1
|
Wu JS, Chu PY, Hsu WY, Chuang TH, Yu YC, Pan YC, Lin YT, Tang CH, Lee CL, Wu YC. Fractionation and identification of ocular protective compounds from kochiae fructus against oxidative damage in retinal pigment epithelium cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119328. [PMID: 39793774 DOI: 10.1016/j.jep.2025.119328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/26/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kochiae Fructus, the ripe fruit of Kochia scoparia, is a traditional Chinese medicine commonly used to treat eye discomforts and vision problems. Although Kochiae Fructus is mentioned in many classical Chinese medical texts, its protective effects and the roles of its active phytochemicals in eye treatment still lack scientific exploration. AIM OF THE STUDY This study aimed to clarify the protective effects and identify the active fractions and compounds of Kochiae Fructus against oxidative stress-induced retinal pigment epithelium (RPE) cell death. MATERIALS AND METHODS Liquid-liquid partitioning was employed to prepare active fractions. Silica gel, RP-18, and Sephadex™ LH-20 gels were used as stationary phases to purify the compounds through column chromatography. Cell models were established by treating ARPE-19 (RPE cell line) with hydrogen peroxide (H₂O₂) and tert-butyl hydroperoxide (TBHP), respectively. RESULTS The methanol aqueous fraction from ethyl acetate-soluble extract (KSEM) alleviated oxidant-induced RPE cell death in a dose-dependent manner and activated the Nrf2/HO-1 pathway, a critical defense mechanism against oxidative damage. The isolated flavonols in fraction KSEM, hyperoside, quercetin, and kaempferol, reduced oxidant-induced cell mortality, with quercetin showing the strongest effect. Moreover, combination treatment revealed that hyperoside and kaempferol synergistically enhanced the protective effects of quercetin, making the combined treatment more efficient than quercetin alone. CONCLUSIONS The protective effects of Kochiae Fructus against oxidative damage in RPE cell was validated with the KSEM fraction. The compound quercetin was identified as the important contributor. However, the synergistic effect of the hyperoside, quercetin, and kaempferol mixture may play a dominant role in the protective action of Kochiae Fructus. Overall, these findings highlight the potential of Kochiae Fructus and its flavonol mixture as a basis for developing treatments targeting ocular diseases associated with oxidative damage in retina.
Collapse
Affiliation(s)
- Jian-Sheng Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 0402, Taiwan; Department of Ophthalmology, Changhua Christian Hospital, Changhua, 50006, Taiwan.
| | - Pei-Yi Chu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan; Master Program of Pharmaceutical Manufacture, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan.
| | - Wei-Yu Hsu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan.
| | - Ta-Hsien Chuang
- School of Pharmacy, China Medical University, Taichung, 406040, Taiwan.
| | - Ying-Chun Yu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan.
| | - Yi-Cheng Pan
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan.
| | - Yi-Ting Lin
- Department of Cosmeceutics, China Medical University, Taichung, 40604, Taiwan.
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 0402, Taiwan; Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, 40402, Taiwan.
| | - Chia-Lin Lee
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Department of Cosmeceutics, China Medical University, Taichung, 40604, Taiwan.
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan; Master Program of Pharmaceutical Manufacture, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
2
|
Scalici A, Miller-Fleming TW, Shuey MM, Baker JT, Betti M, Hirbo J, Knapik EW, Cox NJ. Gene and phenome-based analysis of the shared genetic architecture of eye diseases. Am J Hum Genet 2025; 112:318-331. [PMID: 39879988 DOI: 10.1016/j.ajhg.2025.01.004] [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: 06/21/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025] Open
Abstract
While many eye disorders are linked through defects in vascularization and optic nerve degeneration, genetic correlation studies have yielded variable results despite shared features. For example, glaucoma and myopia both share optic neuropathy as a feature, but genetic correlation studies demonstrated minimal overlap. By leveraging electronic health record (EHR) resources that contain genetic variables such as genetically predicted gene expression (GPGE), researchers have the potential to improve the identification of shared genetic drivers of disease by incorporating knowledge of shared features to identify disease-causing mechanisms. In this study, we examined shared genetic architecture across eye diseases. Our gene-based approach used transcriptome-wide association methods to identify shared transcriptomic profiles across eye diseases within BioVU, Vanderbilt University Medical Center's (VUMC's) EHR-linked biobank. Our phenome-based approach leveraged phenome-wide association studies (PheWASs) to identify eye disease comorbidities. Using the beta estimates from the significantly associated comorbidities, we constructed a phenotypic risk score (PheRS) representing a weighted sum of an individual's eye disease comorbidities. This PheRS is predictive of eye disease status and associated with the altered GPGE of significant genes in an independent population. The implementation of both gene- and phenome-based approaches can expand genetic associations and shed greater insight into the underlying mechanisms of shared genetic architecture across eye diseases.
Collapse
Affiliation(s)
- Alexandra Scalici
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tyne W Miller-Fleming
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Megan M Shuey
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James T Baker
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Betti
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jibril Hirbo
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ela W Knapik
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
3
|
Zeng Y, Liu Y, Chen X, Kenny J, Rong R, Xia X. Global, regional, and national burden of blindness and vision loss attributable to smoking from 1990 to 2021, and forecasts to 2030: findings from the Global Burden of Disease Study 2021. BMC Public Health 2025; 25:440. [PMID: 39905351 PMCID: PMC11792669 DOI: 10.1186/s12889-025-21573-2] [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: 10/23/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
OBJECTIVE This study aims to systematically elucidate the burden of blindness and vision loss (BVL) attributable to smoking from 1990 to 2021 and to forecast the trends in BVL burden over the next decade. METHODS We extracted data on years lived with disability (YLDs) and age-standardized YLDs rate (ASYR) related to blindness and vision loss (BVL) caused by smoking, including cataracts and age-related macular degeneration (AMD), from the Global Burden of Disease (GBD) database for the years 1990 to 2021. These data were disaggregated by age, gender, sociodemographic index (SDI), region, and country. Temporal trends in the burden of smoking-induced BVL were evaluated by calculating the average annual percentage changes (AAPCs). RESULTS BVL attributable to smoking presents a significant disease burden, with global BVL-related YLDs attributable to smoking increasing from 1990 to 2021, while ASYR showed a declining trend. In 2021, the global BVL-related YLDs and ASYR attributable to smoking were estimated at 284.03 thousand and 3.27 per 100,000 population. The ASYR for cataract and AMD are 2.60 and 0.68 per 100,000, respectively. The burden was notably higher in males than females, highlighting significant gender disparities. Regionally, the highest burdens were observed in South Asia, Southeast Asia, and North Africa. It is expected that the number of global BVL-related YLDs will increase further by 2030. CONCLUSION Smoking has imposed a substantial disease burden on BVL over the past three decades. The burden is predominantly concentrated among males, particularly older individuals and those in low to middle-SDI regions. Moreover, the burden of smoking-induced BVL is expected to continue improving over the next decade.
Collapse
Affiliation(s)
- Yi Zeng
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Key Specialty of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Yixian Liu
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Key Specialty of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xingyi Chen
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Key Specialty of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jennifer Kenny
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Key Specialty of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Rong Rong
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China.
- National Clinical Key Specialty of Ophthalmology, Changsha, Hunan, 410008, P.R. China.
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China.
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China.
- National Clinical Key Specialty of Ophthalmology, Changsha, Hunan, 410008, P.R. China.
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China.
| |
Collapse
|
4
|
Wu G, Notomi S, Xu Z, Fukuda Y, Maehara Y, Tao Y, Zhao H, Ishikawa K, Murakami Y, Hisatomi T, Sonoda KH. Lamp2 Deficiency Enhances Susceptibility to Oxidative Stress-Induced RPE Degeneration. Invest Ophthalmol Vis Sci 2025; 66:2. [PMID: 39898910 PMCID: PMC11798339 DOI: 10.1167/iovs.66.2.2] [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: 08/29/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Purpose Autophagy and lysosomal degradation are vital processes that protect cells from oxidative stress. This study investigated the role of lysosome-associated membrane protein 2 (Lamp2), a lysosomal protein essential for autophagosome maturation and lysosome biogenesis, in maintaining retinal health under oxidative stress. Methods To induce oxidative stress, young Lamp2 knockout (KO) and wild-type mice received an intravenous injection of a low dose (10 mg/kg) of sodium iodate (NaIO3). We examined retinal histopathology and morphological changes in the RPE. The involvement of resident microglia or infiltrating macrophages was assessed using immunostaining, flow cytometry, and real-time PCR for chemokines and cytokines. Results After administering a low-dose NaIO3, Lamp2 KO mice showed significant RPE degeneration, whereas wild-type mice had minimal damage. Histological analysis and electron microscopy revealed significant thinning of the outer nuclear layer and loss of RPE epithelial polarity in Lamp2 KO mice. Additionally, there was a significant increase in ionized calcium-binding adaptor molecule 1-positive microglia and macrophages in the outer retina. Early proliferation of CD45lowMHC-IIlow resident microglia was followed by the infiltration of CD45highLy6Chigh monocytes and the engraftment of CD11b+CD45high monocyte-derived macrophages. Transcript levels of monocyte chemoattractant protein 1, macrophage inflammatory protein 1β, Il- 1β, and Il-6 also increased in the retinas of Lamp2 KO mice. Furthermore, pretreatment with the macrophage-depleting agent clodronate prevented NaIO3-induced RPE degeneration and macrophage infiltration in Lamp2 KO mice. Conclusions Lamp2 deficiency, when combined with oxidative stress, leads to RPE degeneration in vivo. Lysosomal dysfunction also promotes macrophage engraftment and triggers neurotoxic inflammation.
Collapse
Affiliation(s)
- Guannan Wu
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoji Notomi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ziming Xu
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yosuke Fukuda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Maehara
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yan Tao
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Huanyu Zhao
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keijiro Ishikawa
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshio Hisatomi
- Department of Ophthalmology, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Li S, Su D, Hu S, Hu Q, Sun D. Epigallocatechin gallate ameliorates retinal pigment epithelial cell damage via the CYFIP2 /AKT pathway. Toxicol Appl Pharmacol 2025; 495:117124. [PMID: 39667565 DOI: 10.1016/j.taap.2024.117124] [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: 06/25/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 12/14/2024]
Abstract
Age-related macular degeneration (AMD) is a representative age-related ophthalmic disease, and the pathogenesis of AMD remains unclear. This research intended to determine whether epigallocatechin gallate (EGCG) could alleviate the progression of AMD and the possible mechanism. We constructed three groups of mice (young, aged, and EGCG), and HE and TUNEL staining of retinal tissues was performed to observe the structural changes in the retinal pigment epithelial (RPE) layer and the level of apoptosis, respectively. Through RNA-Sequencing analysis of retinal tissues and by RT-qPCR, GO, KEGG, and literature analyses, we identified cytoplasmic fragile X mental retardation 1-interacting protein 2 (CYFIP2) as a possible effector gene for EGCG action and validated its role by immunofluorescent and western blotting experiments. The CCK-8 and Hoechst 33342 apoptosis assays, and western blotting and qRT-PCR assays showed that EGCG reduced hydrogen peroxide (H2O2)-induced apoptosis in adult human RPE (ARPE-19) cells, and the expression of Cyfip2 was changed accordingly. RNA interference analysis indicated that Cyfip2 knockdown alleviated H2O2-induced ARPE apoptosis, while its overexpression weakened EGCG's protective effect. Western blot analysis showed that Cyfip2 mediated the anti-apoptotic effect of EGCG by modulating the level of protein kinase B (Akt) phosphorylation in ARPE cells, and the activation level of phosphorylated AKT (p-AKT Ser473) in retinal tissue of the EGCG-fed group was higher than that of the aged group. Taken together, this study suggests that EGCG plays a protective role in the development of AMD and the apoptosis of ARPE cells through the Cyfip2/AKT pathway.
Collapse
Affiliation(s)
- Sijia Li
- Harbin Medical University, Harbin 150086, China; Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Dongmei Su
- Department of Genetics, NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning, Health Department, Beijing 100081, China; Graduate School, Peking Union Medical College, Beijing 100081, China
| | - Shanshan Hu
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Qiang Hu
- Harbin Medical University, Harbin 150086, China
| | - Dawei Sun
- Department of Ophthalmology, the second affiliated hospital of Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
6
|
Wei MH, Li JX, Mi J, Wang Q, Xu F, Xu C. Associations between co-exposure to multiple heavy metals and age-related macular degeneration: A cross-sectional study. J Trace Elem Med Biol 2025; 87:127573. [PMID: 39643952 DOI: 10.1016/j.jtemb.2024.127573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/06/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND & AIMS Accumulating evidence suggests that exposure to single heavy metal can facilitate the progression of age-related macular degeneration (AMD). However, the effects of exposure to mixtures of heavy metals on AMD remain largely unexplored. This study aims to investigate both the joint and individual impacts of arsenic (As), mercury (Hg), cadmium (Cd), and lead (Pb) on AMD within a co-exposure framework. METHODS Data from subjects participating the US National Health and Nutrition Examination Survey (NHANES, 2005-2008) were analyzed. Concentrations of As, Hg, Cd, and Pb were determined in urine by inductively coupled plasma dynamic reaction cell mass spectrometry (ICP-DRC-MS) for As and Hg, and inductively coupled plasma mass spectrometry (ICP-MS) for Cd and Pb. The weighted quantile sum (WQS) and Bayesian kernel machine regression (BKMR) models were employed to assess the effects of heavy metal mixtures on AMD risk. RESULTS Both WQS and BKMR analyses consistently revealed a significant overall association between heavy metal mixtures and the risk of all types of AMD. The combined effect was more evident among patients with early AMD compared to those with late AMD. Cd and Hg were the main contributors driving these combined effects within the context of metal mixtures. Elevated urinary levels of Cd were positively correlated with an increased risk for all types as well as early AMD. Higher exposure to Hg corresponded with an elevated risk for early AMD. Furthermore, BKMR analysis indicated that the influence of Cd on early AMD exhibited a non-linear pattern. CONCLUSIONS Our findings suggest that co-exposure to As, Hg, Cd, and Pb is associated with an elevated risk for developing AMD, particularly in its early stages. Furthermore, excessive exposure to Cd and Hg has been identified as key contributing factors in this process.
Collapse
Affiliation(s)
- Mu-Hong Wei
- Department of Epidemiology and Statistics, School of Public Health, Bengbu Medical University, No. 2600 Donghai Avenue, Bengbu 233000, China
| | - Ju-Xiao Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Jing Mi
- Department of Epidemiology and Statistics, School of Public Health, Bengbu Medical University, No. 2600 Donghai Avenue, Bengbu 233000, China
| | - Qing Wang
- The First People's Hospital of Guiyang City, No.97 Boai Road, Nanming District, Guiyang City, China
| | - Feng Xu
- Biomedical Research Core Facilities, Westlake University, Hangzhou 310027, China
| | - Che Xu
- Department of ophthalmology, the First Affiliated Hospital of Bengbu Medical University, No.287 Changhuai Road, Bengbu 233000, China.
| |
Collapse
|
7
|
Wang X, Zhang C, Jiang H. Association of dietary inflammatory index with ocular diseases: a population-based cross-sectional study. Eur J Med Res 2025; 30:62. [PMID: 39891276 PMCID: PMC11783699 DOI: 10.1186/s40001-025-02294-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Our research was designed to investigate the relationship between dietary inflammatory index (DII) and risk of ocular diseases, including glaucoma, cataract, age-related macular degeneration (ARMD), and diabetic retinopathy. METHODS We used the National Health and Nutrition Examination Survey (NHANES) data from 2005 to 2008 to conduct this study. The correlation between DII and risk of ocular diseases was examined using weighted multivariable logistic regression analysis, restricted cubic spline (RCS) plots, and subgroup analysis. RESULTS In total, 2885 participants from the NHANES database were included. The DII scores were divided into four group: Q1 (- 4.438-0.386), Q2 (0.387-1.848), Q3 (1.849-3.073), and Q4 (3.074-4.970). RCS shown that there was a U-shaped correlation between DII and prevalence of glaucoma, cataract, ARMD, and diabetic retinopathy. After adjusting for underlying confounding variables, compared to Q1 group, the odd ratios (ORs) with 95 percent confidence intervals (CIs) for glaucoma, cataract, ARMD, and diabetic retinopathy across the quartiles were [0.97 (0.54, 1.75), 1.20 (0.68, 2.11), and 1.29 (0.73, 2.30)], [0.87 (0.56, 1.35), 1.12 (0.73, 1.73), and 1.16 (0.75, 1.80)], [0.85 (0.53, 1.36), 0.66 (0.40, 1.09), and 0.97 (0.61, 1.56)] and [0.86 (0.63, 1.18), 0.89 (0.65, 1.22), and 1.04 (0.75, 1.45)] for DII, respectively. CONCLUSIONS Reducing the intake of pro-inflammatory foods may be an effective measure to prevent the onset of ocular disease, including glaucoma, cataract, ARMD, and diabetic retinopathy. However, eating only anti-inflammatory foods is not the best choice.
Collapse
Affiliation(s)
- Xue Wang
- Department of Ophthalmology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Zhenhua East Road, Lianyungang, 222000, Jiangsu, China.
| | - Can Zhang
- Department of Ophthalmology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Zhenhua East Road, Lianyungang, 222000, Jiangsu, China
| | - Haitao Jiang
- Department of Ophthalmology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Zhenhua East Road, Lianyungang, 222000, Jiangsu, China
| |
Collapse
|
8
|
Wang SW, Li P, Liu SY, Huang DL, Zhang SJ, Zeng XX, Lan T, Mao KL, Gao Y, Cheng YF, Shen Q, Ruan YP, Mao ZJ. Astragaloside IV inhibits retinal pigment epithelial cell senescence and reduces IL-1β mRNA stability by targeting FTO-mediated m 6A methylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156408. [PMID: 39848020 DOI: 10.1016/j.phymed.2025.156408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Resistance to senescence in retinal pigment epithelial (RPE) cells can delay the progression of age-related macular degeneration (AMD). However, the mechanisms underlying RPE cell senescence remain inadequately understood, and effective therapeutic strategies are lacking. While astragaloside IV (Ast) has demonstrated anti-aging properties, its specific effects on RPE cell senescence and potential mechanisms are not yet fully clarified. PURPOSE This study aimed to explore the impacts of Ast on RPE cell senescence and to uncover the molecular mechanisms involved. METHODS The therapeutic efficacy of Ast was assessed using sodium iodate (NaIO3)-induced adult retinal pigment epithelial cell line 19 (ARPE-19) cell models and an AMD mouse model. To investigate the mechanisms by which Ast mitigated RPE cell senescence, RNA sequencing (RNA-seq), drug affinity responsive target stability-mass spectrometry (DARTS-MS), cellular thermal shift assay (CETSA), reverse transcription quantitative PCR (RT-qPCR), as well as western blotting were conducted. RESULTS Ast significantly inhibited NaIO3-treated ARPE-19 cell senescence and protected against NaIO3-induced AMD in mice. RNA-seq analysis revealed that Ast significantly attenuated inflammation-related signaling pathways and reduced the mRNA levels of interleukin-1 beta (IL-1β). Specifically, Ast decreased the stability of IL-1β mRNA while enhancing its N6-methyladenosine (m6A) methylation. Furthermore, Ast directly interacted with fat mass and obesity-associated protein (FTO). Knockdown or pharmacological inhibition of FTO mitigated the senescence and IL-1β expression in NaIO3-treated ARPE-19 cells. FTO was essential for Ast to inhibit cellular senescence and IL-1β expression. Additionally, inhibition or knockdown of FTO conferred also provided resistance to AMD in the murine model. CONCLUSION Our results indicated that Ast significantly attenuated RPE cell senescence and showed anti-AMD properties. FTO was demonstrated to be a promising therapeutic target for AMD treatment. These findings may provide a deeper understanding of the molecular mechanisms underlying RPE cell senescence in AMD and offer potential strategies for its prevention and management.
Collapse
Affiliation(s)
- Si-Wei Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| | - Ping Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shi-Yu Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - De-Lian Huang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Si-Jia Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xi-Xi Zeng
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Tian Lan
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Kai-Li Mao
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Yuan Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi-Fan Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qing Shen
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Ye-Ping Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Zhu-Jun Mao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Ophthalmology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| |
Collapse
|
9
|
Yeh WJ, Yan C, Wu CH. Photoprotective Effects of Phytochemicals on Blue Light-Induced Retinal Damage: Current Evidence and Future Perspectives. Nutrients 2025; 17:331. [PMID: 39861461 PMCID: PMC11768023 DOI: 10.3390/nu17020331] [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: 12/20/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
The widespread use of light-emitting diodes (LEDs) has increased blue light (BL) exposure, raising concerns about its potential adverse effects on ocular health. Prolonged exposure to BL has been implicated in the pathogenesis of various retinal disorders, including age-related macular degeneration (AMD), primarily through mechanisms involving oxidative stress and inflammation mediated by the overproduction of reactive oxygen species (ROS). This review synthesizes current evidence on the photoprotective properties of dietary bioactive compounds, (e.g., anthocyanins, curcumin, quercetin, myricetin, and resveratrol), with a focus on their potential to mitigate BL-induced retinal damage. Accumulating research suggests that dietary antioxidants, particularly polyphenols, may offer photoprotective benefits. These phytochemicals act by neutralizing ROS and enhancing the retina's endogenous antioxidant capacity. Based on these findings, this review advocates for a food-first approach in future investigations, emphasizing the development of evidence-based dietary recommendations to bolster retinal health and mitigate the risk of BL-related ocular diseases. Considering the current lack of empirical clinical studies examining the impact of BL on human ocular health, future research in the field of BL hazard should prioritize two key approaches: conducting large-scale epidemiological dietary surveys and implementing clinical trials on functional ingredients that have demonstrated beneficial effects against photodamage in preclinical animal studies.
Collapse
Affiliation(s)
| | | | - Chi-Hao Wu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan; (W.-J.Y.); (C.Y.)
| |
Collapse
|
10
|
Jiang X, Liu C, Zhang Q, Lv Y, Lu C, Su W, Zhou J, Zhang H, Gong H, Liu Y, Yuan S, Chen Y, Qu D. Strategic delivery of rapamycin and ranibizumab with intravitreal hydrogel depot disrupts multipathway-driven angiogenesis loop for boosted wAMD therapy. J Control Release 2025; 377:239-255. [PMID: 39528095 DOI: 10.1016/j.jconrel.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Autophagic dysfunction-induced deterioration of the retinal microenvironment drives the progression of wet age-related macular degeneration (wAMD). The efficacy of single-target anti-VEGF antibodies in treating wAMD has long been suboptimal due to the intricate interplay between autophagy dysfunction, oxidative stress, and angiogenesis. Here, we introduce an intravitreal hydrogel depot, named Rab&Rapa-M@G, consisting of rapamycin-loaded microemulsion (Rapa-M, an mTOR inhibitor), ranibizumab (anti-VEGF antibody), and a thermosensitive hydrogel matrix. A single intravitreal injection of Rab&Rapa-M@G can sustainably deliver Rapa-M and ranibizumab to the retinal pigment epithelium for at least 14 days. This formulation significantly improves retinal autophagic flux homeostasis and reduces oxidative stress injury in wAMD mice by modulating the AMPK/mTOR/HIF-1α/VEGF and AMPK/ROS/HO-1/VEGF pathways. Consequently, it synergistically disrupts the "autophagic dysfunction-oxidative stress-angiogenesis" loop, leading to a remarkable reduction in choroidal neovascularization area and retinal damage compared to ranibizumab alone. Notably, the sequential administration of ranibizumab and Rab&Rapa-M@G further enhances the overall anti-wAMD efficacy, achieved through sequential delivery of Rab and Rapa, allowing for a more precise grasp of the treatment window. In conclusion, this hydrogel depot design, with its sequential and sustained delivery of mTOR inhibitors and anti-VEGF antibodies, offers a promising strategy for multi-target synergistic therapy in wAMD.
Collapse
Affiliation(s)
- Xi Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Congyan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Qun Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Yanli Lv
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Chen Lu
- The first affiliated hospital of Nanjing medical university, Nanjing 210000, China
| | - Wenting Su
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Jing Zhou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Huangqin Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Huiling Gong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Songtao Yuan
- The first affiliated hospital of Nanjing medical university, Nanjing 210000, China.
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
11
|
Wang T, Song Y, Bell BA, Anderson BD, Lee TT, Yu W, Dunaief JL. Complement C3 knockout protects photoreceptors in the sodium iodate model. Exp Eye Res 2025; 250:110161. [PMID: 39557279 PMCID: PMC11625604 DOI: 10.1016/j.exer.2024.110161] [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: 06/17/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Complement factor 3 (C3) has emerged as a primary therapeutic target in age-related macular degeneration (AMD) supported by genetic, histologic, and clinical trial evidence. Yet, the site(s) of action are unclear. The purpose of this study was to test the effect of C3 knockout on photoreceptors and retinal pigment epithelial cells (RPE) in the sodium iodate (NaIO3) model, which mirrors some features of AMD. C3-/- and WT mice, both on a C57Bl/6J background, were injected intraperitoneally with 25 mg/kg NaIO3. Electroretinography and optical coherence tomography were performed 7 days later to assess retinal function and structure, respectively. Then, mice were euthanized for retinal immunohistochemistry, quantitative real-time PCR and enzyme-linked immunosorbent assays. NaIO3 increased C3 protein levels in the neural retina but not RPE. WT but not C3-/- mice showed NaIO3-induced iC3b deposition on photoreceptor outer segments. C3-/- mice were partially protected against photoreceptor layer thinning. There was partial preservation of rod and cone function in the C3-/- group. Neither RPE structure nor function was protected. These results suggest outer segment opsonization contributes to photoreceptor death in this model, and that targeting C3 can protect photoreceptor structure and function when RPE cells are stressed.
Collapse
Affiliation(s)
- Tan Wang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ying Song
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Brent A Bell
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Brandon D Anderson
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Timothy T Lee
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Weihong Yu
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Joshua L Dunaief
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
12
|
Xue Y, Ye L, Huang C, Ye H. Lights and Shadows of Cytokines in Age-Related Eye Diseases: A Narrative Literature Review. J Biochem Mol Toxicol 2025; 39:e70121. [PMID: 39756061 DOI: 10.1002/jbt.70121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
The eye is considered to be an immune-privileged region. However, several parts of the eye have distinct mechanisms for delivering immune cells to the injury sites or even in response to aging. Although these immune responses are intended to be protective, the visual acuity can be compromised by the release of pro-inflammatory cytokines by immune cells, which induce chronic inflammation and fibrosis. Age-related eye diseases (AREDs) are the primary cause of vision impairment (VI) in the elderly, with a poor comprehension of their pathophysiology. Age-related eye diseases affect both the anterior and posterior segments, resulting in diminished quality of life and risk of irreversible blindness. Immune system dysregulation and the upregulation of pro-inflammatory cytokines have been linked to AREDs, underscoring the need to comprehend inflammation's impact on ocular disorders to enhance patient symptom management. In this framework, the PubMed database was searched using the medical subject headings (MeSH) terms "Age-related eye diseases," "dry eye syndrome," "glaucoma," "cataract," "diabetic retinopathy," "inflammation," "interleukin," and "cytokine" with the aim of overview the role of cytokines in AREDs and discuss their potential therapeutic approaches.
Collapse
Affiliation(s)
- Yuyu Xue
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan, China
| | - Lu Ye
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Chan Huang
- School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan, China
| | - Hejiang Ye
- Department of Ophthalmology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Wei S, Li J, Zhang Y, Li Y, Wang Y. Ferroptosis in eye diseases: a systematic review. Eye (Lond) 2025; 39:18-27. [PMID: 39379520 PMCID: PMC11733247 DOI: 10.1038/s41433-024-03371-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/09/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Ferroptosis is a type of iron-dependent cell death that differs from apoptosis, necroptosis, autophagy, and other forms of cell death. It is mainly characterized by the accumulation of intracellular lipid peroxides, redox imbalance, and reduced levels of glutathione and glutathione peroxidase 4. Studies have demonstrated that ferroptosis plays an important regulatory role in the occurrence and development of neurodegenerative diseases, stroke, traumatic brain injury, and ischemia-reperfusion injuries. Multiple mechanisms, such as iron metabolism, ferritinophagy, p53, and p62/Keap1/Nrf2, as well as the combination of FSP1/CoQ/NADPH and hepcidin/FPN-1 can alter the vulnerability to ferroptosis. Nevertheless, there has been limited research on the development and management of ferroptosis in the realm of eye disorders, with most studies focusing on retinal conditions such as age-related macular degeneration and retinitis pigmentosa. This review offers a thorough examination of the disruption of iron homeostasis in eye disorders, investigating the underlying mechanisms. We anticipate that the occurrence of ferroptotic cell death will not only establish a fresh field of study in eye diseases, but also present a promising therapeutic target for treating these diseases.
Collapse
Affiliation(s)
- Shengsheng Wei
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Jing Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yaohua Zhang
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yong Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yan Wang
- Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China.
- Nankai University Eye Institute, Nankai University, Tianjin, China.
| |
Collapse
|
14
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 DOI: 10.1016/j.preteyeres.2024.101306] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
15
|
Chen J, Lu T, Chen C, Zheng W, Lu L, Li N. Elevation of ANXA1 associated with potential protective mechanism against ferroptosis and immune cell infiltration in age-related macular degeneration. Eur J Med Res 2024; 29:615. [PMID: 39710756 DOI: 10.1186/s40001-024-02163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD), is a neurodegenerative ocular disease. This study investigated the role of ferroptosis-related genes and their interaction with immune cell infiltration in AMD. METHODS We screened differential expression genes (DEGs) of AMD from data sets in Gene Expression Omnibus. We identified ferroptosis-related differentially expressed genes (ferroDEGs) by intersecting DEGs with ferroptosis-related genes. Protein-protein interactions network and Cytoscape were used for screening hub genes. Next, we analyzed immune cell infiltration using CIBERSORT and examined the crosstalk between hub ferroDEGs and immune cell infiltration. Hub genes expression in each cell cluster and the proportions of different cell clusters between AMD and normal samples were examined using single-cell data. The hub ferroDEG expressions were verified in cell and mouse models using RT-qPCR, western blot, and immunofluorescence assay. The roles of ANXA1 in ferroptosis and its crosstalk with microglia were investigated. RESULTS We identified hub ferroDEGs that include six genes (ANXA1, DKK1, CD44, VIM, TGFB2, DUSP1). Functional analysis of those hub ferroDEGs was found to be correlated with leukocyte migration and chemotaxis, macrophage migration, and gliogenesis. The high-risk ferroptosis group exhibited elevated levels of CD8+ T cells, activated NK cells, and M2 macrophages. Single-cell sequencing data revealed a high degree of cell heterogeneity in macular degeneration and the monocytes proportion in the macular area was higher in AMD samples. Moreover, we observed elevated mRNA and protein levels of CD44, ANXA1 (P < 0.01), while ANXA1 knockdown reduced GPX4 expression in the cell model. Finally, we validated increased ANXA1 expression and observed its colocalization with microglia in mouse models using immunofluorescence assays. CONCLUSIONS This study offers insights into the AMD pathogenesis and identifies ANXA1 as a potential target related to protecting from ferroptosis and immune response for future research.
Collapse
Affiliation(s)
- Jing Chen
- Department of Ophthalmology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Tu Lu
- 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, 510060, China
- Department of Ophthalmology, First Hospital of China Medical University, Shenyang, China
| | - Chen 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, 510060, China
| | - Wenbin Zheng
- Department of Ophthalmology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Lin Lu
- 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, 510060, China.
| | - Naiyang Li
- Department of Ophthalmology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China.
| |
Collapse
|
16
|
Zhou Y, Xue F. Revolutionary drug repositioning: the preventive and therapeutic potential of metformin and other antidiabetic drugs in age-related macular degeneration. Front Pharmacol 2024; 15:1507860. [PMID: 39720591 PMCID: PMC11666363 DOI: 10.3389/fphar.2024.1507860] [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: 10/08/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness among the elderly worldwide. Anti-vascular endothelial growth factor (anti-VEGF) injections remain the first-line therapy for AMD. However, their high cost and the need for frequent administration pose challenges to long-term adherence, highlighting the need for accessible and cost-effective preventive strategies. Emerging evidence suggests that traditional antidiabetic drugs, such as metformin, sulfonylureas, and thiazolidinediones, may offer neuroprotective benefits, opening new avenues for AMD prevention. Among these, metformin has emerged as the most promising candidate, demonstrating significant potential in reducing AMD risk, even at low cumulative doses, primarily through AMP-activated protein kinase (AMPK) activation. Sulfonylureas, although effective in stimulating insulin secretion, carry risks such as hypoglycemia, hyperinsulinemia, and a possible association with increased cancer risk. Similarly, thiazolidinediones, while improving insulin sensitivity, are associated with adverse effects, including cardiovascular risks and macular edema, limiting their broader application in AMD prevention. This paper explores the preventive potential and underlying mechanisms of these antidiabetic drugs in AMD and discusses the role of artificial intelligence in optimizing individualized prevention strategies. By advancing precision medicine, these approaches may improve public health outcomes and reduce the burden of aging-related vision loss.
Collapse
|
17
|
Li M, Xu Q, Fan Q, Li H, Zhang Y, Jiang F, Qu Y. Small molecule SIRT1 activators counteract oxidative stress-induced inflammasome activation and nucleolar stress in retinal degeneration. Int Immunopharmacol 2024; 142:113167. [PMID: 39303543 DOI: 10.1016/j.intimp.2024.113167] [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: 06/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The nicotinamide adenosine dinucleotide-dependent deacetylase Sirtuin 1 (SIRT1) has been identified as a protective factor that inhibits the activation of nucleotide-binding and oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome. However, whether pharmacological SIRT1 activators can protect retinal pigment epithelial (RPE) cells against oxidative and inflammatory injuries related to age-related macular degeneration remains to be explored. METHODS Two small molecule specific SIRT1 activators (SRT2104 and CAY10602) were tested, with resveratrol being used as a positive control. Mouse models with sodium iodate-induced retinal degeneration were constructed. ARPE-19 cells in culture were used for in vitro experiments. The effects of SIRT1 activators on H2O2-induced ARPE-19 cell injury were determined by reactive oxygen species quantification, western blotting, flow cytometry and immunofluorescence staining. In vivo, the severity of retinal damage was assessed using flash electroretinography and histopathological analysis. RESULTS In vitro, SRT2104, CAY10602 and resveratrol significantly attenuated H2O2-induced cell death, nucleolar stress response, and reactive oxygen species accumulation. In H2O2-stimulated cells, SIRT1 activators reduced the level of NLRP3, inhibited the activation of caspase-1, and decreased the production of interleukin (IL)-1β and IL-18. The inhibitory effects of SIRT1 activators on caspase-1 activation and IL-1β production were blunted by SIRT1 gene silencing. In vivo, treatment with SRT2104 or CAY10602 in mice with sodium iodate-induced retinal degeneration markedly improved the retinal functions and reduced the loss of RPE cells. CONCLUSION Our study suggests that small molecule SIRT1 activators are effective for protection of RPE cells against oxidative stress-induced NLRP3 inflammasome activation, highlighting potential applications in the treatment of macular degeneration associated RPE dysfunctions.
Collapse
Affiliation(s)
- Mengyao Li
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine (202132001), Jinan, Shandong Province, China
| | - Qian Xu
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine (202132001), Jinan, Shandong Province, China
| | - Qian Fan
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine (202132001), Jinan, Shandong Province, China
| | - Haiming Li
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine (202132001), Jinan, Shandong Province, China
| | - Yu Zhang
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine (202132001), Jinan, Shandong Province, China
| | - Fan Jiang
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine (202132001), Jinan, Shandong Province, China.
| | - Yi Qu
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine (202132001), Jinan, Shandong Province, China.
| |
Collapse
|
18
|
Zhu M, Yu J. Salidroside alleviates ferroptosis in FAC-induced Age-related macular degeneration models by activating Nrf2/SLC7A11/GPX4 axis. Int Immunopharmacol 2024; 142:113041. [PMID: 39260309 DOI: 10.1016/j.intimp.2024.113041] [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: 06/29/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is a significant contributor to irreversible impairment in visual capability, particularly in its non-neovascular (dry) form. Ferroptosis, an emerging form of programmed necrosis, involves generating lipid peroxidation (LOS) through free iron and reactive oxygen species (ROS). Salidroside, a glycoside from Rhodiola rosea, known for anti-inflammatory and antioxidant properties. The research aim was exploring whether ferroptosis exists in dry AMD pathogenesis and elucidate salidroside's protective mechanisms against ferroptosis in AMD murine models and ARPE-19 cells. METHODS ARPE-19 cells were treated with varying concentrations of ferrous ammonium citrate (FAC) and salidroside. In an in vivo model, C57BL/6 mice were administered intraperitoneal injections of salidroside for 7 consecutive days, followed by an intravitreal injection (IVT) of FAC. After 7 days, the eyeballs were harvested for subsequent analyses. Ferroptosis markers were assessed using western blotting, immunofluorescence staining, and flow cytometry. To further elucidate the modulatory role of Nrf2 in ferroptosis, ARPE-19 cells were transfected with si-Nrf2. RESULTS In vitro, FAC-treated ARPE-19 cells exhibited reduced viability, decreased mitochondrial membrane potential (MMP), and accumulation of iron and lipid peroxidation (LOS) products. In vivo, FAC administration by IVT led to outer nuclear layer thinning and compromised tight junctions in RPE cells. The GPX4, Nrf2, and SLC7A11 expressions were downregulated both in vitro and in vivo. Salidroside upregulated Nrf2 and ameliorated these outcomes, but its effects were attenuated in ARPE-19 cells transfected with si-Nrf2. CONCLUSION Our study establishes that FAC induces RPE cell ferroptosis within dry AMD, and salidroside exerts therapeutic effects by triggering Nrf2/SLC7A11/GPX4 signaling axis.
Collapse
Affiliation(s)
- Meijiang Zhu
- Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China.
| | - Jing Yu
- Tongji University School of Medicine, Shanghai Tenth People's Hospital, Shanghai, China.
| |
Collapse
|
19
|
Mulfaul K, Khan AH, Schwarte SG, Voigt AP, Moore RF, Potempa LA, Wang K, Scheetz TE, Stone EM, Tucker BA, Mullins RF. Elevation of Granulocyte Colony Stimulating Factor in Human AMD Donor RPE-Choroid. Invest Ophthalmol Vis Sci 2024; 65:15. [PMID: 39641748 PMCID: PMC11629913 DOI: 10.1167/iovs.65.14.15] [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: 05/13/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Purpose Choroidal inflammation, complement deposition, and accumulation of C-reactive protein (CRP) are involved in age-related macular degeneration (AMD) pathology. The pro-inflammatory signals that regulate immune cell recruitment in the choroid of patients with AMD remain to be determined. We performed cytokine profiling of human AMD and age-matched control donor tissue to identify inflammatory molecules upregulated in AMD tissue. Methods Protein was isolated from 25 AMD and 21 control donor RPE/choroid macular punches. Total protein was quantified, and 50 µg assayed for expression of 40 cytokines using an inflammation array. We validated the elevated expression of granulocyte colony stimulating factor (G-CSF) protein by ELISA in a second cohort of 22 control and 26 AMD donors. To identify an AMD associated stressor responsible for upregulating G-CSF we assayed for changes in G-CSF protein secretion in RPE/choroid organ cultures treated with the monomeric (m)CRP, an inflammatory protein elevated in AMD. Results Using a multiplex array, we identified elevated G-CSF protein in the choroid of AMD donors compared to age-matched non-AMD controls. Differential expression of G-CSF was confirmed via ELISA in an independent cohort of samples (P = 0.01). The mCRP, which is deposited in AMD choroids, increased G-CSF protein secretion in RPE/choroid organ cultures. Single nuclei RNA sequencing identified choroidal endothelial cells and fibroblasts as the primary cell types responsible for increased G-CSF secretion in response to mCRP. The G-CSF receptor is expressed primarily by choroidal macrophages and dendritic cells and anti-G-CSFR colocalizes with anti-CD45 and anti-CD68 in human donor choroid tissue. Conclusions Elevated G-CSF expression in AMD donor tissue as a result of increased levels of mCRP may be involved in immune cell recruitment in AMD contributing to inflammatory stress in the choroid.
Collapse
Affiliation(s)
- Kelly Mulfaul
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Adnan H. Khan
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Samantha G. Schwarte
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Andrew P. Voigt
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Rachel F. Moore
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Lawrence A. Potempa
- Roosevelt University College of Pharmacy, Schaumburg, Illinois, United States
| | - Kai Wang
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, United States
| | - Todd E. Scheetz
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Edwin M. Stone
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Budd A. Tucker
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, United States
| | - Robert F. Mullins
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, United States
- Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
20
|
Yeh WJ, Chien PT, Wen YT, Wu CH. A comprehensive review of experimental models for investigating blue light-induced ocular damage: Insights into parameters, limitations, and new opportunities. Exp Eye Res 2024; 249:110142. [PMID: 39490726 DOI: 10.1016/j.exer.2024.110142] [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: 07/27/2024] [Revised: 09/26/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Light-emitting diodes (LEDs) are widely used in modern lighting and electronic devices, including smartphones, computer monitors, tablets, televisions, and vehicle lights. Blue light (BL) hazards to eye health have received increasing attention because white LED bulbs emit higher levels of BL than traditional lighting sources. At wavelengths of 400-500 nm, BL is characterized by its high energy and risks associated with prolonged exposure, which may lead to photochemical damage and morphological alterations in the retina. Recent research has revealed that the harmful effects of BL are intricately linked to light intensity and exposure frequency, with mechanisms involving the overproduction of reactive oxygen species through photooxidative processes. This growing body of knowledge deepens our understanding of photodamage and opens avenues for exploring protective strategies for our eyes. Although current clinical trials assessing the safety of BL exposure remain limited, the development of experimental models that mimic physiological conditions has revealed BL toxicity. This review categorizes and evaluates BL-induced retinopathy in vivo, providing a comprehensive overview of the associated experimental parameters, including photosensitive fluorophores, light wavelength, illuminance, irradiance, exposure duration, animal strains, and their unique lesion patterns. Moreover, this study underscores the need for further research to evaluate photoprotective agents, which may offer valuable insights to the ongoing discussion on preserving ocular health in our increasingly illuminated digital environments.
Collapse
Affiliation(s)
- Wan-Ju Yeh
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan
| | - Pin-Ting Chien
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97403, Taiwan
| | - Chi-Hao Wu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan.
| |
Collapse
|
21
|
Hultgren NW, Petcherski A, Torriano S, Komirisetty R, Sharma M, Zhou T, Burgess BL, Ngo J, Osto C, Shabane B, Shirihai OS, Kelesidis T, Williams DS. Productive infection of the retinal pigment epithelium by SARS-CoV-2: Initial effects and consideration of long-term consequences. PNAS NEXUS 2024; 3:pgae500. [PMID: 39712068 PMCID: PMC11660945 DOI: 10.1093/pnasnexus/pgae500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/14/2024] [Indexed: 12/24/2024]
Abstract
As the SARS-CoV-2 coronavirus continues to evolve and infect the global population, many individuals are likely to suffer from post-acute sequelae of SARS-CoV-2 infection (PASC). Manifestations of PASC include vision symptoms, but little is known about the ability of SARS-CoV-2 to infect and impact the retinal cells. Here, we demonstrate that SARS-CoV-2 can infect and perturb the retinal pigment epithelium (RPE) in vivo, after intranasal inoculation of a transgenic mouse model of SARS-CoV-2 infection, and in cell culture. Separate lentiviral studies showed that SARS-CoV-2 Spike protein mediates viral entry and replication in RPE cells, while the Envelope and ORF3a proteins induce morphological changes. Infection with major variants of SARS-CoV-2 compromised the RPE barrier function and phagocytic capacity. It also caused complement activation and production of cytokines and chemokines, resulting in an inflammatory response that spread across the RPE layer. This inflammatory signature has similarities to that associated with the onset of age-related macular degeneration (AMD), a major cause of human blindness, resulting from RPE pathology that eventually leads to photoreceptor cell loss. Thus, our findings suggest that post-acute sequelae of SARS-CoV-2 infection of the RPE may have long-term implications for vision, perhaps comparable to the increased occurrence of AMD found among individuals infected by HIV, but with greater public health consequences due to the much larger number of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Nan W Hultgren
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Anton Petcherski
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Simona Torriano
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Ravikiran Komirisetty
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Madhav Sharma
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tianli Zhou
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Barry L Burgess
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Jennifer Ngo
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Corey Osto
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Byourak Shabane
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Orian S Shirihai
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Theodoros Kelesidis
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David S Williams
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
- Department of Neurobiology, David Geffen School of Medicine; Molecular Biology Institute; Brain Research Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
22
|
Bakker LM, Boulton ME, Różanowska MB. (Photo)toxicity of Partially Oxidized Docosahexaenoate and Its Effect on the Formation of Lipofuscin in Cultured Human Retinal Pigment Epithelial Cells. Antioxidants (Basel) 2024; 13:1428. [PMID: 39594569 PMCID: PMC11591205 DOI: 10.3390/antiox13111428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Docosahexaenoate is a cytoprotective ω-3 polyunsaturated lipid that is abundant in the retina and is essential for its function. Due to its six unsaturated double bonds, docosahexaenoate is highly susceptible to oxidation and the formation of products with photosensitizing properties. This study aimed to test on cultured human retinal pigment epithelial cells ARPE-19 the (photo)cytotoxic potential of partly oxidized docosahexaenoate and its effect on the formation of lipofuscin from phagocytosed photoreceptor outer segments (POSs). The results demonstrate that the cytoprotective effects of docosahexaenoate do not counteract the deleterious effects of its oxidation products, leading to the concentration-dependent loss of cell metabolic activity, which is exacerbated by concomitant exposure to visible light. Partly oxidized docosahexaenoate does not cause permeability of the cell plasma membrane but does cause apoptosis. While vitamin E can provide partial protection from the (photo)toxicity of partly oxidized docosahexaenoate, zeaxanthin undergoes rapid photodegradation and can exacerbate the (photo)toxicity. Feeding cells with POSs enriched in partly oxidized docosahexaenoate results in a greater accumulation of intracellular fluorescent lipofuscin than in cells fed POSs without the addition. In conclusion, partly oxidized docosahexaenoate increases the accumulation of lipofuscin-like intracellular deposits, is cytotoxic, and its toxicity increases during exposure to light. These effects may contribute to the increased progression of geographic atrophy observed after long-term supplementation with docosahexaenoate in age-related macular degeneration patients.
Collapse
Affiliation(s)
- Linda M. Bakker
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK;
| | - Michael E. Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Małgorzata B. Różanowska
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK;
- Cardiff Institute for Tissue Engineering and Repair (CITER), Cardiff University, Cardiff CF10 3AX, Wales, UK
| |
Collapse
|
23
|
Geathers JS, Grillo SL, Karakoleva E, Campbell GP, Du Y, Chen H, Barber AJ, Zhao Y, Sundstrom JM. Sodium Iodate: Rapid and Clinically Relevant Model of AMD. FRONT BIOSCI-LANDMRK 2024; 29:380. [PMID: 39614439 DOI: 10.31083/j.fbl2911380] [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: 06/01/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Age-related macular degeneration (AMD) is the most common cause of vision loss in people above the age of 50, affecting approximately 10% of the population worldwide and the incidence is rising. Hyperreflective foci (HRF) are a major predictor of AMD progression. The purpose of this study was to use the sodium iodate mouse model to study HRF formation in retinal degeneration. METHODS Sodium iodate (NaIO3) treated rodents were studied to characterize HRF. 3-month-old male wild-type (WT) C57Bl/6J mice were injected with phosphate-buffered saline (PBS) or varying doses of NaIO3 (15-60 mg/kg). Optical Coherence Tomography (OCT) images were collected at baseline and several days post-NaIO3 injection. Retinal thicknesses were measured using Bioptigen software. Seven days post-injection, eyes were prepared for either transmission electron microscopy (TEM), Hematoxylin & Eosin (H&E), or immunofluorescence. RESULTS OCT imaging of the mice given higher doses of NaIO3 revealed HRF formation in the neural retina (n = 4). The amount of HRF correlated with the degree of retinal tissue loss. H&E and TEM imaging of the retinas seven days post-NaIO3 injection revealed several pigmented bodies in multiple layers of the retina (n = 3-5). Immunofluorescence revealed that some pigmented bodies were positive for macrophage markers and an epithelial-to-mesenchymal transition marker, while all were retinal pigment epithelium (RPE) 65-negative (n = 4). CONCLUSIONS The data suggest that NaIO3 induces the formation of HRF in the outer retina and their abundance correlates with retinal tissue loss. The experiments in this study highlight NaIO3 as a clinically relevant model of intermediate AMD that can be used to study HRF formation and to discover new treatment targets.
Collapse
Affiliation(s)
- Jasmine S Geathers
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Stephanie L Grillo
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Ema Karakoleva
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Gregory P Campbell
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Yixuan Du
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Han Chen
- Section of Research Resources, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Alistair J Barber
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Yuanjun Zhao
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jeffrey M Sundstrom
- Department of Ophthalmology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
24
|
Hernandez BJ, Strain M, Suarez MF, Stamer WD, Ashley-Koch A, Liu Y, Klingeborn M, Bowes Rickman C. Small Extracellular Vesicle-Associated MiRNAs in Polarized Retinal Pigmented Epithelium. Invest Ophthalmol Vis Sci 2024; 65:57. [PMID: 39589346 PMCID: PMC11601136 DOI: 10.1167/iovs.65.13.57] [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: 05/21/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024] Open
Abstract
Purpose Oxidative stress in the retinal pigmented epithelium (RPE) has been implicated in age-related macular degeneration by impacting endocytic trafficking, including the formation, content, and secretion of extracellular vesicles (EVs). Using our model of polarized primary porcine RPE (pRPE) cells under chronic subtoxic oxidative stress, we tested the hypothesis that RPE miRNAs packaged into EVs are secreted in a polarized manner and contribute to maintaining RPE homeostasis. Methods Small EVs (sEVs) enriched for exosomes were isolated from apical and basal conditioned media from pRPE cells grown for up to four weeks with or without low concentrations of hydrogen peroxide using two sEV isolation methods, leading to eight experimental groups. The sEV miRNA expression was profiled using miRNA-Seq with Illumina MiSeq, followed by quality control and bioinformatics analysis for differential expression using the R computing environment. Expression of selected miRNAs were validated using qRT-PCR. Results We identified miRNA content differences carried by sEVs isolated using two ultracentrifugation-based methods. Regardless of the sEV isolation method, miR-182 and miR-183 were enriched in the cargo of apically secreted sEVs, and miR-122 in the cargo of basally secreted sEVs from RPE cells during normal homeostatic conditions. After oxidative stress, miR-183 levels were significantly decreased in the cargo of apically released sEVs from stressed RPE cells. Conclusions We curated RPE sEV miRNA datasets based on cell polarity and oxidative stress. Unbiased miRNA analysis identified differences based on polarity, stress, and sEV isolation methods. These findings suggest that miRNAs in sEVs may contribute to RPE homeostasis and function in a polarized manner.
Collapse
Affiliation(s)
- Belinda J. Hernandez
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Madison Strain
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, North Carolina, United States
| | - Maria Fernanda Suarez
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Allison Ashley-Koch
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, North Carolina, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Mikael Klingeborn
- McLaughlin Research Institute, Great Falls, Montana, United States
- Touro College of Osteopathic Medicine Montana, Great Falls, Montana, United States
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Cell Biology, Duke University, Durham, North Carolina, United States
| |
Collapse
|
25
|
Klochkov V, Chan CM, Lin WW. Methylglyoxal: A Key Factor for Diabetic Retinopathy and Its Effects on Retinal Damage. Biomedicines 2024; 12:2512. [PMID: 39595078 PMCID: PMC11592103 DOI: 10.3390/biomedicines12112512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Diabetic retinopathy is the most common retinal vascular disease, affecting the retina's blood vessels and causing chronic inflammation, oxidative stress, and, ultimately, vision loss. Diabetes-induced elevated glucose levels increase glycolysis, the main methylglyoxal (MGO) formation pathway. MGO is a highly reactive dicarbonyl and the most rapid glycation compound to form endogenous advanced glycation end products (AGEs). MGO can act both intra- and extracellularly by glycating molecules and activating the receptor for AGEs (RAGE) pathway. Conclusions: This review summarizes the sources of MGO formation and its actions on various cell pathways in retinal cells such as oxidative stress, glycation, autophagy, ER stress, and mitochondrial dysfunction. Finally, the detoxification of MGO by glyoxalases is discussed.
Collapse
Affiliation(s)
- Vladlen Klochkov
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City 23148, Taiwan
| | - Chi-Ming Chan
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City 23148, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Wan-Wan Lin
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| |
Collapse
|
26
|
Boulakh L, Isaksen JL, Poulsen HE, Faber J, Heegaard S, Nygaard B, Kanters JK, Toft PB, Mortens Udholm P, Bek T, Buch Hesgaard H, Ellervik C. Thyroid dysfunction and exudative age-related macular degeneration - A longitudinal nationwide registry-based cohort study. Acta Ophthalmol 2024; 102:813-820. [PMID: 38712900 DOI: 10.1111/aos.16705] [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: 12/19/2023] [Accepted: 04/22/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE The association between thyroid dysfunction and exudative age-related macular degeneration (AMD) is unknown. METHODS In this Danish longitudinal nationwide registry-based cohort study we included all Danish residents aged 50-100 between 2008 and 2018. Using the Danish national registries, we studied the association between thyroid dysfunction and exudative AMD. Thyroid dysfunction was classified as two consecutive redeemed prescriptions of thyroid hormones (hypothyroidism) or anti-thyroid medication (hyperthyroidism). Exudative AMD was classified as an ICD diagnosis of AMD and a code for anti-VEGF treatment. All patients are treated for exudative AMD in a hospital in Denmark, and we therefore have complete registration of this patient group. RESULTS We included 2 087 305 individuals, of which 1 072 567 (51.4%) were women; 59 318 (2.8%) had hypothyroidism, and 33 922 (1.6%) had hyperthyroidism. During a median follow-up of 11 years, 26 998 (1.3%) people developed exudative AMD. Hypothyroidism (adjusted hazard ratio [HR]: 1.17; 95% confidence interval [CI] 1.10-1.25; p < 0.001) and hyperthyroidism (HR: 1.23; 95% CI:1.13-1.34; p < 0.001) were both associated with the development of exudative AMD. The age-stratified analyses yielded similar results to the main analyses, except that the risks were exaggerated in the older part of the population. CONCLUSION This is the first longitudinal nationwide study showing that both hypo- and hyperthyroidism are associated with an increased risk of exudative AMD. AMD is a quantitative problem in the population and our findings could have a public health impact. Further studies are needed to study the underlying mechanisms of the association.
Collapse
Affiliation(s)
- Lena Boulakh
- Department of Ophthalmology, Rigshospitalet - Glostrup, Glostrup, Denmark
| | - Jonas L Isaksen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Enghusen Poulsen
- Department of Endocrinology, Bispebjerg Fredriksberg Hospital, Frederiksberg, Denmark
- Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Faber
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Endocrinology, Herlev-Gentofte Hospital, Hellerup, Denmark
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet - Glostrup, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet-Glostrup, Glostrup, Denmark
| | - Birte Nygaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Endocrinology, Herlev-Gentofte Hospital, Hellerup, Denmark
| | - Jørgen Kim Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre of Physiological Research, University of California, San Francisco, San Francisco, California, USA
| | - Peter Bjerre Toft
- Department of Ophthalmology, Rigshospitalet - Glostrup, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | - Helena Buch Hesgaard
- Institute of Neuroscience and Physiology, Gothenburg University, Göteborg, Sweden
- Department of Ophthalmology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Data Support, Soroe, Region Zealand, Denmark
- Department of Laboratory Medicine, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Hyttinen JMT, Koskela A, Blasiak J, Kaarniranta K. Autophagy in drusen biogenesis secondary to age-related macular degeneration. Acta Ophthalmol 2024; 102:759-772. [PMID: 39087629 DOI: 10.1111/aos.16744] [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: 03/25/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Age-related macular degeneration (AMD) is an emerging cause of blindness in aged people worldwide. One of the key signs of AMD is the degeneration of the retinal pigment epithelium (RPE), which is indispensable for the maintenance of the adjacent photoreceptors. Because of impaired energy metabolism resulting from constant light exposure, hypoxia, and oxidative stress, accumulation of drusen in AMD-affected eyes is observed. Drusen contain damaged cellular proteins, lipoprotein particles, lipids and carbohydrates and they are related to impaired protein clearance, inflammation, and extracellular matrix modification. When autophagy, a major cellular proteostasis pathway, is impaired, the accumulations of intracellular lipofuscin and extracellular drusen are detected. As these aggregates grow over time, they finally cause the disorganisation and destruction of the RPE and photoreceptors leading to visual loss. In this review, the role of autophagy in drusen biogenesis is discussed since impairment in removing cellular waste in RPE cells plays a key role in AMD progression. In the future, means which improve intracellular clearance might be of use in AMD therapy to slow the progression of drusen formation.
Collapse
Affiliation(s)
- Juha M T Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ali Koskela
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, Plock, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
- Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
28
|
Carr BJ, Skitsko D, Kriese LM, Song J, Li Z, Ju MJ, Moritz OL. prominin-1-null Xenopus laevis develop subretinal drusenoid-like deposits, cone-rod dystrophy and RPE atrophy. J Cell Sci 2024; 137:jcs262298. [PMID: 39355864 PMCID: PMC11586525 DOI: 10.1242/jcs.262298] [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: 06/04/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
Prominin-1 (PROM1) variants are associated with inherited, non-syndromic vision loss. We used CRISPR/Cas9 to induce prom1-null mutations in Xenopus laevis and then tracked retinal disease progression from the ages of 6 weeks to 3 years. We found that prom1-null-associated retinal degeneration in frogs was age-dependent and involved retinal pigment epithelium (RPE) dysfunction preceding photoreceptor degeneration. Before photoreceptor degeneration occurred, aging prom1-null frogs developed larger and increasing numbers of cellular debris deposits in the subretinal space and outer segment layer, which resembled subretinal drusenoid deposits (SDDs) in their location, histology and representation as seen by color fundus photography and optical coherence tomography (OCT). Evidence for an RPE origin of these deposits included infiltration of pigment granules into the deposits, thinning of the RPE as measured by OCT, and RPE disorganization as measured by histology and OCT. The appearance and accumulation of SDD-like deposits and RPE thinning and disorganization in our animal model suggests an underlying disease mechanism for prom1-null-mediated blindness that involves death and dysfunction of the RPE preceding photoreceptor degeneration, instead of direct effects upon photoreceptor outer segment morphogenesis, as was previously hypothesized.
Collapse
Affiliation(s)
- Brittany J. Carr
- The University of Alberta, Faculty of Medicine and Dentistry, Department of Ophthalmology and Visual Sciences, Edmonton, AB T6G 2E1, Canada
- The University of Alberta, Faculty of Medicine and Dentistry, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Dominic Skitsko
- The University of British Columbia, Faculty of Medicine, Department of Ophthalmology and Visual Sciences, Vancouver, BC V5Z 0A6, Canada
| | - Linnea M. Kriese
- The University of Alberta, Faculty of Medicine and Dentistry, Department of Ophthalmology and Visual Sciences, Edmonton, AB T6G 2E1, Canada
- The University of Alberta, Faculty of Medicine and Dentistry, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Jun Song
- The University of British Columbia, Faculty of Applied Science, Faculty of Medicine, School of Biomedical Engineering, Vancouver, BC V6T 2B9, Canada
| | - Zixuan Li
- The University of Alberta, Faculty of Medicine and Dentistry, Department of Ophthalmology and Visual Sciences, Edmonton, AB T6G 2E1, Canada
- The University of Alberta, Faculty of Medicine and Dentistry, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Myeong Jin Ju
- The University of British Columbia, Faculty of Medicine, Department of Ophthalmology and Visual Sciences, Vancouver, BC V5Z 0A6, Canada
- The University of British Columbia, Faculty of Applied Science, Faculty of Medicine, School of Biomedical Engineering, Vancouver, BC V6T 2B9, Canada
| | - Orson L. Moritz
- The University of British Columbia, Faculty of Medicine, Department of Ophthalmology and Visual Sciences, Vancouver, BC V5Z 0A6, Canada
| |
Collapse
|
29
|
Zhang Y, Tang X, Liu L, Cai D, Gou S, Hao S, Li Y, Shen J, Chen Y, Zhao Y, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Zhang Z, Wang X, Deng S, Xiao Z, Yao L, Du F. GLO1 regulates hepatocellular carcinoma proliferation and migration through the cell cycle pathway. BMC Cancer 2024; 24:1297. [PMID: 39434012 PMCID: PMC11492659 DOI: 10.1186/s12885-024-12927-x] [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: 07/06/2023] [Accepted: 09/10/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant tumor characterized by a high mortality rate. The occurrence and progression of HCC are linked to oxidative stress. Glyoxalase-1 (GLO1) plays an important role in regulating oxidative stress, yet the underlying mechanism remains unclear. GLO1 may serve as a prognostic biomarker and therapeutic target for HCC. METHODS Based on TCGA database hepatocellular carcinoma samples, we conducted a bioinformatics analysis to explore the correlation between GLO1 expression and HCC cell proliferation and viability. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that differentially expressed genes (DEGs) were mainly enriched in the cell cycle pathway. We analyzed the relationships between GLO1 and 24 genes enriched in the cell cycle pathway using a protein-protein interaction (PPI) network. Finally, experimental validation was performed to assess GLO1's impact on the distribution of cells at different cell cycle stages and on the proliferation and migration of HCC cells. RESULTS Our study demonstrated that GLO1 was overexpressed in HCC tissues and was associated with a poor prognosis. Data analysis indicated that overexpression of GLO1 activated the cell cycle pathway and positively correlated with expression of the majority of key cell cycle genes. Experimental validation showed that GLO1 expression affects the number of HCC cells in G2 and S phases and regulates HCC cell proliferation and migration. CONCLUSIONS GLO1 represents a promising therapeutic target for HCC, providing valuable insights into its role in the viability and proliferation of HCC cells.
Collapse
Affiliation(s)
- Yao Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Xiaolong Tang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Lin Liu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Siyu Hao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Yan Li
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zhuo Zhang
- Key Laboratory of Luzhou City for Aging Medicine, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China.
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Lei Yao
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, 610072, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
30
|
Azuma K, Suzuki T, Kobayashi K, Nagahara M, Imai H, Suga A, Iwata T, Shiraya T, Aihara M, Ueta T. Retinal pigment epithelium-specific ablation of GPx4 in adult mice recapitulates key features of geographic atrophy in age-related macular degeneration. Cell Death Dis 2024; 15:763. [PMID: 39426958 PMCID: PMC11490617 DOI: 10.1038/s41419-024-07150-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss in the elderly population, particularly the late-stage of dry AMD known as geographic atrophy (GA), lacks effective treatment options. Genetic mouse models of AMD have revealed the significance of impaired lipid metabolism and anti-oxidative capacity in early/intermediate stage of AMD, but remains unclear in GA that severely damages visual function. Here, to investigate the potential relevance of peroxidized lipids in RPE for late-stage dry AMD, GPx4fl/fl mice underwent subretinal injections of RPE-specific AAV-Cre vector or control AAV vector. RPE-specific GPx4 deficiency led to rapid RPE degeneration resembling key features of late-stage dry AMD, including preceding loss of RPE cell polarity, accumulation of acrolein, malondialdehyde, and 4-hydroxynonenal, photoreceptor loss, lipofuscin-laden subretinal melanophage infiltration, and complement activation. Treatment with α-tocopherol and ferrostatin-1 mitigated RPE degeneration, and shrunk mitochondria were observed in GPx4 deficient mice, suggesting involvement of ferroptosis. Unexpectedly, necrostatin-1s, an inhibitor of necroptosis, also ameliorated RPE degeneration, and activation of RIP3 and MLKL along with inactivation of caspase-8 was observed, indicating crosstalk between ferroptosis and necroptosis pathways. Our findings shed light on the intricate mechanisms underlying RPE degeneration in AMD and highlight GPx4/lipid peroxidation as potential therapeutic targets. RPE-specific ablation of GPx4 in mice provides a valuable tool for further elucidating the interplay between lipid peroxidation, cell death pathways, and AMD pathogenesis, offering new insights for preclinical research and therapeutic development targeting GA.
Collapse
Affiliation(s)
- Kunihiro Azuma
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo Ward, Japan
- Department of Ophthalmology, National Center for Global Health and Medicine, Shinjuku Ward, Japan
| | - Takafumi Suzuki
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo Ward, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Masako Nagahara
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo Ward, Japan
| | - Hirotaka Imai
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Akiko Suga
- Molecular and Cellular Biology Division, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Takeshi Iwata
- Molecular and Cellular Biology Division, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Tomoyasu Shiraya
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo Ward, Japan
| | - Makoto Aihara
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo Ward, Japan
| | - Takashi Ueta
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo Ward, Japan.
| |
Collapse
|
31
|
Chen TE, Lo J, Huang SP, Chang KC, Liu PL, Wu HE, Chen YR, Chang YC, Liu CC, Lee PY, Lai YH, Wu PC, Wang SC, Li CY. Glaucine inhibits hypoxia-induced angiogenesis and attenuates LPS-induced inflammation in human retinal pigment epithelial ARPE-19 cells. Eur J Pharmacol 2024; 981:176883. [PMID: 39128809 DOI: 10.1016/j.ejphar.2024.176883] [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: 09/16/2023] [Revised: 07/24/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Glaucine is an aporphine alkaloid with anti-inflammatory, bronchodilator and anti-cancer activities. However, the effects of glaucine in the regulation of age-related macular degeneration (AMD) remain unclear. Herein, we aimed to investigate the anti-angiogenetic and anti-inflammatory effects of glaucine in ARPE-19 cells. ARPE-19 cells were treated with N-(methoxyoxoacetyl)-glycine, methyl ester (DMOG) and cobalt chloride (CoCl2) for induction of hypoxia, while lipopolysaccharide (LPS) treatment was used for elicitation of inflammatory response. Cell viability was analyzed using 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay. The expression of hypoxia-inducible factor (HIF-1α) and vascular endothelial growth factor (VEGF) were measured by Western blot. The secretion of VEGF, interleukin (IL)-6 and monocyte chemoattractant protein-1 (MCP-1) was detected using enzyme-linked immunosorbent assay (ELISA). Human umbilical vein endothelial cells (HUVECs) were used for tube formation analysis. Expression of HIF-1α and secretion of VEGF were significantly increased under DMOG and CoCl2 induction, whereas glaucine significantly attenuated both HIF-1α expression and VEGF secretion by DMOG- and CoCl2-induced ARPE-19 cells. In addition, glaucine suppressed the tube formation by DMOG- and CoCl2-induced HUVEC cells. Moreover, glaucine also attenuated the production of IL-6 and MCP-1 by LPS-induced ARPE-19 cells. This study indicated that glaucine exhibited anti-angiogenic and anti-inflammatory effects, suggesting that glaucine might have benefits for the treatment of AMD.
Collapse
Affiliation(s)
- Ting-En Chen
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Jung Lo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Shu-Pin Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Institute of Medical Science and Technology, College of Medicine, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pitts-burgh, PA, 15213, USA
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Hsin-En Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yuan-Ru Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yo-Chen Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ching-Chih Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Ophthalmology, Chi Mei Medical Center, Tainan 71004, Taiwan
| | - Po-Yen Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yu-Hung Lai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Pei-Chang Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan.
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan.
| |
Collapse
|
32
|
Li S, Qiu Y, Li Y, Wu J, Yin N, Ren J, Shao M, Yu J, Song Y, Sun X, Gao S, Cao W. Serum metabolite biomarkers for the early diagnosis and monitoring of age-related macular degeneration. J Adv Res 2024:S2090-1232(24)00434-X. [PMID: 39369956 DOI: 10.1016/j.jare.2024.10.001] [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: 05/22/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024] Open
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is a leading cause of irreversible blindness worldwide, with significant challenges for early diagnosis and treatment. OBJECTIVES To identify new biomarkers that are important for the early diagnosis and monitoring of the severity/progression of AMD. METHODS We investigated the diagnostic and monitoring potential of blood metabolites in a cohort of 547 individuals (167 healthy controls, 240 individuals with other eye diseases as eye disease controls, and 140 individuals with AMD) from 2 centers over three phases: discovery phase 1, discovery phase 2, and an external validation phase. The samples were analyzed via a mass spectrometry-based, widely targeted metabolomic workflow. In discovery phases 1 and 2, we built a machine learning algorithm to predict the probability of AMD. In the external validation phase, we further confirmed the performance of the biomarker panel identified by the algorithm. We subsequently evaluated the performance of the identified biomarker panel in monitoring the progression and severity of AMD. RESULTS We developed a clinically specific three-metabolite panel (hypoxanthine, 2-furoylglycine, and 1-hexadecyl-2-azelaoyl-sn-glycero-3-phosphocholine) via five machine learning models. The random forest model effectively discriminated patients with AMD from patents in the other two groups and showed acceptable calibration (area under the curve (AUC) = 1.0; accuracy = 1.0) in both discovery phases 1 and 2. An independent validation phase confirmed the diagnostic model's efficacy (AUC = 0.962; accuracy = 0.88). The three-biomarker panel model demonstrated an AUC of 1.0 in differentiating the severity of AMD via RF machine learning, which was consistent across both the discovery and external validation phases. Additionally, the biomarker concentrations remained stable under repeated freeze-thaw cycles (P > 0.05). CONCLUSIONS This study reveals distinct metabolite variations in the serum of AMD patients, paving the way for the development of the first routine laboratory test for AMD.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai 200031, China; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| | - Yichao Qiu
- Department of Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Yingzhu Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Jianing Wu
- Department of Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Ning Yin
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Jun Ren
- Department of Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Mingxi Shao
- Department of Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Jian Yu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai 200031, China; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Yunxiao Song
- Department of Clinical Laboratory, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200031, China
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai 200031, China; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Shunxiang Gao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China.
| | - Wenjun Cao
- Department of Clinical Laboratory, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai 200031, China; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| |
Collapse
|
33
|
Bresciani G, Manai F, Felszeghy S, Smedowski A, Kaarniranta K, Amadio M. VEGF and ELAVL1/HuR protein levels are increased in dry and wet AMD patients. A new tile in the pathophysiologic mechanisms underlying RPE degeneration? Pharmacol Res 2024; 208:107380. [PMID: 39216841 DOI: 10.1016/j.phrs.2024.107380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Age-related macular degeneration (AMD) is a common retinal pathology characterized by degeneration of macula's retinal pigment epithelium (RPE) and photoreceptors, visual impairment, or loss. Compared to wet AMD, dry AMD is more common, but lacks cures; therefore, identification of new potential therapeutic targets and treatments is urgent. Increased oxidative stress and declining antioxidant, detoxifying systems contribute to the pathophysiologic mechanisms underlying AMD. The present work shows that the Embryonic Lethal Abnormal Vision-Like 1/Human antigen R (ELAVL1/HuR) and the Vascular Endothelial Growth Factor (VEGF) protein levels are higher in the RPE of both dry and wet AMD patients compared to healthy subjects. Moreover, increased HuR protein levels are detected in the retina, and especially in the RPE layer, of a dry AMD model, the nuclear factor erythroid 2-related factor 2 (Nrf2) / peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) double knock-out mouse. The crosstalk among Nrf2, HuR and VEGF has been also studied in ARPE-19 cells in basal and stressful conditions related to the AMD context (i.e., oxidative stress, autophagy impairment, Nrf2 deficit), offering new evidence of the mutual influence between Nrf2 and HuR, of the dependence of VEGF expression and secretion by these two factors, and of the increased susceptibility of cells to stressful conditions in Nrf2- or HuR-impaired contexts. Overall, this study shows evidence of the interplay among Nrf2, HuR and VEGF, essential factors for RPE homeostasis, and represents an additional piece in the understanding of the complex pathophysiologic mechanisms underlying AMD.
Collapse
Affiliation(s)
| | - Federico Manai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Szabolcs Felszeghy
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland; Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Adrian Smedowski
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland; GlaucoTech Co., Katowice, Poland; Department of Ophthalmology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland; Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | | |
Collapse
|
34
|
Guo Y, Chen S, Guan W, Xu N, Zhu L, Du W, Liu Z, Fong HKW, Huang L, Zhao M. Retinal G-protein-coupled receptor deletion exacerbates AMD-like changes via the PINK1-parkin pathway under oxidative stress. FASEB J 2024; 38:e70135. [PMID: 39467145 PMCID: PMC11580724 DOI: 10.1096/fj.202401160rr] [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: 05/23/2024] [Revised: 09/18/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
The intake of high dietary fat has been correlated with the progression of age-related macular degeneration (AMD), affecting the function of the retinal pigment epithelium through oxidative stress. A high-fat diet (HFD) can lead to lipid metabolism disorders, excessive production of circulating free fatty acids, and systemic inflammation by aggravating the degree of oxidative stress. Deletion of the retinal G-protein-coupled receptor (RGR-d) has been identified in drusen. In this study, we investigated how the RGR-d exacerbates AMD-like changes under oxidative stress, both in vivo and in vitro. Fundus atrophy became evident, at 12 months old, particularly in the RGR-d + HFD group, and fluorescence angiography revealed narrower retinal vessels and a reduced perfusion area in the peripheral retina. Although rod electroretinography revealed decreasing trends in the a- and b-wave amplitudes in the RGR-d + HFD group at 12 months, the changes were not statistically significant. Mice in the RGR-d + HFD group showed a significantly thinner and more fragile retinal morphology than those in the WT + HFD group, with disordered and discontinuous pigment distribution in the RGR-d + HFD mice. Transmission electron microscopy revealed a thickened Bruch's membrane along the choriocapillaris endothelial cell wall in the RGR-d + HFD mice, and the outer nuclear layer structure appeared disorganized, with reduced nuclear density. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated significantly lower levels of 25(OH)-vitamin D3 metabolites in the RGR-d + HFD group. Under oxidative stress, RGR-d localized to the mitochondria and reduced the levels of the PINK1-parkin pathway. RGR-d mice fed an HFD were used as a new animal model of dry AMD. Under high-fat-induced oxidative stress, RGR-d accumulated in the mitochondria, disrupting normal mitophagy and causing cellular damage, thus exacerbating AMD-like changes both in vivo and in vitro.
Collapse
Affiliation(s)
- Yue Guo
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Sitong Chen
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Wenxue Guan
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Ningda Xu
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Li Zhu
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Wei Du
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Zhiming Liu
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Henry K. W. Fong
- Department of Ophthalmology, USC Roski Eye InstituteKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Lvzhen Huang
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Mingwei Zhao
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| |
Collapse
|
35
|
Picard E, Youale J, Hyman MJ, Xie E, Achiedo S, Kaufmann GT, Moir J, Daruich A, Crisanti P, Torriglia A, Polak M, Behar-Cohen F, Skondra D, Berdugo M. Glyburide confers neuroprotection against age-related macular degeneration (AMD). Transl Res 2024; 272:81-94. [PMID: 38815899 DOI: 10.1016/j.trsl.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 06/01/2024]
Abstract
Glyburide, a sulfonylurea drug used to treat type 2 diabetes, boasts neuroprotective effects by targeting the sulfonylurea receptor 1 (SUR1) and associated ion channels in various cell types, including those in the central nervous system and the retina. Previously, we demonstrated that glyburide therapy improved retinal function and structure in a rat model of diabetic retinopathy. In the present study, we explore the application of glyburide in non-neovascular ("dry") age-related macular degeneration (AMD), another progressive disease characterized by oxidative stress-induced damage and neuroinflammation that trigger cell death in the retina. We show that glyburide administration to a human cone cell line confers protection against oxidative stress, inflammasome activation, and apoptosis. To corroborate our in vitro results, we also conducted a case-control study, controlling for AMD risk factors and other diabetes medications. It showed that glyburide use in patients reduces the odds of new-onset dry AMD. A positive dose-response relationship is observed from this analysis, in which higher cumulative doses of glyburide further reduce the odds of new-onset dry AMD. In the quest for novel therapies for AMD, glyburide emerges as a promising repurposable drug given its known safety profile. The results from this study provide insights into the multifaceted actions of glyburide and its potential as a neuroprotective agent for retinal diseases; however, further preclinical and clinical studies are needed to validate its therapeutic potential in the context of degenerative retinal disorders such as AMD.
Collapse
Affiliation(s)
- Emilie Picard
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France
| | - Jenny Youale
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France
| | - Max J Hyman
- enter for Health and the Social Sciences, University of Chicago, Chicago, Illinois
| | - Edward Xie
- Stony Brook University Hospital, Stony Brook, NY
| | - Seiki Achiedo
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France
| | | | - John Moir
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois
| | - Alejandra Daruich
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France; AP-HP, Service d'Ophtalmologie, Hôpital universitaire Necker-Enfants Malades, Paris, France
| | - Patricia Crisanti
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France
| | - Alicia Torriglia
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France
| | - Michel Polak
- AP-HP, Service d'endocrinologie, diabétologie et gynécologie pédiatriques, Hôpital universitaire Necker-Enfants Malades, Paris, France; Inserm U1016, Institut Cochin, Paris, France; Inserm UMR 1163, Institut Imagine, Université Paris Cité, Paris, France
| | - Francine Behar-Cohen
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France; AP-HP, OphtalmoPôle, Hôpital Cochin, Department of Ophthalmology and Visual Science, Paris, France.
| | - Dimitra Skondra
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Marianne Berdugo
- Inserm UMRS1138, Team 1: Physiopathology of ocular diseases-Therapeutic innovations, Centre de Recherche des Cordeliers, Paris, France; Sorbonne Université UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Cité, Faculté de Santé, Paris, France
| |
Collapse
|
36
|
Sun Q, Ni Y, Wang K, Zhang H, Liu J, Xu L, Zhao Y. Rhodium nanozyme mitigates RPE degeneration and preserves vision in age-related macular degeneration via antioxidant and anti-inflammatory mechanisms. Mater Today Bio 2024; 28:101230. [PMID: 39296359 PMCID: PMC11408860 DOI: 10.1016/j.mtbio.2024.101230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/17/2024] [Accepted: 09/03/2024] [Indexed: 09/21/2024] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness among elderly people worldwide. However, there are currently no effective treatments for AMD. Oxidative stress-induced retinal pigment epithelium (RPE) degeneration and the inflammatory response are the main causes of AMD. In this study, a polyethylene glycol (PEG)-coated rhodium nanozyme (PEG-RhZ) with excellent reactive oxygen species (ROS) and reactive nitrogen species (RNS) elimination capability was synthesized for the treatment of AMD. PEG-RhZs protected RPE cell viability and barrier function upon exposure to oxidative stress stimuli. Additionally, microglial migration and iNOS, IL-1β and TNF-α expression were inhibited by PEG-RhZs. In the acute phase of the AMD model, PEG-RhZs significantly alleviated RPE oxidative damage and inhibited microglial activation. In the late stage of the AMD model, PEG-RhZs reduced photoreceptor loss and improved vision impairment. Furthermore, PEG-RhZs showed good biocompatibility and stability both in vitro and in vivo. Collectively, our findings suggest the therapeutic potential of PEG-RhZs for AMD treatment. STATEMENT OF SIGNIFICANCE: AMD is a kind of retinal degenerative disease that poses heavy health burden globally. PEG-RhZs exhibiting robust ROS and RNS scavenging capabilities have shown promise in safeguarding retinal pigment epithelium (RPE) from oxidative stress, suppressing microglia activation and the secretion of pro-inflammatory molecules, mitigating loss of retinal photoreceptor cells, and ameliorating visual impairment. The commendable antioxidant properties, biological safety, and biostability of PEG-RhZs offer valuable insights for the clinical management of AMD.
Collapse
Affiliation(s)
- Qian Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yueqi Ni
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kang Wang
- Hubei Province Tobacco Quality Supervision and Test Station, Wuhan, 430030, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lingjuan Xu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
37
|
Warner EF, Vaux L, Boyd K, Widdowson PS, Binley KM, Osborne A. Ocular delivery of Pigment Epithelium-Derived Factor (PEDF) as a neuroprotectant for Geographic Atrophy. Aging Dis 2024; 15:2003-2007. [PMID: 38421833 PMCID: PMC11346394 DOI: 10.14336/ad.2024.0216-1] [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: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
Geographic atrophy (GA) is an advanced form of age-related macular degeneration (AMD), that starts with atrophic lesions in the outer retina that expand to cover the macula and fovea, leading to severe vision loss over time. Pigment Epithelium-Derived Factor (PEDF) has a diverse-range of properties, including its ability to promote cell survival, reduce inflammation, inhibit angiogenesis, combat oxidative stress, regulate autophagy, and stimulate anti-apoptotic pathways, making it a promising therapeutic candidate for GA. However, the relatively short half-life of PEDF protein has precluded its potential as a clinical therapy for GA since it would require frequent injections. Therefore, we describe administration of a PEDF gene, comparing and contrasting delivery routes, viral and non-viral vectors, and consider the critical challenges for PEDF as a neuroprotectant for GA.
Collapse
Affiliation(s)
- Emily F. Warner
- Ikarovec Limited., The Norwich Research Park Innovation Centre, Norwich, NR4 7GJ, United Kingdom
| | | | | | | | | | - Andrew Osborne
- Ikarovec Limited., The Norwich Research Park Innovation Centre, Norwich, NR4 7GJ, United Kingdom
| |
Collapse
|
38
|
Wojciechowski AM, Bell BA, Song Y, Anderson BD, Conomikes A, Petruconis C, Dunaief JL. Inducible RPE-specific GPX4 knockout causes oxidative stress and retinal degeneration with features of age-related macular degeneration. Exp Eye Res 2024; 247:110028. [PMID: 39128667 PMCID: PMC11392608 DOI: 10.1016/j.exer.2024.110028] [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: 06/19/2024] [Revised: 07/11/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of vision loss in the elderly. This disease involves oxidative stress burden in the retina leading to death of retinal pigment epithelial (RPE) cells and photoreceptors. The retina is susceptible to oxidative stress, in part due to high metabolic activity and high concentration of polyunsaturated fatty acids that undergo lipid peroxidation chain reactions. Antioxidant enzymes exist in the retina to combat this stress, including glutathione peroxidase 4 (GPX4). GPX4 specifically reduces oxidized lipids, protecting against lipid peroxidation-induced oxidative stress, which is noted in dry AMD. We hypothesize that Gpx4 knockout within the RPE will result in an environment of chronic oxidative stress yielding degeneration akin to AMD. C57BL/6J mice with a floxed Gpx4 gene were mated with Rpe65Cre/ER mice. Offspring containing Rpe65Cre ± alleles and either Gpx4 WT or Gpx4 fl/fl alleles were administered tamoxifen to induce Gpx4 knockout in Gpx4 fl/fl mice. At sequential timepoints, retinal phenotypes were assessed via in vivo imaging utilizing confocal scanning laser ophthalmoscopy and optical coherence tomography (OCT), and visual function was probed by electroretinography. Retinas were studied post-mortem by immunohistochemical analyses, electron microscopy, plastic sectioning, and quantitative polymerase chain reaction and Western analyses. The RPE-specific Gpx4 knockout model was validated via Western analysis indicating diminished GPX4 protein only within the RPE and not the neural retina. Following Gpx4 knockout, RPE cells became dysfunctional and died, with significant cell loss occurring 2 weeks post-knockout. Progressive thinning of the photoreceptor layer followed RPE degeneration and was accompanied by loss of visual function. OCT and light microscopy showed hyperreflective foci and enlarged, pigmented cells in and above the RPE layer. Electron microscopy revealed decreased mitochondrial cristae and loss of basal and apical RPE ultrastructure. Finally, there was increased carboxyethylpyrrole staining, indicating oxidation of docosahexaenoic acid, and increased levels of mRNAs encoding oxidative stress-associated genes in the RPE and photoreceptors. Overall, we show that RPE-localized GPX4 is necessary for the health of the RPE and outer retina, and that knockout recapitulates phenotypes of dry AMD.
Collapse
Affiliation(s)
- Alaina M Wojciechowski
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Brent A Bell
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ying Song
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Brandon D Anderson
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Alexa Conomikes
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Cecilia Petruconis
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Joshua L Dunaief
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
39
|
Wu S, Zheng F, Sui A, Wu D, Chen Z. Sodium-iodate injection can replicate retinal and choroid degeneration in pigmented mice: Using multimodal imaging and label-free quantitative proteomics analysis. Exp Eye Res 2024; 247:110050. [PMID: 39151777 DOI: 10.1016/j.exer.2024.110050] [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: 05/22/2024] [Revised: 07/12/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible visual loss in the elderly population. Sodium iodate (NaIO3), a stable oxidizing agent, has been injected to establish a reproducible model of oxidative stress-induced RPE and photoreceptor death. The aim of our study was to evaluate the morphological and molecular changes of retina and retinal pigment epithelium (RPE)-choroid in NaIO3-treated mouse using multimodal fundus imaging and label-free quantitative proteomics analysis. Here, we found that following NaIO3 injection, retinal degeneration was evident. Fundus photographs showed numerous scattered yellow-white speckled deposits. Optical coherence tomography (OCT) images indicated disruption of the retinal layers, damage of the RPE layer and accumulation of hyper-reflective matter in multiple layers of the outer retina. Widespread foci of a high fundus autofluorescence (FAF) signal were noticed. Fundus fluorescein angiography (FFA) revealed diffuse intense transmitted fluorescence mixed with scattered spot-like blocked fluorescence. Indocyanine green angiography (ICGA) presented punctate hyperfluorescence. Due to the atrophy of the RPE and Bruch's membrane and choroidal capillary complex, the larger choroidal vessels become more prominent in ICGA and optical coherence tomography angiography (OCTA). Transmission electron microscope (TEM) illustrated abnormal material accumulation and damaged mitochondria. Bioinformatics analysis of proteomics revealed that the differentially expressed proteins participated in diverse biological processes, encompassing phototransduction, NOD-like receptor signaling pathway, phagosome, necroptosis, and cell adhesion molecules. In conclusion, by multimodal imaging, we described the phenotype of NaIO3-treated mouse model mimicking oxidative stress-induced RPE and photoreceptor death in detail. In addition, proteomics analysis identified differentially expressed proteins and significant enrichment pathways, providing insights for future research, although the exact mechanism of oxidative stress-induced RPE and photoreceptor death remains incompletely understood.
Collapse
Affiliation(s)
- Shijing Wu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Hangzhou, 310009, Zhejiang, China
| | - Fang Zheng
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Hangzhou, 310009, Zhejiang, China
| | - Ailing Sui
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Hangzhou, 310009, Zhejiang, China
| | - Di Wu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Hangzhou, 310009, Zhejiang, China.
| | - Zhiqing Chen
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
40
|
Bernd J, Plastino F, Karayannis JJ, Kvanta A, Locri F, André H. Accelerated maturation of ARPE-19 cells for the translational assessment of gene therapy. FASEB J 2024; 38:e70020. [PMID: 39222301 DOI: 10.1096/fj.202301707rr] [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: 08/24/2023] [Revised: 07/16/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
The human retinal pigment epithelium (RPE) cell line ARPE-19 is widely used as an alternative to primary RPE despite losing many features of primary RPE. We aimed to determine whether a combination of RPE-specific laminin (LN) and nicotinamide (NAM) could improve ARPE-19 redifferentiation to resemble mature RPE and improve the assessment of RPE-specific gene therapy strategies. ARPE-19 cells were propagated on tissue culture plastic supplemented with NAM and human recombinant LN521-coating. RPE maturation was performed by immunocytochemistry and gene expression by qPCR. Viral transduction experiments with adeno-associated virus (AAV)1 or AAV2, carrying a VMD2-driven GFP, were assessed at 2- and 4-weeks post-plating in the different culturing conditions with a low multiplicity of infection. The combination of LN521 coating with NAM supplementation promoted cytoskeletal and tight junction protein reorganization. The expression of maturation markers bestrophin-1 and RPE 65 was promoted concomitantly with a reduction of several epithelial-mesenchymal transition markers, such as TNF-α, TGF-β, CDH2, and vimentin. Redifferentiated ARPE-19 transduced at low multiplicity of infection of both AAV1- and AAV2-VMD2-GFP. Expression of GFP was detected at 2 weeks and increased at 4 weeks post-plating. AAV1 exhibited a greater expression efficacy compared to AAV2 in maturated ARPE-19 cells already after 2 weeks with increased efficiency after 4 weeks. Our study demonstrates an improved maturation protocol for ARPE-19 cells in vitro, mimicking an in vivo phenotype with the expression of signature genes and improved morphology. Viral-mediated RPE-specific gene expression demonstrates that the combination cultures mimic in vivo AAV tropism essential to test new gene therapies for RPE-centered diseases.
Collapse
Affiliation(s)
- Jonathan Bernd
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flavia Plastino
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Joanna Jackelin Karayannis
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Anders Kvanta
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Filippo Locri
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Elbedwehy AM, Wu J, Na HK, Baek A, Jung H, Kwon IH, Lee SW, Kim JH, Lee TG. ROS-responsive charge reversal mesoporous silica nanoparticles as promising drug delivery system for neovascular retinal diseases. J Control Release 2024; 373:224-239. [PMID: 39002796 DOI: 10.1016/j.jconrel.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Intravitreal injection of biodegradable implant drug carriers shows promise in reducing the injection frequency for neovascular retinal diseases. However, current intravitreal ocular devices have limitations in adjusting drug release rates for individual patients, thereby affecting treatment effectiveness. Accordingly, we developed mesoporous silica nanoparticles (MSNs) featuring a surface that reverse its charge in response to reactive oxygen species (ROS) for efficient delivery of humanin peptide (HN) to retinal epithelial cells (ARPE-19). The MSN core, designed with a pore size of 2.8 nm, ensures a high HN loading capacity 64.4% (w/w). We fine-tuned the external surface of the MSNs by incorporating 20% Acetyl-L-arginine (Ar) to create a partial positive charge, while 80% conjugated thioketal (TK) methoxy polyethylene glycol (mPEG) act as ROS gatekeeper. Ex vivo experiments using bovine eyes revealed the immobilization of Ar-MSNs-TK-PEG (mean zeta potential: 2 mV) in the negatively charged vitreous. However, oxidative stress reversed the surface charge to -25 mV by mPEG loss, facilitating the diffusion of the nanoparticles impeded with HN. In vitro studies showed that ARPE-19 cells effectively internalize HN-loaded Ar-MSNs-TK, subsequently releasing the peptide, which offered protection against oxidative stress-induced apoptosis, as evidenced by reduced TUNEL and caspase3 activation. The inhibition of retinal neovascularization was further validated in an in vivo oxygen-induced retinopathy (OIR) mouse model.
Collapse
Affiliation(s)
- Ahmed M Elbedwehy
- Department of Nano Science, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea; Nanotechnology Center, Mansoura University, Mansoura 35516, Egypt
| | - Jun Wu
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hee-Kyung Na
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Ahruem Baek
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Haejin Jung
- Flow Cytometry Core Facility of Research Solution Center, Institute of Basic Science, Daejeon 34126, Republic of Korea
| | - Ik Hwan Kwon
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Sang Won Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Ophthalmology, Seoul National University Hospital, Seoul 03080, Republic of Korea; Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea; Institute of Reproductive Medicine and Population, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Tae Geol Lee
- Department of Nano Science, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea.
| |
Collapse
|
42
|
Boulakh L. Intraocular and extraocular manifestations of thyroid dysfunction in Danish patients: A Nationwide Study. Acta Ophthalmol 2024; 102 Suppl 283:3-25. [PMID: 39238437 DOI: 10.1111/aos.16734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 09/07/2024]
Affiliation(s)
- Lena Boulakh
- Department of Ophthalmology, Rigshospitalet-Glostrup, Copenhagen, Denmark
| |
Collapse
|
43
|
Gemae MR, Bassi MD, Wang P, Chin EK, Almeida DR. NAD+ and Niacin Supplementation as Possible Treatments for Glaucoma and Age-Related Macular Degeneration: A Narrative Review. Nutrients 2024; 16:2795. [PMID: 39203931 PMCID: PMC11357639 DOI: 10.3390/nu16162795] [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: 07/31/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Glaucoma and age-related macular degeneration (AMD) are progressive retinal diseases characterized by increased oxidative stress, inflammation, and mitochondrial dysfunction. This review investigates the potential therapeutic benefits of NAD+ and niacin supplementation in managing glaucoma and AMD. A literature search was conducted encompassing keywords such as "niacin", "NAD", "glaucoma", "AMD", and "therapeutics". NAD+ depletion is associated with increased oxidative stress and mitochondrial dysfunction in glaucoma and AMD. Niacin, a precursor to NAD+, has shown promise in replenishing NAD+ levels, improving choroidal blood flow, and reducing oxidative damage. Animal studies in glaucoma models indicate that nicotinamide (NAM) supplementation preserves RGC density and function. Large-scale population-based studies indicate an inverse correlation between niacin intake and glaucoma prevalence, suggesting a preventative role. Randomized controlled trials assessing niacin supplementation showed significant improvements in visual field sensitivity and inner retinal function, with a dose-dependent relationship. In AMD, nicotinamide supplementation may improve rod cell function and protect against oxidative stress-induced damage. Cross-sectional studies reveal that individuals with AMD have a lower dietary intake of niacin. Further studies suggest niacin's role in improving choroidal blood flow and dilating retinal arterioles, potentially mitigating ischemic damage and oxidative stress in AMD. Beyond current management strategies, NAD+ and niacin supplementation may offer novel therapeutic avenues for glaucoma and AMD. Further research is warranted to elucidate their efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Mohamed R. Gemae
- School of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Mario D. Bassi
- School of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Patrick Wang
- Department of Ophthalmology, Kingston Health Science Centre, Queen’s University, Kingston, ON K7L 5G2, Canada
| | - Eric K. Chin
- Retina Consultants of Southern California, Redlands, CA 92374, USA
| | - David R.P. Almeida
- Erie Retina Research & Center for Advanced Surgical Exploration, Erie, PA 16507, USA
| |
Collapse
|
44
|
Wang XL, Gao YX, Yuan QZ, Zhang M. NLRP3 and autophagy in retinal ganglion cell inflammation in age-related macular degeneration: potential therapeutic implications. Int J Ophthalmol 2024; 17:1531-1544. [PMID: 39156786 PMCID: PMC11286452 DOI: 10.18240/ijo.2024.08.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/22/2024] [Indexed: 08/20/2024] Open
Abstract
Retinal degenerative diseases were a large group of diseases characterized by the primary death of retinal ganglion cells (RGCs). Recent studies had shown an interaction between autophagy and nucleotide-binding oligomerization domain-like receptor 3 (NLRP3) inflammasomes, which may affect RGCs in retinal degenerative diseases. The NLRP3 inflammasome was a protein complex that, upon activation, produces caspase-1, mediating the apoptosis of retinal cells and promoting the occurrence and development of retinal degenerative diseases. Upregulated autophagy could inhibit NLRP3 inflammasome activation, while inhibited autophagy can promote NLRP3 inflammasome activation, which leaded to the accelerated emergence of drusen and lipofuscin deposition under the neurosensory retina. The activated NLRP3 inflammasome could further inhibit autophagy, thus forming a vicious cycle that accelerated the damage and death of RGCs. This review discussed the relationship between NLRP3 inflammasome and autophagy and its effects on RGCs in age-related macular degeneration, providing a new perspective and direction for the treatment of retinal diseases.
Collapse
Affiliation(s)
- Xiao-Li Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yun-Xia Gao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiong-Zhen Yuan
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ming Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
45
|
Ozturk E, Cankaya C, Yildizli Y. Correlation between corneal endothelial layer features and age-related macular degeneration severity. Int Ophthalmol 2024; 44:345. [PMID: 39122890 DOI: 10.1007/s10792-024-03271-x] [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: 03/20/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
PURPOSE This study aimed to investigate the relationship between corneal endothelial layer features and the severity of age-related macular degeneration (AMD). METHODS The study included 119 patients, with 47 females and 72 males. Patients were categorized into four groups based on the AREDS grading system: no AMD (group 1), mild AMD (group 2), moderate AMD (group 3), and advanced AMD (group 4). Only the right eye of patients with both eyes suitable for the study was included. Corneal endothelial cell density (CD), coefficient of variation (CoV), hexagonal cell ratio (HEX), and central corneal thickness (CCT) were measured using specular microscopy (Konan Medical Inc., Nishinomiya, Japan). RESULTS Group 1 had 40 patients, group 2 had 27 patients, and groups 3 and 4 had 26 patients each. Significant differences were observed between the mean endothelial CD, CoV, and HEX values among the groups, while no significant difference was found in CCT values (p = 0.049, p = 0.002, p = 0.004, and p = 0.883, respectively). A mild negative correlation was observed between AMD severity and CD and HEX values, while a mild positive correlation was found between AMD severity and CoV. CONCLUSION Increasing severity of AMD may negatively impact corneal endothelial layer values.
Collapse
Affiliation(s)
- Emrah Ozturk
- Department of Ophthalmology, Inonu University School of Medicine, Malatya, Turkey.
- Department of Ophthalmology, Malatya Turgut Ozal University School of Medicine, Malatya, Turkey.
| | - Cem Cankaya
- Department of Ophthalmology, Inonu University School of Medicine, Malatya, Turkey
| | - Yakup Yildizli
- Department of Ophthalmology, Inonu University School of Medicine, Malatya, Turkey
| |
Collapse
|
46
|
Liu R, Dang JN, Lee R, Lee JJ, Kesavamoorthy N, Ameri H, Rao N, Eoh H. Mycobacterium dormancy and antibiotic tolerance within the retinal pigment epithelium of ocular tuberculosis. Microbiol Spectr 2024; 12:e0078824. [PMID: 38916325 PMCID: PMC11302011 DOI: 10.1128/spectrum.00788-24] [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: 03/28/2024] [Accepted: 05/23/2024] [Indexed: 06/26/2024] Open
Abstract
Tuberculosis (TB) is a leading cause of death among infectious diseases worldwide due to latent TB infection, which is the critical step for the successful pathogenic cycle. In this stage, Mycobacterium tuberculosis resides inside the host in a dormant and antibiotic-tolerant state. Latent TB infection can also lead to multisystemic diseases because M. tuberculosis invades virtually all organs, including ocular tissues. Ocular tuberculosis (OTB) occurs when the dormant bacilli within the ocular tissues reactivate, originally seeded by hematogenous spread from pulmonary TB. Histological evidence suggests that retinal pigment epithelium (RPE) cells play a central role in immune privilege and in protection from antibiotic effects, making them an anatomical niche for invading M. tuberculosis. RPE cells exhibit high tolerance to environmental redox stresses, allowing phagocytosed M. tuberculosis bacilli to maintain viability in a dormant state. However, the microbiological and metabolic mechanisms determining the interaction between the RPE intracellular environment and phagocytosed M. tuberculosis are largely unknown. Here, liquid chromatography-mass spectrometry metabolomics were used to illuminate the metabolic state within RPE cells reprogrammed to harbor dormant M. tuberculosis bacilli and enhance antibiotic tolerance. Timely and accurate diagnosis as well as efficient chemotherapies are crucial in preventing the poor visual outcomes of OTB patients. Unfortunately, the efficacy of current methods is highly limited. Thus, the results will lead to propose a novel therapeutic option to synthetically kill the dormant M. tuberculosis inside the RPE cells by modulating the phenotypic state of M. tuberculosis and laying the foundation for a new, innovative regimen for treating OTB. IMPORTANCE Understanding the metabolic environment within the retinal pigment epithelium (RPE) cells altered by infection with Mycobacterium tuberculosis and mycobacterial dormancy is crucial to identify new therapeutic methods to cure ocular tuberculosis. The present study showed that RPE cellular metabolism is altered to foster intracellular M. tuberculosis to enter into the dormant and drug-tolerant state, thereby blunting the efficacy of anti-tuberculosis chemotherapy. RPE cells serve as an anatomical niche as the cells protect invading bacilli from antibiotic treatment. LC-MS metabolomics of RPE cells after co-treatment with H2O2 and M. tuberculosis infection showed that the intracellular environment within RPE cells is enriched with a greater level of oxidative stress. The antibiotic tolerance of intracellular M. tuberculosis within RPE cells can be restored by a metabolic manipulation strategy such as co-treatment of antibiotic with the most downstream glycolysis metabolite, phosphoenolpyruvate.
Collapse
Affiliation(s)
- Rachel Liu
- Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Joshua N. Dang
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rhoeun Lee
- Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Jae Jin Lee
- Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Niranjana Kesavamoorthy
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hossein Ameri
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Narsing Rao
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hyungjin Eoh
- Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
47
|
Cvekl A, Vijg J. Aging of the eye: Lessons from cataracts and age-related macular degeneration. Ageing Res Rev 2024; 99:102407. [PMID: 38977082 DOI: 10.1016/j.arr.2024.102407] [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: 03/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
Aging is the greatest risk factor for chronic human diseases, including many eye diseases. Geroscience aims to understand the effects of the aging process on these diseases, including the genetic, molecular, and cellular mechanisms that underlie the increased risk of disease over the lifetime. Understanding of the aging eye increases general knowledge of the cellular physiology impacted by aging processes at various biological extremes. Two major diseases, age-related cataract and age-related macular degeneration (AMD) are caused by dysfunction of the lens and retina, respectively. Lens transparency and light refraction are mediated by lens fiber cells lacking nuclei and other organelles, which provides a unique opportunity to study a single aging hallmark, i.e., loss of proteostasis, within an environment of limited metabolism. In AMD, local dysfunction of the photoreceptors/retinal pigmented epithelium/Bruch's membrane/choriocapillaris complex in the macula leads to the loss of photoreceptors and eventually loss of central vision, and is driven by nearly all the hallmarks of aging and shares features with Alzheimer's disease, Parkinson's disease, cardiovascular disease, and diabetes. The aging eye can function as a model for studying basic mechanisms of aging and, vice versa, well-defined hallmarks of aging can be used as tools to understand age-related eye disease.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jan Vijg
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
48
|
Servillo A, Sacconi R, Oldoni G, Barlocci E, Tombolini B, Battista M, Fantaguzzi F, Rissotto F, Mularoni C, Parravano M, Zucchiatti I, Querques L, Bandello F, Querques G. Advancements in Imaging and Therapeutic Options for Dry Age-Related Macular Degeneration and Geographic Atrophy. Ophthalmol Ther 2024; 13:2067-2082. [PMID: 38833127 PMCID: PMC11246354 DOI: 10.1007/s40123-024-00970-7] [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: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss in the elderly, with dry AMD (d-AMD) leading to geographic atrophy (GA) and significant visual impairment. Multimodal imaging plays a crucial role in d-AMD diagnosis and management, allowing for detailed classification of patient phenotypes and aiding in treatment planning and prognosis determination. Treatment approaches for d-AMD have recently witnessed profound change with the development of specific drugs targeting the complement cascade, with the first anticomplement agents recently approved for GA treatment. Additionally, emerging strategies such as gene therapy and laser treatments may offer potential benefits, though further research is needed to fully establish their efficacy. However, the lack of effective therapies capable of restoring damaged retinal cells remains a major challenge. In the future, genetic treatments aimed at preventing the progression of d-AMD may emerge as a powerful approach. Currently, however, their development is still in the early stages.
Collapse
Affiliation(s)
- Andrea Servillo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Riccardo Sacconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gloria Oldoni
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Eugenio Barlocci
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Beatrice Tombolini
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Marco Battista
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Federico Fantaguzzi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Federico Rissotto
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Cecilia Mularoni
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | | | - Ilaria Zucchiatti
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Lea Querques
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Francesco Bandello
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giuseppe Querques
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
49
|
Preya UH, Sayed S, Nguyen NL, Kim JT. Potential role of CTSS in AMDImmune modulatory and anti-angiogenic effects of cathepsin S knockdown in ARPE-19 cells. Exp Eye Res 2024; 245:109981. [PMID: 38914301 DOI: 10.1016/j.exer.2024.109981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
We aimed to determine the role of cathepsin S (CTSS) in modulating oxidative stress-induced immune and inflammatory reactions and angiogenesis in age-related macular degeneration. Human retinal pigment epithelium cells line ARPE-19 (immature) were maintained and treated with H2O2. The expression of CTSS, inflammatory cytokines, and complement factors induced by oxidative stress was compared between cells incubated without (control) and with CTSS knockdown (using small interfering ribonucleic acid; siRNA). To evaluate the role of CTSS in angiogenesis, we assayed tube formation using human umbilical vein endothelial cells and conditioned medium from ARPE-19 cells. We also used a mouse model of laser-induced choroidal neovascularization. CTSS levels were higher in ARPE-19 cells treated with H2O2 than in control cells. Oxidative stress-induced CTSS resulted in significantly elevated transcription of nuclear factor kappa B-dependent inflammatory cytokines, complement factors C3a and C5a, membrane attack complex (C5b-9), and C3a and C5a receptors. siRNA-mediated knockdown of CTSS reduced the number of inflammatory signals. Furthermore, oxidative stress-induced CTSS regulated the expression of peroxisome proliferator-activated receptor γ and vascular endothelial growth factor A/Akt serine/threonine kinase family signaling, which led to angiogenesis. Tube formation assays and mouse models of choroidal neovascularization revealed that CTSS knockdown ameliorated angiogenesis in vitro and in vivo. The present findings suggest that CTSS modulates the complement pathway, inflammatory reactions, and neovascularization, and that CTSS knockdown induces potent immunomodulatory effects. Hence, it could be a promising target for the prevention and treatment of early- and late-stage age-related macular degeneration.
Collapse
Affiliation(s)
- Umma Hafsa Preya
- Ophthalmology Department, School of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Shithima Sayed
- Ophthalmology Department, School of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Ngoc Lan Nguyen
- Ophthalmology Department, School of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Jee Taek Kim
- Ophthalmology Department, School of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea; Chung-Ang University Hospital, Dongjak-gu, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Wagner N, Tsai T, Reinehr S, Theile J, Dick HB, Joachim SC. Retinal debris triggers cytotoxic damage in cocultivated primary porcine RPE cells. Front Neurosci 2024; 18:1401571. [PMID: 39114482 PMCID: PMC11303199 DOI: 10.3389/fnins.2024.1401571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction One of the most common causes of vision loss in the elderly population worldwide is age-related macular degeneration (AMD). Subsequently, the number of people affected by AMD is estimated to reach approximately 288 million by the year 2040. The aim of this study was to develop an ex vivo model that simulates various aspects of the complex AMD pathogenesis. Methods For this purpose, primary porcine retinal pigment epithelial cells (ppRPE) were isolated and cultured. One group was exposed to medium containing sodium iodate (NaIO3) to induce degeneration. The others were exposed to different supplemented media, such as bovine serum albumin (BSA), homogenized porcine retinas (HPR), or rod outer segments (ROOS) for eight days to promote retinal deposits. Then, these ppRPE cells were cocultured with porcine neuroretina explants for another eight days. To assess the viability of ppRPE cells, live/dead assay was performed at the end of the study. The positive RPE65 and ZO1 area was evaluated by immunocytochemistry and the expression of RLBP1, RPE65, and TJP1 was analyzed by RT-qPCR. Additionally, drusen (APOE), inflammation (ITGAM, IL6, IL8, NLRP3, TNF), oxidative stress (NFE2L2, SOD1, SOD2), and hypoxia (HIF1A) markers were investigated. The concentration of the inflammatory cytokines IL-6 and IL-8 was determined in medium supernatants from day 16 and 24 via ELISA. Results Live/dead assay suggests that especially exposure to NaIO3 and HPR induced damage to ppRPE cells, leading in a significant ppRPE cell loss. All supplemented media resulted in decreased RPE-characteristic markers (RPE65; ZO-1) and gene expression like RLBP1 and RPE65 in the cultured ppRPE cells. Besides, some inflammatory, oxidative as well as hypoxic stress markers were altered in ppRPE cells cultivated with NaIO3. The application of HPR induced an enhanced APOE expression. Pre-exposure of the ppRPE cells led to a diminished number of cones in all supplemented media groups compared to controls. Discussion Overall, this novel coculture model represents an interesting initial approach to incorporating deposits into coculture to mimic AMD pathogenesis. Nevertheless, the effects of the media used need to be investigated in further studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|