1
|
Wan X, Zhang Y, Zhang K, Mou Y, Jin X, Huang X. The alterations of ocular surface metabolism and the related immunity inflammation in dry eye. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2025; 5:1-12. [PMID: 39758836 PMCID: PMC11699629 DOI: 10.1016/j.aopr.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 01/07/2025]
Abstract
Background Dry eye disease (DED) stands as a prominent ocular condition of global prevalence, emerging as a growing concern within public health. However, the underlying mechanisms involved in its pathogenesis remain largely unknown. In recent years, with the development of metabolomics, numerous studies have reported alterations in ocular surface metabolism in DED and offered fresh perspectives on the development of DED. Main text The metabolic changes of the ocular surface of DED patients are closely intertwined with the cellular metabolism process and immune inflammation changes. This article expounds upon the correlation between ocular surface metabolism and immune inflammation alterations in DED in terms of glycolysis, lipid metabolism, amino acid metabolism, cellular signaling pathways, and immune inflammation regulation. Conclusions The alterations in ocular surface metabolism of patients with dry eye are closely associated with their inflammatory status. Our work contributes novel insights into the pathogenesis of dry eye diseases and offers innovative molecular targets for diagnosing, detecting, and managing DED patients.
Collapse
Affiliation(s)
- Xiaojie Wan
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Yu Zhang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Kaiye Zhang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Yujie Mou
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Xiuming Jin
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Xiaodan Huang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| |
Collapse
|
2
|
Chen X, Zhang C, Peng F, Wu L, Zhuo D, Wang L, Zhang M, Li Z, Tian L, Jie Y, Huang Y, Yang X, Li X, Lei F, Cheng Y. Identification of glutamine as a potential therapeutic target in dry eye disease. Signal Transduct Target Ther 2025; 10:27. [PMID: 39837870 PMCID: PMC11751114 DOI: 10.1038/s41392-024-02119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
Dry eye disease (DED) is a prevalent inflammatory condition significantly impacting quality of life, yet lacks effective pharmacological therapies. Herein, we proposed a novel approach to modulate the inflammation through metabolic remodeling, thus promoting dry eye recovery. Our study demonstrated that co-treatment with mesenchymal stem cells (MSCs) and thymosin beta-4 (Tβ4) yielded the best therapeutic outcome against dry eye, surpassing monotherapy outcomes. In situ metabolomics through matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) revealed increased glutamine levels in cornea following MSC + Tβ4 combined therapy. Inhibition of glutamine reversed the anti-inflammatory, anti-apoptotic, and homeostasis-preserving effects observed with combined therapy, highlighting the critical role of glutamine in dry eye therapy. Clinical cases and rodent model showed elevated expression of glutaminase (GLS1), an upstream enzyme in glutamine metabolism, following dry eye injury. Mechanistic studies indicated that overexpression and inhibition of GLS1 counteracted and enhanced, respectively, the anti-inflammatory effects of combined therapy, underscoring GLS1's pivotal role in regulating glutamine metabolism. Furthermore, single-cell sequencing revealed a distinct subset of pro-inflammatory and pro-fibrotic corneal epithelial cells in the dry eye model, while glutamine treatment downregulated those subclusters, thereby reducing their inflammatory cytokine secretion. In summary, glutamine effectively ameliorated inflammation and the occurrence of apoptosis by downregulating the pro-inflammatory and pro-fibrotic corneal epithelial cells subclusters and the related IκBα/NF-κB signaling. The present study suggests that glutamine metabolism plays a critical, previously unrecognized role in DED and proposes an attractive strategy to enhance glutamine metabolism by inhibiting the enzyme GLS1 and thus alleviating inflammation-driven DED progression.
Collapse
Affiliation(s)
- Xiaoniao Chen
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China.
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China.
| | - Chuyue Zhang
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Fei Peng
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Lingling Wu
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Deyi Zhuo
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liqiang Wang
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Min Zhang
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Zhaohui Li
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Tian
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ying Jie
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yifei Huang
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xinji Yang
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoqi Li
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fengyang Lei
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yu Cheng
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
3
|
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
|
4
|
Li Y, Sun Y, Xie D, Chen H, Zhang Q, Zhang S, Wen F, Ou JS, Zhang M, Su L, Li X, Wen WP, Chi W. AIP1 Regulates Ocular Angiogenesis Via NLRP12-ASC-Caspase-8 Inflammasome-Mediated Endothelial Pyroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405834. [PMID: 39527457 DOI: 10.1002/advs.202405834] [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: 05/28/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Pathological ocular angiogenesis is a significant cause of irreversible vision loss and blindness worldwide. Currently, most studies have focused on the angiogenesis factors in ocular vascular diseases, and very few endogenous anti-angiogenic compounds have been found. Moreover, although inflammation is closely related to the predominant processes involved in angiogenesis, the mechanisms by which inflammation regulates pathological ocular angiogenesis remain obscure. In this study, a vascular endothelial cells (VECs)-specific anti-angiogenic factor is identified, apoptosis signal-regulating kinase 1(ASK1)-interacting protein-1 (AIP1) as a key pathogenic regulator in a typical ocular angiogenesis model, oxygen-induced retinopathy (OIR), using single-cell RNA sequencing. It is demonstrated that AIP1 inhibited pathological angiogenesis by preventing a particular inflammatory death pathway, namely pyroptosis, in retinal VECs. The assembly of a noncanonical inflammasome is further uncovered, the NLRP12-ASC-caspase-8 inflammasome, which is promoted by decreased AIP1 in OIR. This inflammasome elicited gasdermin D (GSDMD)-dependent endothelial pyroptosis, which in turn promoted the release of vascular endothelial growth factor (VEGF) and interleukin (IL)-1β. Suppression of NLRP12-CASP8-GSDMD axis and AIP1 upregulation reduced VEGF signaling, limiting new vessel formation. These findings reveal a previously uncharacterized inflammatory angiogenic process involving VECs pyroptosis-inducing retinal neovascularization, paving the way for promising therapeutic avenues targeting angiogenesis via AIP1 or pyroptosis.
Collapse
Affiliation(s)
- Yonghao Li
- Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Jinan University, Shenzhen, Guangdong, 518043, China
| | - Yimeng Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, 510060, China
| | - Dasen Xie
- Xiamen Key Laboratory of Ophthalmology, Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, Fujian, 361003, China
| | - Hui Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, 510060, China
| | - Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, 510060, China
| | - Shaochong Zhang
- Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Jinan University, Shenzhen, Guangdong, 518043, China
| | - Feng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, 510060, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Min Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, 510060, China
| | - Lishi Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, 510060, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, 510060, China
| | - Wei-Ping Wen
- Department of Otolaryngology, the Sixth Affiliated Hospital of Sun Yat-sen University, Otorhinolaryngology Institute of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Wei Chi
- Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Jinan University, Shenzhen, Guangdong, 518043, China
| |
Collapse
|
5
|
Wang S, Zheng W, Li T, Yu D, Zhang Q, Ju Y, Wei L. Global research hotspots and trends in anti-inflammatory studies in dry eye: a bibliometric analysis (2004-2024). Front Med (Lausanne) 2024; 11:1451990. [PMID: 39669988 PMCID: PMC11634594 DOI: 10.3389/fmed.2024.1451990] [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: 06/20/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Background Inflammation plays a crucial role in the pathophysiology of dry eye (DE). This study aims to provide a comprehensive overview of the current status, hotspots and trends in DE anti-inflammatory research through bibliometric analysis. Method All publications were searched using the Web of Science Core Collection (WoSCC) database from 2004 to 2024. Bibliometric analyses were performed using VOSviewer, R-bibliometrix, and CiteSpace, and data were managed using Microsoft Office Excel 2019. Results There were 603 papers published between 2004 and 2024 included in this study, with the number of papers increasing each year. The United States was the major contributor, with the largest number of publications and the greatest impact. Baylor College of Medicine was the most influential research institution. Pflugfelder, Stephen C. and Tsubota, Kazuo were the most prolific authors in this area of research, while Dana, Reza was the most cited author in the field with the highest impact. The Journal with the highest number of publications was Investigative Ophthalmology & Visual Science, while the highest impact journal was Ocular Surface. Research hotspots were focused on the mechanisms of inflammation in DE and interventions for anti-inflammatory therapy. Future studies would favor more inflammation-related targeted therapies and physical therapies. Conclusion This study is the first bibliometric analysis to comprehensively summarize research trends and developments in DE anti-inflammatory treatments, pointing out recent research frontiers and hot directions for scholars studying DE anti-inflammatory treatments.
Collapse
Affiliation(s)
- Shan Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wei Zheng
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Ting Li
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Dongxu Yu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Qi Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yuan Ju
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Lijuan Wei
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Stinnett GS, Kuo CH, Ono SJ. Impact of inflammasomes on the ocular surface. Curr Opin Allergy Clin Immunol 2024; 24:368-374. [PMID: 38900843 PMCID: PMC11356675 DOI: 10.1097/aci.0000000000001004] [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] [Indexed: 06/22/2024]
Abstract
PURPOSE OF REVIEW The ocular surface is prone to inflammation due to exposure to environmental irritants and pathogens. Inflammasomes are intracellular, multiprotein complexes that communicate potentially dangerous signals to the immune system. The identification of inflammasomes in various inflammatory ocular surface conditions can aid in the development of therapeutics to treat these chronic inflammatory conditions. RECENT FINDINGS Several inflammasomes have been associated with ocular surface disorders including dry eye disease, keratitis, and allergies. Mechanisms for activation of these inflammasomes with regards to specific disorders have been explored in models to aid in the development of targeted treatments. SUMMARY Research efforts continue to characterize the types of inflammasomes and activators of these in inflammatory ocular surface conditions. Various therapies targeting specific inflammasome types or pyroptosis are being tested preclinically to assess effects on decreasing the associated chronic inflammation.
Collapse
Affiliation(s)
- Gwen S. Stinnett
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Chuan-Hui Kuo
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, Eye Care Centre, The University of British Columbia, Vancouver, BC, Canada
| | - Santa J. Ono
- Departments of Ophthalmology & Visual Science, Microbiology & Immunology and Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
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
|
10
|
Yu L, Meng Q, Li M, Tian L, Wu X, Jie Y. Heating-driven self-assembled glycyrrhizin nanomicelles loading bisdemethoxycurcumin: Preparation, characterization, and efficacy evaluation on experimental dry eye. Colloids Surf B Biointerfaces 2024; 245:114206. [PMID: 39255746 DOI: 10.1016/j.colsurfb.2024.114206] [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: 04/13/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
In this study, a simple but novel preparation method was developed by heating a mixture of dipotassium glycyrrhizinate (DG) and bisdemethoxycurcumin (BDMC) in aqueous solution, and a DG self-assembled nanomicelles-loading BDMC (named B@DNM) ophthalmic solution was successfully fabricated with this heating-driven process. AutoDock simulation analysis revealed that Pi-Alkyl hydrophobic interactions between BDMC and DG played important role in this self-assembled B@DNM. The optimized B@DNM, with a DG:BDMC mass ratio of 40:1 and heating time of 6 h, had a high encapsulation efficacy of 96.70 ± 0.13 % and particle sizes of 117.50 ± 6.07 nm. The apparent solubility of BDMC in B@DNM was significantly improved from bare BDMC (10.40 ± 0.16 μg/ml to 1405.60 ± 6.78 μg/ml) in artificial tears after 4 h incubation. B@DNM had great storage stability as an aqueous ophthalmic solution. B@DNM showed significantly improved in vitro antioxidant activity. Ex vivo hen's egg test-chorioallantoic membrane assay and long-term in vivo mouse eye tolerance evaluation showed that B@DNM had good ocular safety profiles. B@DNM showed improved in vivo corneal permeation profiles in the mouse eyes. Topical administration of B@DNM achieved a significantly improved efficacy on a mouse model of dry eye disease (DED), including accelerating corneal wound healing, restoring corneal sensitivity, and inhibiting corneal neovascularization. Regulation of the high mobility group box 1 signal pathway was involved in B@DNM's strong therapeutic effects. These findings demonstrate that heating is a simple method to prepare ocular nanoformulation with DG, and B@DNM might be a potential ocular drug for treating DED.
Collapse
Affiliation(s)
- Linrong Yu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China; Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | | | - Mengshuang Li
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China; Qingdao Women and Children's Hospital, Qingdao, China
| | - Lei Tian
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xianggen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China.
| | - Ying Jie
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Hou C, Xiao J, Wang Y, Pan X, Liu K, Lu K, Wang Q. Astaxanthin activated the SLC7A11/GPX4 pathway to inhibit ferroptosis and enhance autophagy, ameliorating dry eye disease. Front Pharmacol 2024; 15:1407659. [PMID: 39224780 PMCID: PMC11366873 DOI: 10.3389/fphar.2024.1407659] [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: 03/27/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
Dry eye disease (DED) is a common eye disease in clinical practice. The crucial pathogenesis of DED is that hyperosmolarity activates oxidative stress signaling pathways in corneal epithelial and immune cells and, thus, produces inflammatory molecules. The complex pathological changes in the dry eye still need to be elucidated to facilitate treatment. In this study, we found that astaxanthin (AST) can protect against DED through the SLC7A11/GPX4 pathway. After treatment with AST, the SLC7A11/GPX4 pathway was positively activated in DED both in vivo and in vitro, accompanied by enhanced autophagy and decreased ferroptosis. In hyperosmolarity-induced DED corneal epithelial cells, AST increased the expression of ferritin to promote iron storage and reduce Fe2+ overload. It increased glutathione (GSH) and GPX4, scavenged reactive oxygen species (ROS) and lipid peroxide, and rescued the mitochondrial structure to prevent ferroptosis. Furthermore, inhibition of ferroptosis by ferrostatin-1 (Fer-1), iron chelator deferoxamine mesylate (DFO), or AST could activate healthy autophagic flux. In addition, in a dry eye mouse model, AST upregulated SLC7A11 and GPX4 and inhibited ferroptosis. To summarize, we found that AST can ameliorate DED by reinforcing the SLC7A11/GPX4 pathway, which mainly affects oxidative stress, autophagy, and ferroptosis processes.
Collapse
Affiliation(s)
- Chenting Hou
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Eye Hospital of Shandong Province, Jinan, China
| | - Jie Xiao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Youhai Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinghui Pan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kangrui Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kang Lu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Li X, Chen C, Chen Y, Jiang K, Zhao X, Zhang F, Li Y. Oridonin ameliorates ocular surface inflammatory responses by inhibiting the NLRP3/caspase-1/GSDMD pyroptosis pathway in dry eye. Exp Eye Res 2024; 245:109955. [PMID: 38843984 DOI: 10.1016/j.exer.2024.109955] [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: 09/27/2023] [Revised: 04/26/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Chronic inflammation is one of the central drivers in the development of dry eye disease (DED), in which pyroptosis induced by the NLRP3/caspase-1/gasdermin D (GSDMD) pathway plays a key role. This pathway has become a major target for the treatment of a variety of inflammatory disorders. Oridonin (Ori) is a naturally occurring substance with anti-inflammatory properties obtained from Rabdosia rubescens. Whether Ori can exert an anti-inflammatory effect on DED, and its anti-inflammatory mechanism of action, are still unknown. This experiment is intended to investigate the impact of Ori on the hyperosmolarity-induced NLRP3/caspase-1/GSDMD pyroptosis pathway in immortalized human corneal epithelial (HCE-T) cells, as well as its efficacy and mechanism of action on ocular surface injury in DED mice. Our study showed that Ori could inhibit hyperosmotic-induced pyroptosis through the NLRP3/caspase-1/GSDMD pathway in HCE-T cells, and similarly, Ori inhibited the expression of this pathway in DED mice. Moreover, Ori was protective against hyperosmolarity-induced HCE-T cell damage. In addition, we found that the morphology and number of HCE-T cells were altered under culture conditions of various osmolarities. With increasing osmolarity, the proliferation, migration, and healing ability of HCE-T cells decreased significantly, and the expression of N-GSDMD was elevated. In a mouse model of DED, Ori application inhibited the expression of the NLRP3/caspase-1/GSDMD pyroptosis pathway, improved DED signs and injury, decreased corneal sodium fluorescein staining scores, and increased tear volume. Thus, our study suggests that Ori has potential applications for the treatment of DED, provides potential novel therapeutic approaches to treat DED, and provides a theoretical foundation for treating DED using Ori.
Collapse
Affiliation(s)
- Xiaojing Li
- Medical College, Graduate School of Medicine, Qingdao University, Qingdao, 266071, China; Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Chen Chen
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Ying Chen
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Kaiwen Jiang
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Xinmei Zhao
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Fenglan Zhang
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| | - Yuanbin Li
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| |
Collapse
|
13
|
Sun Y, Li F, Liu Y, Qiao D, Yao X, Liu GS, Li D, Xiao C, Wang T, Chi W. Targeting inflammasomes and pyroptosis in retinal diseases-molecular mechanisms and future perspectives. Prog Retin Eye Res 2024; 101:101263. [PMID: 38657834 DOI: 10.1016/j.preteyeres.2024.101263] [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: 07/30/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Retinal diseases encompass various conditions associated with sight-threatening immune responses and are leading causes of blindness worldwide. These diseases include age-related macular degeneration, diabetic retinopathy, glaucoma and uveitis. Emerging evidence underscores the vital role of the innate immune response in retinal diseases, beyond the previously emphasized T-cell-driven processes of the adaptive immune system. In particular, pyroptosis, a newly discovered programmed cell death process involving inflammasome formation, has been implicated in the loss of membrane integrity and the release of inflammatory cytokines. Several disease-relevant animal models have provided evidence that the formation of inflammasomes and the induction of pyroptosis in innate immune cells contribute to inflammation in various retinal diseases. In this review article, we summarize current knowledge about the innate immune system and pyroptosis in retinal diseases. We also provide insights into translational targeting approaches, including novel drugs countering pyroptosis, to improve the diagnosis and treatment of retinal diseases.
Collapse
Affiliation(s)
- Yimeng Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Fan Li
- Eye Center, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yunfei Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Dijie Qiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xinyu Yao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Guei-Sheung Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dequan Li
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chuanle Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Tao Wang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Guangming District, Shenzhen, 518132, China; School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao You'anMen Street, Beijing, 100069, 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
|
14
|
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
|
15
|
Li X, Peng H, Kang J, Sun X, Liu J. IL-1β induced down-regulation of miR-146a-5p promoted pyroptosis and apoptosis of corneal epithelial cell in dry eye disease through targeting STAT3. BMC Ophthalmol 2024; 24:144. [PMID: 38553670 PMCID: PMC10981279 DOI: 10.1186/s12886-024-03396-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 03/14/2024] [Indexed: 04/01/2024] Open
Abstract
AIM To elaborate the underlying mechanisms by which IL-1β promote progression of Dry eye disease(DED) through effect on pyroptosis and apoptosis of corneal epithelial cells(CECs). METHODS 400 mOsM solutions were used to establish the DED model (hCECs- DED). RT-qPCR was performed to measure IL-1β mRNA and miR-146a-5p in CECs. Western blotting was performed to measure STAT3, GSDMD, NLRP3, and Caspase-1 levels. Cell counting kit-8 assay was adopted to check cell viability. Apoptosis was detected by flow cytometry. ELISAs were performed to determine IL-18, IL-33 and LDH. The luciferase test detects targeting relationships. RESULTS After treatment with 400 mOsM solution, cell viability decreased and apoptosis increased. Compared with hCECs, IL-1β was increased and miR-146a-5p was decreased in hCECs-DED. At the same time, GSDMD, NLRP3, Caspase-1, IL-18, IL-33 and LDH were significantly higher in hCECs-DED than in hCECs, while IL-1β silencing reversed this effect. In addition, IL-1β negatively regulated miR-146a-5p. MiR-146a-5p mimics eliminated the inhibition of hCECs-DED pyroptosis and apoptosis caused by IL-1β silencing. At the same time, miR-146a-5p reduced STAT3 levels in hCECs. CONCLUSION Highly expressed IL-1β promoted pyroptosis and apoptosis of hCECs- DED through downregulated miR-146a-5p and inhibited STAT3.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, 650500, Kunming, Yunnan, China
| | - Hua Peng
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, 650500, Kunming, Yunnan, China
| | - Jianshu Kang
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, 650500, Kunming, Yunnan, China
| | - Xiaomei Sun
- Department of Ophthalmology, Affiliated Hospital of Yunnan University, 650500, Kunming, Yunnan, China
| | - Jian Liu
- Department of Ophthalmology, China Academy of C.M.S. Eye Hospital, NO. 33 Lugu Road, Shijingshan District, 100040, Beijing, China.
| |
Collapse
|
16
|
Chen J, Gong Y, Sun X, Chen N, Zhao Z, Zhang W, Zheng Y. Prostaglandin E2 may clinically alleviate dry eye disease by inducing Th17 cell differentiation. Chem Biol Drug Des 2024; 103:e14477. [PMID: 38361150 DOI: 10.1111/cbdd.14477] [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: 11/10/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/17/2024]
Abstract
Dry eye (DE) is a multifactorial ocular surface disease characterised by an imbalance in tear homeostasis. The pathogenesis of DE is complex and related to environmental, immunological (e.g., T helper 17 cells) and other factors. However, the DE disease pathogenesis remains unclear, thereby affecting its clinical treatment. This study aimed to explore the mechanism through which prostaglandin E2 (PGE2) affects DE inflammation by regulating Th17. The DE mouse model was established through subcutaneous injection of scopolamine hydrobromide. The tear secretion test and break-up time (BUT) method were used to detect tear secretion and tear film BUT, respectively. Enzyme-linked immunosorbent assay (ELISA) was used to detect the concentrations of PGE2, interleukin (IL)-17, IL-6 and tumour necrosis factor (TNF-α) in tear fluid and those of PGE2 and IL-17 in the serum. RT-qPCR and western blotting were used to test the mRNA and protein expression levels of IL-17 and retinoid-related orphan receptor-γt (RORγt). PGE2 was highly expressed in the DE mouse model. The mRNA and protein levels of IL-17 and the key Th17 transcription factor RORγt were increased in tissues of the DE mice. Moreover, PGE2 promoted tear secretion, reduced the BUT, increased the IL-17 concentration in tears and increased the Th17 cell proportion in DE, whereas the PGE2 receptor inhibitor AH6809 reversed the effects of PGE2 on tear secretion, BUT, and the Th17 cell proportion in draining lymph node (DLN) cells. Taken together, the study findings indicate that PGE2 could induce DE-related symptoms by promoting Th17 differentiation.
Collapse
Affiliation(s)
- Jingyao Chen
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
- Ophthalmology Department of Kunming First People's Hospital, Kunming, China
| | - Yu Gong
- Kunming Medical University, Kunming, China
| | - Xiaoyu Sun
- Kunming Medical University, Kunming, China
| | - Nuo Chen
- Kunming Medical University, Kunming, China
| | - Zijun Zhao
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Weijia Zhang
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Yixin Zheng
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| |
Collapse
|
17
|
Ban J, Chang S, Ma P, Wang X, Liu F. lncRNA Profiling of Exosomes and Its Communication Role in Regulating Silica-Stimulated Macrophage Apoptosis and Fibroblast Activation. Biomolecules 2024; 14:146. [PMID: 38397383 PMCID: PMC10886698 DOI: 10.3390/biom14020146] [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: 11/23/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Long-term silica particle exposure leads to interstitial pulmonary inflammation and fibrosis, called silicosis. Silica-activated macrophages secrete a wide range of cytokines resulting in persistent inflammation. In addition, silica-stimulated activation of fibroblast is another checkpoint in the progression of silicosis. The pathogenesis after silica exposure is complex, involving intercellular communication and intracellular signaling pathway transduction, which was ignored previously. Exosomes are noteworthy because of their crucial role in intercellular communication by delivering bioactive substances, such as lncRNA. However, the expression profile of exosomal lncRNA in silicosis has not been reported yet. In this study, exosomes were isolated from the peripheral serum of silicosis patients or healthy donors. The exosomal lncRNAs were profiled using high-throughput sequencing technology. Target genes were predicted, and functional annotation was performed using differentially expressed lncRNAs. Eight aberrant expressed exosomal lncRNAs were considered to play a key role in the process of silicosis according to the OPLS-DA. Furthermore, the increased expression of lncRNA MSTRG.43085.16 was testified in vitro. Its target gene PARP1 was critical in regulating apoptosis based on bioinformatics analysis. In addition, the effects of exosomes on macrophage apoptosis and fibroblast activation were checked based on a co-cultured system. Our findings suggested that upregulation of lncRNA MSTRG.43085.16 could regulate silica-induced macrophage apoptosis through elevating PARP1 expression, and promote fibroblast activation, implying that the exosomal lncRNA MSTRG.43085.16 might have potential as a biomarker for the early diagnosis of silicosis.
Collapse
Affiliation(s)
- Jiaqi Ban
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, China; (J.B.); (S.C.); (P.M.)
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, School of Public Health, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Shuai Chang
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, China; (J.B.); (S.C.); (P.M.)
| | - Pengwei Ma
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, China; (J.B.); (S.C.); (P.M.)
| | - Xin Wang
- Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China;
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, China; (J.B.); (S.C.); (P.M.)
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
| |
Collapse
|
18
|
Liao K, Zeng H, Yang X, He D, Wang B, Yuan J. KCNK5 Regulating Potassium Efflux and Inducing Pyroptosis in Corneal Epithelial Cells Through TNFSF10-Mediated Autophagy in Dry Eye. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 38236186 PMCID: PMC10807492 DOI: 10.1167/iovs.65.1.34] [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: 11/09/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024] Open
Abstract
Purpose The purpose of this study was to elucidate the involvement of potassium two pore domain channel subfamily K member 5 (KCNK5)-mediated potassium efflux in the pathogenesis of dry eye and to unravel the underlying molecular mechanisms. Methods To induce experimental dry eye in adult wild-type C57BL/6 mice, scopolamine was administered via subcutaneous injection, and the mice were subjected to desiccating stress. To create an in vitro model of dry eye, desiccation stress was applied to the human corneal epithelial cell line (HCE-T). Intracellular potassium concentration was quantified using inductively coupled plasma mass spectrometry. Cellular death was assessed through lactate dehydrogenase assays. Gene expression profiling was conducted through both RNA sequencing and quantitative real-time PCR. Protein analysis was carried out through Western blotting and immunofluorescence staining. Assessment of the corneal epithelial defect area was conducted through fluorescein sodium staining. Tear secretion was quantified using the phenol red cotton thread method. Results Potassium efflux was observed to further facilitate corneal epithelial pyroptosis. KCNK5 exhibited upregulation in both in vivo and in vitro models of dry eye. The overexpression of KCNK5 was observed to induce potassium efflux and activate the NLR family pyrin domain containing 3 (NLRP3) inflammasome-mediated pyroptosis in vitro. Silencing KCNK5 effectively mitigated pyroptosis in dry eye. Additionally, the overexpression of KCNK5 results in the downregulation of TNF superfamily member 10 (TNFSF10) and subsequent impairment of autophagy. TNFSF10 supplementation could promote autophagy and mitigate pyroptosis in dry eye. Conclusions The upregulation of KCNK5 mediates TNFSF10 to impair autophagy and induce pyroptosis in dry eye. Consequently, targeting KCNK5 may represent a novel and promising approach to therapeutic intervention in the management of dry eye.
Collapse
Affiliation(s)
- Kai Liao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Hao Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Bowen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
19
|
Zhang W, Yin J, Deng Y, Gong Y, Sun X, Chen J. Prostaglandin E2 promotes Th17 differentiation induces corneal epithelial cell apoptosis and participates in the progression of dry eye. Arch Biochem Biophys 2024; 751:109823. [PMID: 37984760 DOI: 10.1016/j.abb.2023.109823] [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: 07/01/2023] [Revised: 10/23/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
This study is mainly based on T helper type 17 (Th17) cells analysis of the mechanism of prostaglandin E2 (PGE2) promoting the progression of dry eye (DE). Scopolamine and dry environment were used to induce mice DE model. Celecoxib was used to inhibit PGE2. Corneal epithelial cells and CD4+ T cells were used to construct a co-culture system. The osmotic pressure was increased by adding NaCl to simulate DE in vitro. AH6809 and E7046 were used to pre-culture to inhibit EP2/4 in T cells to verify the effect of exogenous PGE2 on Th17 cell differentiation and corneal epithelial cell apoptosis. The function of Th17 cells was analyzed by detecting RORγt and interleukin-17 (IL-17). PGE2 was instilled on the ocular surface to induce DE symptoms of mice. AH6809 and E7046 were used to inhibit EP2/4. The corneal epithelial cell apoptosis was observed by TUNEL. The proportion of Th17 cells in corneal tissue and draining lymph nodes (DLNs) was detected by flow cytometry. In DE mice, the concentration of PGE2 and IL-17 increased in tears, and the proportion of Th17 increased, while inhibition of PGE2 alleviated the symptoms of DE and inhibited Th17 differentiation. Hypertonic environment induces corneal epithelial cells to secrete PGE2. PGE2 promoted the expression of EP2/4 and the differentiation of Th17 cells in vitro. The hypertonic environment promoted PGE2 level and the apoptosis of corneal epithelial cells in the co-culture system. PGE2 alone did not cause corneal epithelial cell apoptosis, while PGE2 promoted apoptosis by promoting Th17. Blocking EP2/4 reduced the induction of Th17 differentiation by PGE2 and the promoted corneal epithelial cell apoptosis. Animal experiments showed that exogenous PGE2 induced DE symptoms. Blocking EP2/4 not only inhibited the proportion of Th17, but also alleviated the apoptosis of corneal epithelial cells caused by PGE2. PGE2 induces aggravation of inflammation by promoting the level of Th17 in the ocular surface, and causes corneal epithelial cell apoptosis, thereby participating in the progression of DE.
Collapse
Affiliation(s)
- Weijia Zhang
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Jianwei Yin
- Department of Anesthesiology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yachun Deng
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yu Gong
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Xiaoyu Sun
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Jingyao Chen
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
20
|
Zhang Z, Yang L, Lei X, Yu J, Wang L, Cao H, Gu H. Mechanism of non-small cell lung cancer cell-derived exosome miR-196b-5p promoting pyroptosis of tumor T cells and tumor cell proliferation by downregulating ING5. J Biochem Mol Toxicol 2024; 38:e23629. [PMID: 38229318 DOI: 10.1002/jbt.23629] [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: 03/08/2023] [Revised: 11/21/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024]
Abstract
In the world, lung cancer is one of the most common malignant cancers and has become the leading cause of death of cancers in China, among which non-small cell lung cancer (NSCLC) accounts for a relatively high proportion, but there is a lack of effective treatment at present. An animal model of NSCLC was established, and BEAS-2b, H1299, Lewis, and T cells were used for subsequent experimental verification. The level of miR-196b-5p was detected by quantitative real-time polymerase chain reaction. Growth inhibitor 5 (ING5), CD9, CD63, HSP70, Caspase-1, NLRP3, and GSDMD-NT were detected by western blot. The level of ING5 was confirmed by immunohistochemistry, the location of miR-196b-5p was analyzed by fluorescence in situ hybridization (FISH), cell viability was investigated by Cell Counting Kit-8 kit, and interleukin (IL)-1β and IL-18 were confirmed by enzyme-linked immunosorbent assay. Cell apoptosis was detected by flow cytometry. In addition, the binding site was verified by dual-luciferase reporter gene experiments. Tumor volume was measured. TUNEL staining was used to detect apoptosis. Flow cytometry was used to measure the levels of CD8 T, CD4 T, and Treg cells in tumors. miR-196-5p was highly expressed in exosomes secreted by tumor cells. miR-196-5p negatively targeted ING5 to promote the growth of tumor cells. Cancer-derived exosomes promote pyroptosis of T cells to further aggravate the development of cancer. Exosome-derived miR-196b-5p promoted pyroptosis of T cells. Exosome-derived miR-196b-5p inhibited the level of ING5 to promote tumor growth and accelerate the process of NSCLC.
Collapse
Affiliation(s)
- Zhixian Zhang
- Department of Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lei Yang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuefen Lei
- Department of Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia Yu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lijuan Wang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hongming Cao
- Department of Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hou Gu
- Department of Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
21
|
Ran S, Shu Q, Gao X. Dermatophagoides Pteronyssinus 1 (DerP1) May Trigger NLRP3-Mediated Corneal Epithelial Cell Pyroptosis by Elevating Interleukin-33 Expression Levels. Curr Eye Res 2023; 48:1100-1111. [PMID: 37615401 DOI: 10.1080/02713683.2023.2250583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023]
Abstract
PURPOSE To characterize the in vivo effects of Dermatophagoides pteronyssinus 1 (DerP1) in mice and determine the underlying NLRP3 inflammasome-mediated pyroptosis signaling mechanisms in the human corneal epithelial cells (HCECs). METHODS DerP1 was used to induce allergic conjunctivitis in C57 mice. HCECs were sensitized with DerP1 in vitro to mimic their condition observed in allergic conjunctivitis in vivo. Transmission electron microscopy was used to evaluate pyroptosis in the HCECs, enzyme-linked immunosorbent assays to assess interleukin (IL)-33, IL-1β and IL-4 levels, flow cytometry to detect the proportion of Th2 cells, MTT assays to assess cell metabolic activity, immunofluorescence to evaluate the effects of DerP1 on functional HCEC phenotypes, and Western blot assays to detect the expression of NOD-like receptor family pyrin domain-containing 3 (NLRP3), gasdermin D (GSDMD), N-terminal fragment of GSDMD (GSDMD-N), pro-caspase-1, cleaved caspase-1, IL-1β, and IL-33. IL-33 expression in the HCECs was knocked down via lentivirus transfection. RESULTS In vivo, DerP1 promotes pyroptosis, production of Th2 inflammatory cytokines and IL-33, and NLRP3 activation in mouse corneas. In vitro, pyroptotic bodies were found in the HCECs after sensitization with DerP1. Various concentrations of DerP1 increased the expression levels of NLRP3, GSDMD, GSDMD-N, pro-caspase-1, cleaved caspase-1, and IL-1β in the HCECs, with the largest increase observed after exposure to 20 µM DerP1. In vitro, recombinant human IL-33 mediated the expression of pyroptotic biomarkers in the HCECs, whereas IL-33 silencing diminished 20 µM DerP1-induced increase in their expression levels. CONCLUSIONS DerP1 induces pyroptosis and allergic conjunctivitis, the expression of Th2 inflammatory cytokines, NLRP3 activation, and IL-33 in mouse corneas in our model. These effects would attribute to its activating NLRP3-GSDMD signaling pathway axis via enhancing IL-33 expression in HCECs.
Collapse
Affiliation(s)
- Shengming Ran
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qinxin Shu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xu Gao
- Department of Ophthalmology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| |
Collapse
|
22
|
Latifi-Navid H, Barzegar Behrooz A, Jamehdor S, Davari M, Latifinavid M, Zolfaghari N, Piroozmand S, Taghizadeh S, Bourbour M, Shemshaki G, Latifi-Navid S, Arab SS, Soheili ZS, Ahmadieh H, Sheibani N. Construction of an Exudative Age-Related Macular Degeneration Diagnostic and Therapeutic Molecular Network Using Multi-Layer Network Analysis, a Fuzzy Logic Model, and Deep Learning Techniques: Are Retinal and Brain Neurodegenerative Disorders Related? Pharmaceuticals (Basel) 2023; 16:1555. [PMID: 38004422 PMCID: PMC10674956 DOI: 10.3390/ph16111555] [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: 09/27/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Neovascular age-related macular degeneration (nAMD) is a leading cause of irreversible visual impairment in the elderly. The current management of nAMD is limited and involves regular intravitreal administration of anti-vascular endothelial growth factor (anti-VEGF). However, the effectiveness of these treatments is limited by overlapping and compensatory pathways leading to unresponsiveness to anti-VEGF treatments in a significant portion of nAMD patients. Therefore, a system view of pathways involved in pathophysiology of nAMD will have significant clinical value. The aim of this study was to identify proteins, miRNAs, long non-coding RNAs (lncRNAs), various metabolites, and single-nucleotide polymorphisms (SNPs) with a significant role in the pathogenesis of nAMD. To accomplish this goal, we conducted a multi-layer network analysis, which identified 30 key genes, six miRNAs, and four lncRNAs. We also found three key metabolites that are common with AMD, Alzheimer's disease (AD) and schizophrenia. Moreover, we identified nine key SNPs and their related genes that are common among AMD, AD, schizophrenia, multiple sclerosis (MS), and Parkinson's disease (PD). Thus, our findings suggest that there exists a connection between nAMD and the aforementioned neurodegenerative disorders. In addition, our study also demonstrates the effectiveness of using artificial intelligence, specifically the LSTM network, a fuzzy logic model, and genetic algorithms, to identify important metabolites in complex metabolic pathways to open new avenues for the design and/or repurposing of drugs for nAMD treatment.
Collapse
Affiliation(s)
- Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
- Departments of Ophthalmology and Visual Sciences and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3T 2N2, Canada;
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan 6517838636, Iran;
| | - Maliheh Davari
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Masoud Latifinavid
- Department of Mechatronic Engineering, University of Turkish Aeronautical Association, 06790 Ankara, Turkey;
| | - Narges Zolfaghari
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Sepideh Taghizadeh
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Mahsa Bourbour
- Department of Biotechnology, Alzahra University, Tehran 1993893973, Iran;
| | - Golnaz Shemshaki
- Department of Studies in Zoology, University of Mysore, Manasagangothri, Mysore 570005, India;
| | - Saeid Latifi-Navid
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil 5619911367, Iran;
| | - Seyed Shahriar Arab
- Biophysics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 1411713116, Iran;
| | - Zahra-Soheila Soheili
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran 1666673111, Iran;
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
23
|
Xi X, Wang M, Chen Q, Ma J, Zhang J, Li Y. DNMT1 regulates miR-20a/TXNIP-mediated pyroptosis of retinal pigment epithelial cells through DNA methylation. Mol Cell Endocrinol 2023; 577:112012. [PMID: 37506869 DOI: 10.1016/j.mce.2023.112012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Pyroptosis of retinal pigment epithelium (RPE) cells is associated with the etiology of diabetic retinopathy (DR). In this study, we investigated the effect of DNMT1 on RPE cell pyroptosis by regulating miR-20a/TXNIP expression through DNA methylation. METHODS High glucose (HG)-induced ARPE-19 cells and mice were injected with streptozotocin (STZ) to generate DR cells and animal models. RT‒qPCR was used to detect the expression of miR-20a, and methylation-specific PCR (MS-PCR) was used to determine the occurrence of methylation of miR-20a. The expression of pyroptosis-related proteins (caspase-1 and NLRP3) and DNA methyltransferase (DNMT1) was detected by western blotting, and the expression of inflammatory factors (IL-1β and IL-18) was detected by ELISA. Apoptosis was detected by flow cytometry and TUNEL. HE staining was used to observe the pathological changes in retinal tissue in mice. RESULTS In HG-induced DR cell models, the expression of miR-20a was significantly downregulated, while the expression of inflammatory factors (IL-1β, IL-18) and pyroptosis-associated proteins (caspase-1, NLRP3) was significantly upregulated. Transfection of miR-20a mimic can effectively reverse HG-induced pyroptosis and release of inflammatory factors. DNMT1 promotes miR-20a methylation and inhibits the expression of miR-20a. DNMT1-mediated methylation is involved in the pyroptosis process of high glucose-induced RPE cells, and silencing DNMT1 can promote the expression of miR-20a, thereby inhibiting the release of IL-1β and IL-18 and reducing the occurrence of cell pyroptosis. miR-20a targets negative regulation of TXNIP expression, and overexpression of TXNIP can effectively reverse the inhibitory effect of miR-20a on pyroptosis. The methylation inhibitor 5-AZ can inhibit the occurrence of pyroptosis and DR processes, while treatment with a miR-20a inhibitor or OE-TXNIP can reverse the effect of 5-AZ. CONCLUSION DNMT1 promotes DNA methylation, decreases the expression of miR-20a and increases the expression of TXNIP, which ultimately leads to the occurrence of pyroptosis in RPE cells.
Collapse
Affiliation(s)
- Xiaoting Xi
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Min Wang
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, 200031, China
| | - Qianbo Chen
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jia Ma
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Junyan Zhang
- Department of Clinical Epidemiology and Evidence-based Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Yan Li
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| |
Collapse
|
24
|
Huang L, Tao Y, Wu X, Wu J, Shen M, Zheng Z. The role of NLRP12 in inflammatory diseases. Eur J Pharmacol 2023; 956:175995. [PMID: 37572944 DOI: 10.1016/j.ejphar.2023.175995] [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: 04/20/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
Nucleotide-binding leucine-rich repeat-containing receptor 12 (NLRP12), a highly conserved protein containing an N-terminal pyrin domain (PYD), a nucleotide-binding domain and a C-terminal leucine-rich repeat region, belongs to the nucleotide-binding oligomerization domain-like receptor-containing PYD (NLRP) family and is a cytoplasmic sensor that plays a negative role in inflammation. NLRP12 is involved in multiple disease processes, including formation of inflammasomes and regulation of both canonical and noncanonical inflammatory signaling pathways. NLRP12 and pathogenic infections are closely linked, and alterations in NLRP12 expression and activity are associated with inflammatory diseases. In this review, we begin with a summary of the mechanisms of negative regulation by NLRP12. We then underscore the important roles of NLRP12 in the onset and progression of inflammation, infectious disease, host defense, carcinogenesis and COVID-19. Finally, we highlight factors that influence NLRP12 activity, including synthetic and naturally derived agonists, and are regarded as potential therapeutic agents to overcome inflammatory diseases.
Collapse
Affiliation(s)
- Lili Huang
- Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315100, Zhejiang, China
| | - Youli Tao
- Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315100, Zhejiang, China
| | - Xiping Wu
- Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315100, Zhejiang, China
| | - Jianzhang Wu
- The Eye Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Mengya Shen
- Affiliated Hospital of Jiaxing University, Jiaxing Maternity and Child Health Care Hospital in Zhejiang Province, Jiaxing, 314000, Zhejiang, China.
| | - Zhiwei Zheng
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
25
|
Han Y, Zhang Y, Yuan K, Wu Y, Jin X, Huang X. Hyperosmolarity promotes macrophage pyroptosis by driving the glycolytic reprogramming of corneal epithelial cells in dry eye disease. Front Med 2023; 17:781-795. [PMID: 37266854 DOI: 10.1007/s11684-023-0986-x] [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: 03/06/2022] [Accepted: 01/28/2023] [Indexed: 06/03/2023]
Abstract
Tear film hyperosmolarity plays a core role in the development of dry eye disease (DED) by mediating the disruption of ocular surface homeostasis and triggering inflammation in ocular surface epithelium. In this study, the mechanisms involving the hyperosmolar microenvironment, glycolysis mediating metabolic reprogramming, and pyroptosis were explored clinically, in vitro, and in vivo. Data from DED clinical samples indicated that the expression of glycolysis and pyroptosis-related genes, including PKM2 and GSDMD, was significantly upregulated and that the secretion of IL-1β significantly increased. In vitro, the indirect coculture of macrophages derived from THP-1 and human corneal epithelial cells (HCECs) was used to discuss the interaction among cells. The hyperosmolar environment was found to greatly induce HCECs' metabolic reprogramming, which may be the primary cause of the subsequent inflammation in macrophages upon the activation of the related gene and protein expression. 2-Deoxy-d-glucose (2-DG) could inhibit the glycolysis of HCECs and subsequently suppress the pyroptosis of macrophages. In vivo, 2-DG showed potential efficacy in relieving DED activity and could significantly reduce the overexpression of genes and proteins related to glycolysis and pyroptosis. In summary, our findings suggested that hyperosmolar-induced glycolytic reprogramming played an active role in promoting DED inflammation by mediating pyroptosis.
Collapse
Affiliation(s)
- Yu Han
- Eye Center, The Second Affiliated Hospital, Zhejiang University 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
| | - Yu Zhang
- Eye Center, The Second Affiliated Hospital, Zhejiang University 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
| | - Kelan Yuan
- Eye Center, The Second Affiliated Hospital, Zhejiang University 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
| | - Yaying Wu
- Eye Center, The Second Affiliated Hospital, Zhejiang University 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
| | - Xiuming Jin
- Eye Center, The Second Affiliated Hospital, Zhejiang University 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 Huang
- Eye Center, The Second Affiliated Hospital, Zhejiang University 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
|
26
|
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
|
27
|
Guo XX, Pu Q, Hu JJ, Chang XJ, Li AL, Li XY. The role of regulated necrosis in inflammation and ocular surface diseases. Exp Eye Res 2023:109537. [PMID: 37302745 DOI: 10.1016/j.exer.2023.109537] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/28/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
In recent decades, numerous types of regulated cell death have been identified, including pyroptosis, ferroptosis and necroptosis. Regulated necrosis is characterized by a series of amplified inflammatory responses that result in cell death. Therefore, it has been suggested to play an essential role in the pathogenesis of ocular surface diseases. The cell morphological features and molecular mechanisms of regulated necrosis are discussed in this review. Furthermore, it summarizes the role of ocular surface diseases, such as dry eye, keratitis, and cornea alkali burn, as potential disease prevention and treatment targets.
Collapse
Affiliation(s)
- Xiao-Xiao Guo
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qi Pu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jing-Jie Hu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xue-Jiao Chang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ao-Ling Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xin-Yu Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
28
|
Chai R, Li Y, Shui L, Ni L, Zhang A. The role of pyroptosis in inflammatory diseases. Front Cell Dev Biol 2023; 11:1173235. [PMID: 37250902 PMCID: PMC10213465 DOI: 10.3389/fcell.2023.1173235] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Programmed cell death has crucial roles in the physiological maturation of an organism, the maintenance of metabolism, and disease progression. Pyroptosis, a form of programmed cell death which has recently received much attention, is closely related to inflammation and occurs via canonical, non-canonical, caspase-3-dependent, and unclassified pathways. The pore-forming gasdermin proteins mediate pyroptosis by promoting cell lysis, contributing to the outflow of large amounts of inflammatory cytokines and cellular contents. Although the inflammatory response is critical for the body's defense against pathogens, uncontrolled inflammation can cause tissue damage and is a vital factor in the occurrence and progression of various diseases. In this review, we briefly summarize the major signaling pathways of pyroptosis and discuss current research on the pathological function of pyroptosis in autoinflammatory diseases and sterile inflammatory diseases.
Collapse
Affiliation(s)
| | | | | | - Longxing Ni
- *Correspondence: Longxing Ni, ; Ansheng Zhang,
| | | |
Collapse
|
29
|
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
|
30
|
Zhao D, Zhao H, He Y, Zhang M. BMSC reduces ROS and inflammation levels by inhibiting TLR4/MYD88/NF-κB signaling axis to alleviate dry eye. RESEARCH SQUARE 2023:rs.3.rs-2739871. [PMID: 37131693 PMCID: PMC10153363 DOI: 10.21203/rs.3.rs-2739871/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Objective To investigate the therapeutic effect of Bone marrow mesenchymal stem cells (BMSCs) on dry eye mice, and to investigate the mechanism of TLR4/MYD88/NF-κB signaling pathway on corneal injury repair in dry eye mice. Methods To establish a hypertonic dry eye cell model. Western blot for measureing the protein expressions of caspase-1, IL-1β,NLRP3 and ASC,and Rt-qpcr for mRNA expression. Flow cytometry for detecting the ROS content and apoptosis rate. CCK-8 for detecting the proliferation activity of cells, and ELISA for the levels of inflammation-related factors.The levels of inflammation-related factors were detected by ELISA. The dry eye mouse model of benzalkonium chloride was established. Three clinical parameters used to evaluate ocular surface damage, namely tear secretion, tear film rupture time and corneal sodium fluorescein staining, were measured with phenol cotton thread. Flow cytometry and TUNEL staining are both for he apoptosis rate. Western blot also for detecting the protein expressions of TLR4, MYD88, NF-κB, inflammation-related factors and apoptosis-related factors . The pathological changes were evaluated by HE and PAS staining. Results In vitro, BMSCs and inhibitors of TLR4, MYD88 and NF-κB showed decreased ROS content, decreased inflammatory factor protein level, decreased apoptotic protein level and increased mRNA expression compared with NaCl group. BMSCS partially reversed cell apoptosis induced by NaCl and improved cell proliferation. In vivo, it reduces corneal epithelial defects, goblet cell loss and inflammatory cytokine production, and increases tear production. In vitro, BMSC and inhibitors of TLR4, MYD88 and NF-κB could protect mice from apoptosis induced by hypertonic stress. In terms of mechanism, NACL-induced NLRP3 inflammasome formation, caspase-1 activation and IL-1β maturation can be inhibited. Conclusion BMSCs treatment can reduce ROS and inflammation levels and alleviate dry eye by inhibiting TLR4/MYD88/NF-κBsignaling pathway.
Collapse
|
31
|
Wang F, Song Y, Liu P, Ma F, Peng Z, Pang Y, Hu H, Zeng L, Luo H, Zhang X. Rapamycin suppresses neuroinflammation and protects retinal ganglion cell loss after optic nerve crush. Int Immunopharmacol 2023; 119:110171. [PMID: 37060809 DOI: 10.1016/j.intimp.2023.110171] [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: 01/28/2023] [Revised: 03/26/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
Pyroptosis, an inflammasome-mediated mode of death, plays an important role in glaucoma. It has been shown that regulating the mTOR pathway can inhibit pyroptosis. Unfortunately, whether rapamycin (RAPA), a specific inhibitor of the mTOR pathway, can inhibit optic nerve crush (ONC)-induced pyroptosis to protect retinal ganglion cells (RGCs) has not been investigated. Our research aimed to confirm the effect of intravitreal injection of RAPA on RGCs. Furthermore, we used the ONC model to explore the underlying mechanisms. First, we observed that intravitreal injection of RAPA alleviated RGC damage induced by various types of injury. We then used the ONC model to further explore the potential mechanism of RAPA. Mechanistically, RAPA not only reduced the activation of glial cells in the retina but also inhibited retinal pyroptosis-induced expression of inflammatory factors such as nucleotide-binding oligomeric domain-like receptor 3 (NLRP3), apoptosis-associated speckle-like protein containing a CARD (ASC), N-terminal of gasdermin-D (GSDMD-N), IL-18 and IL-1β. Moreover, RAPA exerted protective effects on RGC axons, possibly by inhibiting glial activation and regulating the mTOR/ROCK pathway. Therefore, this study demonstrates a novel mechanism by which RAPA protects against glaucoma and provides further evidence for its application in preclinical studies.
Collapse
Affiliation(s)
- Feifei Wang
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Yuning Song
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Peiyu Liu
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Fangli Ma
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Zhida Peng
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Yulian Pang
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Ling Zeng
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Hongdou Luo
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China.
| |
Collapse
|
32
|
Shen J, Liang Y, Bi Z, Yin X, Chen C, Zhao X, Liu S, Li Y. Cyclosporin A improves the hyperosmotic response in an experimental dry eye model by inhibiting the HMGB1/TLR4/NF-κB signaling pathway. Exp Eye Res 2023; 229:109418. [PMID: 36806672 DOI: 10.1016/j.exer.2023.109418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/29/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Hyperosmolarity is closely related to dry eye disease (DED), which induces corneal epithelial cell structure and dysfunction leading to ocular surface inflammation. Cyclosporine A (CSA) is a cyclopeptide consisting of 11 deduced amino acids. It has an immunosuppressive effect and shows a vital function in inhibiting the inflammatory response. The mechanism of CSA in DED is still not entirely clear. This experiment aimed to investigate the possible mechanism of CSA in the hyperosmotic DED model. This study found that CSA can inhibit the transcript levels of DED high mobility group protein 1 (HMGB1), Toll-like receptor 4 (TLR4) and nuclear transcription factor κB (NF-κB) in signaling pathways. In addition, the study also found that 550 mOsm/L can induce the formation of DED models in vivo or in vitro. Furthermore, different concentrations of CSA have different effects on the expression of HMGB1 in human corneal epithelial cells under hyperosmotic stimulation, and high concentrations of CSA may increase the expression of HMGB1. In addition, CSA effectively reduced the corneal fluorescence staining score of the DE group and increased the tear volume of mice. Therefore, this experimental investigation might supply new evidence for the mechanism of CSA in DED, provide a potential new therapy for treating DED, and provide a theoretical basis for CSA treatment of DED.
Collapse
Affiliation(s)
- Jiachao Shen
- Department of Ophthalmology, Binzhou Medical College, Yantai, 264000, China; Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Yan Liang
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Zhaojing Bi
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xin Yin
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Chen Chen
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xinmei Zhao
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Shujun Liu
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China.
| | - Yuanbin Li
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China.
| |
Collapse
|
33
|
Yang X, Zuo X, Zeng H, Liao K, He D, Wang B, Yuan J. IFN-γ Facilitates Corneal Epithelial Cell Pyroptosis Through the JAK2/STAT1 Pathway in Dry Eye. INVESTIGATIVE OPTHALMOLOGY & VISUAL SCIENCE 2023; 64:34. [PMID: 36988949 PMCID: PMC10064915 DOI: 10.1167/iovs.64.3.34] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Purpose To investigate the effect of gamma interferon (IFN-γ) on corneal epithelial pyroptosis in an experimental dry eye (DE) model and explore the underlying molecular mechanisms. Methods Experimental DE was established in adult wild-type (WT) C57BL/6 mice and Ifng-knockout mice on a C57BL/6 background by subcutaneous injection of scopolamine (1.5 mg/0.3 mL, three times per day) and exposure to desiccating stress. An immortalized human corneal epithelial cell line (HCE-T) was treated with IFN-γ under hyperosmolar conditions. Corneal epithelial defects, tear production, and conjunctival goblet cells were detected by fluorescein sodium staining, the phenol red cotton test, and periodic acid-Schiff staining. The mRNA expression was measured by quantitative real-time PCR. Changes in protein expression were analyzed by Western blotting and immunofluorescence staining. Cell Counting Kit-8 and lactate dehydrogenase assays and in situ TUNEL staining were used to assess cell death. Results The expression of IFNG and its related genes was increased in the corneas of DE mice, whereas genetic deletion of Ifng ameliorated desiccating stress-induced dry eye symptoms. We further found that IFN-γ activated the JAK2/STAT1 signaling pathway inducing corneal epithelial pyroptosis. Topical application of a STAT1 inhibitor in vivo or siRNA targeting STAT1 in vitro suppressed pyroptosis of corneal epithelial cells. In addition, the production of reactive oxygen species (ROS) was elevated in DE, and a reduction in excessive ROS release prevented pyroptosis. Conclusions The increase in IFN-γ participates in the pathogenesis of dry eye and promotes corneal epithelial pyroptosis by activating the JAK2/STAT1 signaling pathway. Oxidative stress might be in downstream of JAK2/STAT1, thereby contributing to pyroptosis.
Collapse
|
34
|
Yu Y, Li K, Xue R, Liu S, Liu X, Wu K. A20 functions as a negative regulator of the lipopolysaccharide-induced inflammation in corneal epithelial cells. Exp Eye Res 2023; 228:109392. [PMID: 36717050 DOI: 10.1016/j.exer.2023.109392] [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: 09/16/2022] [Revised: 12/01/2022] [Accepted: 01/25/2023] [Indexed: 01/28/2023]
Abstract
A20, also called TNFAIP3, is a crucial regulator of inflammation in various diseases but has not evidenced its function in the cornea. We aimed to evaluate the existence and the functions of A20 in human corneal epithelial (HCE-T) cells. After being treated with lipopolysaccharide (LPS) in different concentrations or at separate times, cells were collected to analyze A20 expressions. We then constructed the A20 knockdown system by siRNA and the A20 overexpressing system by lentivirus transduction. Systems were further exposed to medium with or without LPS for indicated times. Next, we evaluated the production of inflammatory cytokines (IL-6 and IL-8) by qRT-PCR and ELISA. Also, the translocation of P65 and the phosphorylation of P65, P38 and JNK were observed in two systems. In addition, we used the nuclear factor kappa-B (NF-κB) antagonist TPCA-1 for the pretreatment in cells and then detected the A20 expressions. We found a low basal expression of A20 in HCE-T cells, and the expressions could be dose-dependently induced by LPS, peaking at 4 h in protein level after stimulation. Both the A20 knockdown and A20 overexpressing systems were confirmed to be effective. After the LPS treatment, productions of IL-6 and IL-8 were enhanced in the A20 knockdown system and reduced in the A20 overexpressing system. A20 reduced the translocation of P65 into the nucleus and the phosphorylation of P65, P38 and JNK. Furthermore, TPCA-1 pretreatment reduced the expression of A20 in cells. We concluded that A20 is a potent regulator for corneal epithelium's reaction to inflammation, and it thus is expected to be a potential therapy target for ocular surface diseases.
Collapse
Affiliation(s)
- Yubin Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Kunke Li
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Ran Xue
- Department of Ophthalmology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Sihao Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xiuping Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Kaili Wu
- 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
|
35
|
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
|
36
|
Mandell JT, de Rivero Vaccari JP, Sabater AL, Galor A. The inflammasome pathway: A key player in ocular surface and anterior segment diseases. Surv Ophthalmol 2023; 68:280-289. [PMID: 35798189 DOI: 10.1016/j.survophthal.2022.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 01/06/2023]
Abstract
Inflammasomes are multicomplex molecular regulators with an emerging importance in regulating ocular surface and anterior segment health and disease. Key components found in the eye include NF-κB, NLRP3, NLRC4, NLRP6, ASC, IL-1β, IL-18, and caspase-1. The role of NLRP1, NLRC4, AIM2, and NLRP3 inflammasomes in the pathogenesis of infectious ulcers, DED, uveitis, glaucoma, corneal edema, and other diseases is being studied with many developments. Attenuation of these diseases has been explored by blocking various molecules along the inflammasome pathway with agents like NAC, polydatin, calcitriol, glyburide, YVAD, and disulfiram. We provide a background on the inflammasome pathway as it relates to the ocular surface and anterior segment of the eye, discuss the role of inflammasomes in the above diseases in animals and humans, investigate new therapeutic targets, and explore the efficacy of new anti-inflammasome therapies.
Collapse
Affiliation(s)
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, FL, USA
| | | | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Ophthalmology, Miami Veterans Affairs (VA) Medical Center, Miami, FL, USA.
| |
Collapse
|
37
|
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
|
38
|
Scarpellini C, Ramos Llorca A, Lanthier C, Klejborowska G, Augustyns K. The Potential Role of Regulated Cell Death in Dry Eye Diseases and Ocular Surface Dysfunction. Int J Mol Sci 2023; 24:731. [PMID: 36614174 PMCID: PMC9820812 DOI: 10.3390/ijms24010731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
The research on new treatments for dry eye diseases (DED) has exponentially grown over the past decades. The increased prevalence of dry eye conditions, particularly in the younger population, has received much attention. Therefore, it is of utmost importance to identify novel therapeutical targets. Regulated cell death (RCD) is an essential process to control the biological homeostasis of tissues and organisms. The identification of different mechanisms of RCD stimulated the research on their involvement in different human pathologies. Whereas apoptosis has been widely studied in DED and included in the DED vicious cycle, the role of RCD still needs to be completely elucidated. In this review, we will explore the potential roles of different types of RCD in DED and ocular surface dysfunction. Starting from the evidence of oxidative stress and inflammation in dry eye pathology, we will analyse the potential therapeutic applications of the following principal RCD mechanisms: ferroptosis, necroptosis, and pyroptosis.
Collapse
Affiliation(s)
| | | | | | | | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2160 Antwerp, Belgium
| |
Collapse
|
39
|
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: 13] [Impact Index Per Article: 6.5] [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
|
40
|
Cathepsins Trigger Cell Death and Regulate Radioresistance in Glioblastoma. Cells 2022; 11:cells11244108. [PMID: 36552871 PMCID: PMC9777369 DOI: 10.3390/cells11244108] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Treatment of glioblastoma (GBM) remains very challenging, and it is particularly important to find sensitive and specific molecular targets. In this work, we reveal the relationship between the expression of cathepsins and radioresistance in GBM. We analyzed cathepsins (cathepsin B, cathepsin D, cathepsin L, and cathepsin Z/X), which are highly associated with the radioresistance of GBM by regulating different types of cell death. Cathepsins could be potential targets for GBM treatment.
Collapse
|
41
|
Yu X, Niu T, Liu C. Mechanism of LSD1 in oxygen-glucose deprivation/reoxygenation-induced pyroptosis of retinal ganglion cells via the miR-21-5p/NLRP12 axis. BMC Neurosci 2022; 23:63. [PMID: 36357913 PMCID: PMC9650888 DOI: 10.1186/s12868-022-00747-3] [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: 07/08/2022] [Accepted: 10/10/2022] [Indexed: 11/12/2022] Open
Abstract
Background Retinal ganglion cells (RGCs) are important retinal neurons that connect visual receptors to the brain, and lysine-specific demethylase 1 (LSD1) is implicated in the development of RGCs. This study expounded the mechanism of LSD1 in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced pyroptosis of RGCs. Methods Mouse RGCs underwent OGD/R exposure, and then RGC viability was examined using the cell counting kit-8 method. The mRNA levels of Caspase 1, the protein levels of NOD-like receptor family pyrin domain-containing 3 (NLRP3), N-terminal fragment of gasdermin D (GSDMD-N), and cleaved-Caspase1, and the concentrations of interleukin (IL)-1β and IL-18 were respectively examined. Subsequently, LSD1 expression was intervened to explore the underlying effect of LSD1 on OGD/R-induced pyroptosis of RGCs. Afterwards, the enrichments of LSD1 and histone H3 lysine 4 methylation (H3K4me) 1/2 on the microRNA (miR)-21-5p promoter were determined using chromatin-immunoprecipitation assay. And the binding interaction between miR-21-5p and NLRP12 was detected using dual-luciferase and RNA pull-down assays. Finally, the effects of miR-21-5p/NLRP12 on LSD1-mediated pyroptosis of RGCs were verified through functional rescue experiments. Results OGD/R treatment increased pyroptosis of RGCs and LSD1 expression. Silencing LSD1 declined levels of Caspase 1 mRNA, NLRP3, GSDMD-N, cleaved-Caspase1, IL-1β, and IL-18 and limited pyroptosis of OGD/R-treated RGCs. Mechanically, LSD1 suppressed miR-21-5p expression via demethylation of H3K4me2 on the miR-21-5p promoter to hamper the binding of miR-21-5p to NLRP12, and thereby increased NLRP12 expression. Silencing miR-21-5p or overexpressing NLRP12 facilitated OGD/R-induced pyroptosis of RGCs. Conclusion LSD1-mediated demethylation of H3K4me2 decreased miR-21-5p expression to increase NLRP12 expression, promoting pyroptosis of OGD/R-treated RGCs.
Collapse
|
42
|
Xie M, Wang H, Peng J, Qing D, Zhang X, Guo D, Meng P, Luo Z, Wang X, Peng Q. Acacetin protects against depression-associated dry eye disease by regulating ubiquitination of NLRP3 through gp78 signal. Front Pharmacol 2022; 13:984475. [PMID: 36299901 PMCID: PMC9588975 DOI: 10.3389/fphar.2022.984475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Dry eye disease (DED) is a multifactorial syndrome that commonly occurs with depression. However, therapies targeting depression-related dry eye disease are rare. In the current study, we studied the beneficial effect of a natural flavone, acacetin, in depression-associated dry eye disease by utilizing the chronic unpredictable mild stress (CUMS) depression model. Our data showed that acacetin improved the depressive behaviors in sucrose preference test (SPT), tail suspension test (TST) and forced swim test (FST); relieved the dry eye symptoms including corneal epithelial impairments, tear production decrease and goblet cell loss in CUMS mice. Acacetin also inhibited NOD-like receptor protein 3 (NLRP3) inflammasome expression levels and suppressed inflammatory responses via enhancing glycoprotein 78 (gp78)/Insulin induced gene-1 (Insig-1)-controlled NLRP3 ubiquitination in CUMS mice. Furthermore, knockdown of gp78 compromised acacetin-conferred protective efficacy in depression-related dry eye disease. In summary, our findings indicated that acacetin exerts beneficial effect in depression-associated dry eye disease, which is tightly related to gp78-mediated NLRP3 ubiquitination.
Collapse
Affiliation(s)
- Mingxia Xie
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hanqing Wang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jun Peng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Dongqin Qing
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xi Zhang
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Dongwei Guo
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Pan Meng
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhihong Luo
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiaoye Wang
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Xiaoye Wang, ; Qinghua Peng,
| | - Qinghua Peng
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Xiaoye Wang, ; Qinghua Peng,
| |
Collapse
|
43
|
Zuo X, Zeng H, Wang B, Yang X, He D, Wang L, Ouyang H, Yuan J. AKR1C1 Protects Corneal Epithelial Cells Against Oxidative Stress-Mediated Ferroptosis in Dry Eye. Invest Ophthalmol Vis Sci 2022; 63:3. [PMID: 36066316 PMCID: PMC9463717 DOI: 10.1167/iovs.63.10.3] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To evaluate the precise mode of cell death and to investigate the molecular mechanism underlying the initiation of inflammation in dry eye disease (DED). Methods C57BL/6 mice were injected with scopolamine subcutaneously and exposed to desiccating stress to establish a DED mouse model. An immortalized human corneal epithelial cell line (HCEC) was cultured under hyperosmolarity (500 mOsM). Protein expressions were measured using western blot assay and immunofluorescence staining. mRNA expression was analyzed by RNA-sequencing and quantitative RT-PCR. Transmission electron microscopy was used to observe the intracellular ultrastructure. Intracellular Fe2+ was detected by a FerroOrange fluorescent probe. Flow cytometry was used to evaluate the cellular reactive oxygen species and lipid peroxidation. Results Marked changes in ferroptosis-related markers expression, intracellular iron accumulation, and lipid peroxidation were observed in corneal epithelial cells of DED models. When excessive oxidative stress was suppressed, ferroptosis induced by hyperosmolarity in HCECs was restrained, as indicated by decreased iron content and lipid peroxidation levels. Moreover, AKR1C1 was upregulated by the activation of NRF2 in HCECs under hyperosmolarity. When AKR1C1 was knocked down, cell viability was decreased, accompanied by increased lipid peroxidation, whereas overexpression of AKR1C1 produced the opposite results. It was observed consistently that corneal defects and the inflammatory response were promoted after inhibition of AKR1C1 in vivo. Conclusions Excessive oxidative stress-induced ferroptosis participates in DED pathogenesis. The expression of AKR1C1 is triggered by NRF2 to decrease ferroptosis-induced cell damage and inflammation in HCECs. These findings may provide potential makers targeting ferroptosis and AKR1C1 for DED therapy.
Collapse
Affiliation(s)
- Xin Zuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Hao Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Bowen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Li Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Hong Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
44
|
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: 48] [Impact Index Per Article: 16.0] [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
|
45
|
Yeoh WJ, Vu VP, Krebs P. IL-33 biology in cancer: An update and future perspectives. Cytokine 2022; 157:155961. [PMID: 35843125 DOI: 10.1016/j.cyto.2022.155961] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/03/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 family of cytokines that is constitutively expressed in the nucleus of epithelial, endothelial and fibroblast-like cells. Upon cell stress, damage or necrosis, IL-33 is released into the cytoplasm to exert its prime role as an alarmin by binding to its specific receptor moiety, ST2. IL-33 exhibits pleiotropic function in inflammatory diseases and particularly in cancer. IL-33 may play a dual role as both a pro-tumorigenic and anti-tumorigenic cytokine, dependent on tumor and cellular context, expression levels, bioactivity and the nature of the inflammatory environment. In this review, we discuss the differential contribution of IL-33 to malignant or inflammatory conditions, its multifaceted effects on the tumor microenvironment, while providing possible explanations for the discrepant findings described in the literature. Additionally, we examine the emerging and divergent functions of IL-33 in the nucleus, and aspects of IL-33 biology that are currently under-addressed.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Vivian P Vu
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
46
|
Tovar A, Gomez A, Serrano A, Blanco MP, Galor A, Swaminathan SS, de Rivero Vaccari JP, Sabater AL. Role of Caspase-1 as a Biomarker of Ocular Surface Damage. Am J Ophthalmol 2022; 239:74-83. [PMID: 35151638 DOI: 10.1016/j.ajo.2022.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE To examine the potential of caspase-1 as a biomarker for ocular surface damage. DESIGN Cross-sectional study. METHODS A total of 113 tear samples (64 subjects) were analyzed. Sixty-one samples were from individuals with dry eye disease (DED), defined as Ocular Surface Disease Index (OSDI) ≥13 and/or corneal staining (CS) ≥3; 32 were from individuals who used glaucoma medication, irrespective of DED metrics; and 20 were from controls (CS <3 and OSDI <13). All individuals completed a medical history form and underwent an ocular surface assessment. Protein levels of caspase-1 were determined by enzyme-linked immunosorbent assay off Schirmer's strips. The primary analysis compared caspase-1 levels in individuals with signs of ocular surface damage (CS ≥3) in both case groups and controls. Secondary correlational analyses were conducted to examine relationships between caspase-1 levels and ocular signs and symptoms. Finally, area under the curve (AUC) analyses were performed to examine relationships between inflammatory markers and CS. RESULTS The mean age of the population was 58±18 years; 70% were female. Tear samples from individuals with ocular surface damage presented higher caspase-1 levels than the control group. Caspase-1 levels showed a moderate positive correlation with CS (Spearman r = 0.31; P = .001) and eye redness (Spearman r = 0.39; P = .004), and a negative correlation with Schirmer's (Spearman r = -0.46; P < .001) and tear break-up time (Spearman r = -0.33; P = .0006). Caspase-1 showed higher sensitivity and AUC for detecting ocular surface damage than InflammaDry, and its expression was not affected by anti-inflammatory agents. CONCLUSION Caspase-1 levels were higher in the tears of individuals with ocular surface damage, suggesting its potential to be used as a biomarker and/or therapeutic target.
Collapse
Affiliation(s)
- Arianna Tovar
- From the Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA (A.T, A.G, A.S, M.P.B, S.S.S, A.L.S)
| | - Angela Gomez
- From the Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA (A.T, A.G, A.S, M.P.B, S.S.S, A.L.S)
| | - Andres Serrano
- From the Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA (A.T, A.G, A.S, M.P.B, S.S.S, A.L.S)
| | - Maricarmen Perez Blanco
- From the Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA (A.T, A.G, A.S, M.P.B, S.S.S, A.L.S)
| | - Anat Galor
- From the Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA (A.T, A.G, A.S, M.P.B, S.S.S, A.L.S)
| | - Swarup S Swaminathan
- From the Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA (A.T, A.G, A.S, M.P.B, S.S.S, A.L.S)
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA (J.P.R.V)
| | - Alfonso L Sabater
- From the Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA (A.T, A.G, A.S, M.P.B, S.S.S, A.L.S).
| |
Collapse
|
47
|
Li J, Yang K, Pan X, Peng H, Hou C, Xiao J, Wang Q. Long Noncoding RNA MIAT Regulates Hyperosmotic Stress-Induced Corneal Epithelial Cell Injury via Inhibiting the Caspase-1-Dependent Pyroptosis and Apoptosis in Dry Eye Disease. J Inflamm Res 2022; 15:3269-3283. [PMID: 35676970 PMCID: PMC9169976 DOI: 10.2147/jir.s361541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/20/2022] [Indexed: 12/21/2022] Open
Abstract
Purpose The biological role and mechanism of long noncoding RNA (lncRNA) myocardial infarction-associated transcript (MIAT) in dry eye remain to be illustrated. Pyroptosis is a noticeable form of inflammatory activation, which is characteristic of gasdermin D (GSDMD)-driven cell death. The present study was designed to explore the role of MIAT in pyroptosis and apoptosis induced by hyperosmolarity stress (HS) in human corneal epithelial cells (HCECs). Methods HCECs were cultured in 70–120 mM hyperosmotic medium for 24 h to create a dry eye model in vitro. The level of the pyroptosis marker GSDMD was measured, and the cell inflammatory response was evaluated by detecting IL-1β and IL-18 levels. Exogenous caspase-1 inhibitor Ac-YVAD-CHO was used. The pyroptosis in HCECs was examined by caspase-1 activity, immunofluorescent staining, and Western blotting. Flow cytometry was performed to test the apoptosis rate of HCECs. Cell migration and proliferation were detected. The expression of the lncRNA MIAT in HCECs was detected by quantitative real-time PCR. MIAT was knocked down by small interfering RNA (siRNA) transfection. The effects of caspase-1 inhibition on pyroptosis, apoptosis, migration, and proliferation were observed. Results HS promoted pyroptosis in HCECs by elevating caspase-1, GSDMD, and the active cleavage of GSDMD (N-terminal domain, N-GSDMD), and increased the release of IL-1β, IL-18, LDH and the rate of apoptosis, with reduced cell migration. These changes were prevented by the inhibition of caspase-1. The expression of MIAT was significantly increased in HCECs exposed to a hyperosmotic medium. Silencing MIAT increased the expression of GSDMD, caspase-1, and inflammatory chemokines IL-1β and IL-18, and promoted apoptosis while inhibiting migration and proliferation in HCECs. Conclusion The lncRNA MIAT is involved in HS-induced pyroptosis and apoptosis and the inflammatory response of HCECs and provides a new understanding of the pathogenesis of dry eye.
Collapse
Affiliation(s)
- Jinjian Li
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Kun Yang
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Xinghui Pan
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Hui Peng
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Chenting Hou
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Jie Xiao
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Qing Wang
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
- Correspondence: Qing Wang, Ophthalmology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266500, People’s Republic of China, Tel +86 17853290636, Fax +86 532 82911747, Email
| |
Collapse
|
48
|
Roles and Mechanisms of Regulated Necrosis in Corneal Diseases: Progress and Perspectives. J Ophthalmol 2022; 2022:2695212. [PMID: 35655803 PMCID: PMC9152437 DOI: 10.1155/2022/2695212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/24/2022] [Accepted: 05/09/2022] [Indexed: 11/21/2022] Open
Abstract
Regulated necrosis is defined as cell death characterized by loss of the cell membrane integrity and release of the cytoplasmic content. It contributes to the development and progression of some diseases, including ischemic stroke injury, liver diseases, hypertension, and cancer. Various forms of regulated necrosis, particularly pyroptosis, necroptosis, and ferroptosis, have been implicated in the pathogenesis of corneal disease. Regulated necrosis of corneal cells enhances inflammatory reactions in the adjacent corneal tissues, leading to recurrence and aggravation of corneal disease. In this review, we summarize the molecular mechanisms of pyroptosis, necroptosis, and ferroptosis in corneal diseases and discuss the roles of regulated necrosis in inflammation regulation, tissue repair, and corneal disease outcomes.
Collapse
|
49
|
Chen X, Wu J, Lin X, Wu X, Yu X, Wang B, Xu W. Tacrolimus Loaded Cationic Liposomes for Dry Eye Treatment. Front Pharmacol 2022; 13:838168. [PMID: 35185587 PMCID: PMC8855213 DOI: 10.3389/fphar.2022.838168] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/13/2022] [Indexed: 12/27/2022] Open
Abstract
Eye drops are ophthalmic formulations routinely used to treat dry eye. However, the low ocular bioavailability is an obvious drawback of eye drops owing to short ocular retention time and weak permeability of the cornea. Herein, to improve the ocular bioavailability of eye drops, a cationic liposome eye drop was constructed and used to treat dry eye. Tacrolimus liposomes exhibit a diameter of around 300 nm and a surface charge of +30 mV. Cationic liposomes could interact with the anionic ocular surface, extending the ocular retention time and improving tacrolimus amount into the cornea. The cationic liposomes notably prolonged the ocular retention time of eye drops, leading to an increased tacrolimus concentration in the ocular surface. The tacrolimus liposomes were also demonstrated to reduce reactive oxygen species and dry eye-related inflammation factors. The use of drug-loaded cationic liposomes is a good formulation in the treatment of ocular disease; the improved ocular retention time and biocompatibility give tremendous scope for application in the treatment of ocular disease, with further work in the area recommended.
Collapse
Affiliation(s)
- Xiang Chen
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jicheng Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Xueqi Lin
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xingdi Wu
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xuewen Yu
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ben Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Wen Xu
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
50
|
Wang B, Zeng H, Zuo X, Yang X, Wang X, He D, Yuan J. TLR4-Dependent DUOX2 Activation Triggered Oxidative Stress and Promoted HMGB1 Release in Dry Eye. Front Med (Lausanne) 2022; 8:781616. [PMID: 35096875 PMCID: PMC8793023 DOI: 10.3389/fmed.2021.781616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/09/2021] [Indexed: 12/30/2022] Open
Abstract
Dry eye disease (DED) is one of the most common ocular surface diseases worldwide. DED has been characterized by excessive accumulation of reactive oxygen species (ROS), following significant corneal epithelial cell death and ocular surface inflammation. However, the key regulatory factor remains unclear. In this study, we tended to explore whether DUOX2 contributed to DED development and the underlying mechanism. Human corneal epithelial (HCE) cells were treated with hyperosmolarity, C57BL/6 mice were injected of subcutaneous scopolamine to imitate DED. Expression of mRNA was investigated by RNA sequencing (RNA-seq) and quantitative real-time PCR (qPCR). Protein changes and distribution of DUOX2, high mobility group box 1 (HMGB1), Toll-like receptor 4 (TLR4), and 4-hydroxynonenal (4-HNE) were evaluated by western blot assays and immunofluorescence. Cell death was assessed by Cell Counting Kit-8 (CCK8), lactate dehydrogenase (LDH) release, and propidium iodide (PI) staining. Cellular ROS levels and mitochondrial membrane potential (MMP) were analyzed by flow cytometry. RNA-seq and western blot assay indicated a significant increase of DUOX2 dependent of TLR4 activation in DED both in vitro and in vivo. Immunofluorescence revealed significant translocation of HMGB1 within corneal epithelial cells under hyperosmolar stress. Interestingly, after ablated DUOX2 expression by siRNA, we found a remarkable decrease of ROS level and recovered MMP in HCE cells. Moreover, knockdown of DUOX2 greatly inhibited HMGB1 release, protected cell viability and abolished inflammatory activation. Taken together, our data here suggest that upregulation of DUOX2 plays a crucial role in ROS production, thereafter, induce HMGB1 release and cell death, which triggers ocular surface inflammation in DED.
Collapse
Affiliation(s)
- Bowen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - Hao Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - Xin Zuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - Xue Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - Xiaoran Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|