DLM to generate blink profiles, allowing comparison of blink parameters, and their association with DED symptoms and signs. Results The blink parameters based on 30 FPS video presented higher sensitivity and accuracy than those based on eight FPS. The average relative interpalpebral height (IPH), the frequency and proportion of incomplete blinking (IB) were much higher in DED participants than in normal controls (P < 0.001). The IB frequency was closely associated with DED symptoms and signs (|R| ≥ 0.195, P ≤ 0.048), as was IB proportion and the average IPH (R ≥ 0.202, P ≤ 0.042). Conclusions DLM is a powerful tool for analyzing blink videos with high accuracy and sensitivity, and a frame rate ≥ 30 FPS is recommended. The IB frequency is indicative of DED. Translational Relevance The system of DLM-based blink analysis is of great potential for the assessment of IB and diagnosis of DED.
Collapse
Affiliation(s)
- Qinxiang Zheng
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Lei Wang
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Han Wen
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Yueping Ren
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Shenghai Huang
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Furong Bai
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Na Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Jennifer P Craig
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| | - Louis Tong
- Singapore Eye Research Institute, Singapore; Singapore National Eye Centre, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wei Chen
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China; National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| |
Collapse
|
45
|
Di Zazzo A, Coassin M, Surico PL, Bonini S. Age-related ocular surface failure: A narrative review. Exp Eye Res 2022; 219:109035. [DOI: 10.1016/j.exer.2022.109035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/28/2022] [Accepted: 03/13/2022] [Indexed: 12/26/2022]
|
46
|
Thompson B, Davidson EA, Chen Y, Orlicky DJ, Thompson DC, Vasiliou V. Oxidative stress induces inflammation of lens cells and triggers immune surveillance of ocular tissues. Chem Biol Interact 2022; 355:109804. [PMID: 35123994 PMCID: PMC9136680 DOI: 10.1016/j.cbi.2022.109804] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/24/2021] [Accepted: 01/05/2022] [Indexed: 11/03/2022]
Abstract
Recent reports have challenged the notion that the lens is immune-privileged. However, these studies have not fully identified the molecular mechanism(s) that promote immune surveillance of the lens. Using a mouse model of targeted glutathione (GSH) deficiency in ocular surface tissues, we have investigated the role of oxidative stress in upregulating cytokine expression and promoting immune surveillance of the eye. RNA-sequencing of lenses from postnatal day (P) 1-aged Gclcf/f;Le-CreTg/- (KO) and Gclcf/f;Le-Cre-/- control (CON) mice revealed upregulation of many cytokines (e.g., CCL4, GDF15, CSF1) and immune response genes in the lenses of KO mice. The eyes of KO mice had a greater number of cells in the aqueous and vitreous humors at P1, P20 and P50 than age-matched CON and Gclcw/w;Le-CreTg/- (CRE) mice. Histological analyses revealed the presence of innate immune cells (i.e., macrophages, leukocytes) in ocular structures of the KO mice. At P20, the expression of cytokines and ROS content was higher in the lenses of KO mice than in those from age-matched CRE and CON mice, suggesting that oxidative stress may induce cytokine expression. In vitro administration of the oxidant, hydrogen peroxide, and the depletion of GSH (using buthionine sulfoximine (BSO)) in 21EM15 lens epithelial cells induced cytokine expression, an effect that was prevented by co-treatment of the cells with N-acetyl-l-cysteine (NAC), a antioxidant. The in vivo and ex vivo induction of cytokine expression by oxidative stress was associated with the expression of markers of epithelial-to-mesenchymal transition (EMT), α-SMA, in lens cells. Given that EMT of lens epithelial cells causes posterior capsule opacification (PCO), we propose that oxidative stress induces cytokine expression, EMT and the development of PCO in a positive feedback loop. Collectively these data indicate that oxidative stress induces inflammation of lens cells which promotes immune surveillance of ocular structures.
Collapse
Affiliation(s)
- Brian Thompson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA
| | - Emily A Davidson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA
| | - David J Orlicky
- Department of Pathology, Anschutz School of Medicine, University of Colorado, Aurora, CO, USA
| | - David C Thompson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA; Department of Clinical Pharmacy, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA.
| |
Collapse
|
47
|
Xu J, Chen P, Zhao G, Wei S, Li Q, Guo C, Cao Q, Wu X, Di G. Copolymer micelle-administered melatonin ameliorates hyperosmolarity-induced ocular surface damage through regulating PINK1 mediated mitophagy. Curr Eye Res 2022; 47:688-703. [PMID: 35179400 DOI: 10.1080/02713683.2021.2022163] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate the role and mechanism of melatonin-loaded polymer polyvinyl caprolactam-polyvinyl acetate-polyethyleneglycol graft copolymer (PVCL-PVA-PEG) micelles (Mel-Mic) in dry eye disease (DED). METHODS In vitro, the apoptosis and reactive oxygen species (ROS) generation in HCECs were analyzed by immunostaining and flow cytometry (FCM). The effect of Mel-Mic on autophagy and mitophagy was evaluated by immunostaining and western blots. PINK1 knockdown was analyzed by small interfering RNA (siRNA). In vivo, sodium fluorescein staining, tear secretion test, and periodic acid-schiff (PAS) staining were used to determine whether Mel-Mic can alleviate the severity of DED. Small molecule antagonists were pretreated to investigate whether melatonin type 1 and/or 2 receptors (MT1/MT2) mediate the effects of Mel-Mic. RESULTS Mel-Mic improved the solubility and biological activities of Mel in aqueous solutions. Treatment with Mel-Mic decreased the apoptosis of HCECs exposed to hyperosmotic medium, accompanied by downregulation of cleaved Caspase-3 and upregulation of Bcl-2. In addition, Mel-Mic application suppressed ROS overproduction, rescued mitochondrial function, and decreased the level of oxidative stress associated biomarkers (COX-2 and 4-HNE) in HCECs. Interestingly, HCECs treated with Mel-Mic exhibited increased levels of mitophagy markers (PINK1, PARKIN, Beclin 1 and LC3B) and restored impaired mitophagic flux under hyperosmolarity. While PINK1 knock down largely abolished its protective effects. In vivo, compared to vehicle group, topical Mel-Mic solution treated mice showed significantly improved clinical parameters, increased tear production and decreased goblet cells loss in a dose-dependent manner. Also, TEM assay revealed increased autophagosome number in the corneal epithelium of Mel-Mic group. Moreover, luzindole, a non-selective MT1/MT2 antagonist, but not 4-P-PDOT, a selective MT2 antagonist, blocked the protective effect of Mel-Mic. CONCLUSIONS Our findings demonstrated that Mel-Mic ameliorates hyperosmolarity induced ocular surface damage via PINK1 mediated mitophagy and may represent an effective treatment for DED possibly through acting MT1 receptor.
Collapse
Affiliation(s)
- Jing Xu
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, China
| | - Peng Chen
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, China
| | - Guangfen Zhao
- Department of Medicine, The Liaocheng Third People's Hospital. Liaocheng, China
| | - Susu Wei
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, China
| | - Qiqi Li
- College of Chemical Engineering, Qingdao University of Science and Technology. Qingdao, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology. Qingdao, China
| | - Qilong Cao
- Qingdao Haier Biotech Co.Ltd, Qingdao, China
| | - Xianggen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology. Qingdao, China
| | - Guohu Di
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
48
|
Navel V, Sapin V, Henrioux F, Blanchon L, Labbé A, Chiambaretta F, Baudouin C, Dutheil F. Oxidative and antioxidative stress markers in dry eye disease: A systematic review and meta-analysis. Acta Ophthalmol 2022; 100:45-57. [PMID: 33938134 DOI: 10.1111/aos.14892] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/10/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To conduct a systematic review and meta-analysis on the levels of oxidative stress markers and antioxidants in dry eye disease (DED) compared with healthy subject. METHOD The PubMed, Cochrane Library, Embase, Science Direct and Google Scholar databases were searched on 10 January 2021 for studies reporting oxidative and antioxidative stress markers in DED and healthy controls. Main meta-analysis was stratified by type of biomarkers, type of samples (tears, conjunctival cells or biopsies), Sjögren's syndrome (SS) (patients with or without SS) and by geographical zones (Asia or Europe). RESULTS We included nine articles, for a total of 333 patients (628 eye samples) with DED and 165 healthy controls (451 eye samples). There is an overall increase in oxidative stress markers in DED compared with healthy controls (standard mean deviation = 2.39, 95% confidence interval 1.85-2.94), with a significant increase in lipid peroxide (1.90, 0.69-3.11), myeloperoxidase (2.17, 1.06-3.28), nitric oxide synthase 3 (2.52, 0.95-4.08), xanthine oxidase/oxidoreductase (2.41, 1.40-5.43), 4-hydroxy-2-nonenal (4HNE) (4.75, 1.67-7.84), malondialdehyde (3.00, 2.55-3.45) and reactive oxygen species (1.31, 0.94-1.68). Oxidative stress markers were higher in tears, conjunctival cells and conjunctival biopsies of DED than controls. Even if small number of studies were included for antioxidants, catalase seemed to be decreased in DED compared with healthy controls (-2.17, -3.00 to -1.34), with an increase of antioxidants in tears of DED patients without SS (1.13, 0.76-1.49). CONCLUSION Oxidative stress markers, and probably antioxidants, were dysregulated in DED, establishing a local oxidative environment in tears, conjunctival cells and tissues.
Collapse
Affiliation(s)
- Valentin Navel
- University Hospital of Clermont‐Ferrand CHU Clermont‐Ferrand, Ophthalmology Clermont‐Ferrand France
- Université Clermont Auvergne CNRS UMR 6293 INSERM U1103 Genetic Reproduction and Development Laboratory (GReD) Translational Approach to Epithelial Injury and Repair Team Clermont‐Ferrand France
| | - Vincent Sapin
- Université Clermont Auvergne CNRS UMR 6293 INSERM U1103 Genetic Reproduction and Development Laboratory (GReD) Translational Approach to Epithelial Injury and Repair Team Clermont‐Ferrand France
- University Hospital of Clermont‐Ferrand CHU Clermont‐Ferrand, Biochemistry and Molecular Genetics Clermont‐Ferrand France
| | - Fanny Henrioux
- Université Clermont Auvergne CNRS UMR 6293 INSERM U1103 Genetic Reproduction and Development Laboratory (GReD) Translational Approach to Epithelial Injury and Repair Team Clermont‐Ferrand France
| | - Loïc Blanchon
- Université Clermont Auvergne CNRS UMR 6293 INSERM U1103 Genetic Reproduction and Development Laboratory (GReD) Translational Approach to Epithelial Injury and Repair Team Clermont‐Ferrand France
| | - Antoine Labbé
- Department of Ophthalmology III Quinze‐Vingts National Ophthalmology Hospital IHU FOReSIGHT Paris France
- Sorbonne Université INSERM CNRS Institut de la Vision Paris France
- Department of Ophthalmology Ambroise Paré Hospital APHP Université de Versailles Saint‐Quentin en Yvelines Versailles France
| | - Frédéric Chiambaretta
- University Hospital of Clermont‐Ferrand CHU Clermont‐Ferrand, Ophthalmology Clermont‐Ferrand France
- Université Clermont Auvergne CNRS UMR 6293 INSERM U1103 Genetic Reproduction and Development Laboratory (GReD) Translational Approach to Epithelial Injury and Repair Team Clermont‐Ferrand France
| | - Christophe Baudouin
- Department of Ophthalmology III Quinze‐Vingts National Ophthalmology Hospital IHU FOReSIGHT Paris France
- Sorbonne Université INSERM CNRS Institut de la Vision Paris France
- Department of Ophthalmology Ambroise Paré Hospital APHP Université de Versailles Saint‐Quentin en Yvelines Versailles France
| | - Frédéric Dutheil
- Université Clermont Auvergne CNRS LaPSCo Physiological and Psychosocial Stress CHU Clermont‐Ferrand University Hospital of Clermont‐Ferrand, Preventive and Occupational Medicine Witty Fit Clermont‐Ferrand France
| |
Collapse
|
49
|
Ling J, Chan BCL, Tsang MSM, Gao X, Leung PC, Lam CWK, Hu JM, Wong CK. Current Advances in Mechanisms and Treatment of Dry Eye Disease: Toward Anti-inflammatory and Immunomodulatory Therapy and Traditional Chinese Medicine. Front Med (Lausanne) 2022; 8:815075. [PMID: 35111787 PMCID: PMC8801439 DOI: 10.3389/fmed.2021.815075] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Dry eye is currently one of the most common ocular surface disease. It can lead to ocular discomfort and even cause visual impairment, which greatly affects the work and quality of life of patients. With the increasing incidence of dry eye disease (DED) in recent years, the disease is receiving more and more attention, and has become one of the hot research fields in ophthalmology research. Recently, with the in-depth research on the etiology, pathogenesis and treatment of DED, it has been shown that defects in immune regulation is one of the main pathological mechanisms of DED. Since the non-specific and specific immune response of the ocular surface are jointly regulated, a variety of immune cells and inflammatory factors are involved in the development of DED. The conventional treatment of DED is the application of artificial tears for lubricating the ocular surface. However, for moderate-to-severe DED, treatment with anti-inflammatory drugs is necessary. In this review, the immunomodulatory mechanisms of DED and the latest research progress of its related treatments including Chinese medicine will be discussed.
Collapse
Affiliation(s)
- Jiawei Ling
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung-Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Miranda Sin-Man Tsang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xun Gao
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ping Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher Wai-Kei Lam
- Faculty of Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Jiang-Miao Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Chun Kwok Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
50
|
Zhou X, Dai Y, Zhai Z, Hong J. WAY-100635 Alleviates Corneal Lesions Through 5-HT 1A Receptor-ROS-Autophagy Axis in Dry Eye. Front Med (Lausanne) 2022; 8:799949. [PMID: 34970573 PMCID: PMC8712493 DOI: 10.3389/fmed.2021.799949] [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: 10/22/2021] [Accepted: 11/22/2021] [Indexed: 12/01/2022] Open
Abstract
Purpose: To explore whether 5-HT1A receptors are involved in the dry eye disease (DED) mouse model and reveal its underlying mechanism. Methods: A C57BL/6J mouse DED model was established via the administration of 0.2% benzalkonium chloride twice a day for 14 days. Corneal fluorescein sodium staining score and Schirmer I test were checked before, and on days 7, 14, and 21 after treatment. The experiment was randomly divided into control, DED, 5-HT1A receptor agonist with or without N-acetylcysteine (NAC) and 5-HT1A receptor antagonist with or without NAC groups. The mRNA expression of inflammatory cytokines was measured by reverse transcription-quantitative polymerase chain reaction. Cellular reactive oxygen species (ROS) were detected by 2', 7'-dichlorodihydrofluorescein diacetate assays. Western blot analysis was used to measure the expression levels of autophagic proteins microtubule-associated protein 1 light chain 3 (LC3B-I/II) and autophagy-related gene 5 (ATG5). Results: 5-HT1A receptor agonist (8-OH-DPAT) increased corneal fluorescein sodium staining spots and 5-HT1A receptor antagonist (WAY-100635) decreased them. Treatment with 8-OH-DPAT was associated with the gene expression of more inflammatory cytokines, such as interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), C-C motif chemokine ligand 2 (CCL2) and C-X-C motif chemokine ligand 10 (CXCL10) compared with treatment with WAY-100635. An increased expression of LC3B-I/II and ATG5 was observed in corneal epithelial cells in the mouse model of DED. 8-OH-DPAT significantly enhanced the expression of LC3B-I/II and ATG5 by disrupting ROS levels. WAY-100635 alleviates autophagy by inhibiting ROS production. Conclusion: Excessive ROS release through 8-OH-DPAT induction can lead to impaired autophagy and increased inflammatory response in DED. WAY-100635 reduces corneal epithelial defects and inflammation in DED, as well as alleviates autophagy by inhibiting ROS production. The activation of the 5-HT1A receptor-ROS-autophagy axis is critically involved in DED development.
Collapse
Affiliation(s)
- Xujiao Zhou
- Department of Ophthalmology, Eye and Ear, Nose and Throat (ENT) Hospital, Fudan University, Shanghai, China
| | - Yiqin Dai
- Department of Ophthalmology, Eye and Ear, Nose and Throat (ENT) Hospital, Fudan University, Shanghai, China
| | - Zimeng Zhai
- Department of Ophthalmology, Eye and Ear, Nose and Throat (ENT) Hospital, Fudan University, Shanghai, China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye and Ear, Nose and Throat (ENT) Hospital, Fudan University, Shanghai, China.,Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|