1
|
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 2024; 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] [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
|
2
|
Rajala A, Rajala RVS. Age-Related Changes in the Glycolytic Enzymes of M2-Isoform of Pyruvate Kinase and Fructose-1,6-Bisphosphate Aldolase: Implications to Age-Related Macular Degeneration. Aging Dis 2024; 15:2271-2283. [PMID: 38739943 PMCID: PMC11346409 DOI: 10.14336/ad.2024.0077] [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: 01/22/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
Prior studies have emphasized a bioenergetic crisis in the retinal pigment epithelium (RPE) as a critical factor in the development of age-related macular degeneration (AMD). The isoforms Fructose-1,6-bisphosphate aldolase C (ALDOC) and pyruvate kinase M2 (PKM2) have been proposed to play a role in AMD pathogenesis. While PKM2 and ALDOC are crucial for aerobic glycolysis in the neural retina, they are not as essential for the RPE. In this study, we examined the expression and activity of PKM2 and ALDOC in both young and aged RPE cells, as well as in the retina and RPE tissue of mice, including an experimentally induced AMD mouse model. Our findings reveal an upregulation in PKM2 and ALDOC expression, accompanied by increased pyruvate kinase activity, in the aged and AMD mouse RPE. Conversely, there is a decrease in ALDOC expression but an increase in PKM2 expression and pyruvate kinase activity in the aged and AMD retina. Overall, our study indicates that aged and AMD RPE cells tend to favor aerobic glycolysis, while this tendency is diminished in the aged and AMD retina. These results underscore the significance of targeting PKM2 and ALDOC in the RPE as a promising therapeutic approach to address the bioenergetic crisis and prevent vision loss in AMD.
Collapse
Affiliation(s)
- Ammaji Rajala
- Department of Ophthalmology
- Dean McGee Eye Institute, Oklahoma, Oklahoma 73104, USA.
| | - Raju V. S. Rajala
- Department of Ophthalmology
- Department of Biochemistry and Physiology, and
- Department of Cell Biology, University of Oklahoma Health Sciences Center
- Dean McGee Eye Institute, Oklahoma, Oklahoma 73104, USA.
| |
Collapse
|
3
|
Lee PY, Bui BV. Age-related differences in retinal function and structure in C57BL/6J and Thy1-YFPh mice. Neurobiol Aging 2024; 141:171-181. [PMID: 38964014 DOI: 10.1016/j.neurobiolaging.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
Age-related neuronal adaptations are known to help maintain function. This study aims to examine gross age-related in vivo retinal functional adaptations (using electroretinography) in young and middle aged C57BL/6J and Thy1-YFPh mice and to relate this to in vivo retinal structure (using optical coherence tomography). Electroretinography responses were generally larger in Thy1-YFPh mice than in C57BL/6J mice, with similar in vivo retinal layer thicknesses except for longer inner/outer photoreceptor segment in Thy1-YFPh mice. Relative to 3-month-old mice, 12-month-old mice showed reduced photoreceptor (C57BL/6J 84.0±2.5 %; Thy1-YFPh 80.2±5.2 %) and bipolar cell (C57BL/6J 75.6±2.3 %; Thy1-YFPh 68.1±5.5 %) function. There was relative preservation of ganglion cell function (C57BL/6J 79.7±3.7 %; Thy1-YFPh 91.7±5.0 %) with age, which was associated with increased b-wave (bipolar cell) sensitivities to light. Ganglion cell function was correlated with both b-wave amplitude and sensitivity. This study shows that there are normal age-related adaptations to preserve functional output. Different mouse strains may have varied age-related adaptation capacity and should be taken into consideration when examining age-related susceptibility to injury.
Collapse
Affiliation(s)
- Pei Ying Lee
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
4
|
Zhou Y, Zhou W, Rao Y, He J, Huang Y, Zhao P, Li J. Dysregulated energy and protein homeostasis and the loss of GABAergic amacrine cells in aging retina. Exp Eye Res 2024; 245:109985. [PMID: 38945518 DOI: 10.1016/j.exer.2024.109985] [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: 02/26/2024] [Revised: 05/30/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Aging is a major risk factor for the development or the worsening of retinal degenerative conditions. The intricate network of the neural retina determined that the retinal aging is a complicated process. The aim of this study is to delineate the transcriptomic changes of major retinal neurons during aging in C57BL/6 mice at single-cell level. We analyzed the transcriptional profiles of the photoreceptor, bipolar, amacrine, and Müller glial cells of 1.5-2 and 24-30 months old mice using single-cell RNA sequencing technique. We selectively confirmed the differences in gene expression using immunofluorescence staining and RNA in situ hybridization analysis. We found that each retinal cell type had unique changes upon aging. However, they all showed signs of dysregulated glucose and energy metabolism, and perturbed proteostasis. In particular, old Müller glia exhibited the most profound changes, including the upregulation of cell metabolism, stress-responses, antigen-presentation and immune responses and metal ion homeostasis. The dysregulated gliogenesis and differentiation was confirmed by the presence of Müller glia expressing rod-specific genes in the inner nuclear layer and the outer plexiform layer of the old retina. We further pinpointed the specific loss of GABAergic amacrine cells in old retina. Our study emphasized changes of amacrine and Müller glia during retinal aging, provided resources for further research on the molecular and cellular regulatory mechanisms underlying aging-associated retinal deterioration.
Collapse
Affiliation(s)
- Yutong Zhou
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Wenchuan Zhou
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuqing Rao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jincan He
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yue Huang
- Department of Ophthalmology, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 202150, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
5
|
Mu W, Han X, Tong M, Ben S, Yao M, Zhao Y, Xia J, Ren L, Huang C, Li D, Li X, Jiang Q, Yan B. Identification of the Metabolic Signature of Aging Retina. Transl Vis Sci Technol 2024; 13:8. [PMID: 39102240 PMCID: PMC11309042 DOI: 10.1167/tvst.13.8.8] [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: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 08/06/2024] Open
Abstract
Purpose This study aims to explore the metabolic signature of aging retina and identify the potential metabolic biomarkers for the diagnosis of retinal aging. Methods Retinal samples were collected from both young (two months) and aging (14 months) mice to conduct an unbiased metabolic profiling. Liquid chromatography-tandem mass spectrometry analysis was conducted to screen for the metabolic biomarkers and altered signaling pathways associated with retinal aging. Results We identified 166 metabolites differentially expressed between young and aged retinas using a threshold of orthogonal projection to latent structures discriminant analysis variable importance in projection >1 and P < 0.05. These metabolites were significantly enriched in several metabolic pathways, including purine metabolism, citrate cycle, phenylalanine, tyrosine and tryptophan biosynthesis, glycerophospholipid metabolism, and alanine, aspartate and glutamate metabolism. Among these significantly enriched pathways, glycerophospholipid metabolites emerged as promising candidates for retinal aging biomarkers. We assessed the potential of these metabolites as biomarkers through an analysis of their sensitivity and specificity, determined by the area under the receiver-operating characteristic (ROC) curves. Notably, the metabolites like PC (15:0/22:6), PC (17:0/14:1), LPC (P-16:0), PE (16:0/20:4), and PS (17:0/16:1) demonstrated superior performance in sensitivity, specificity, and accuracy in predicting retinal aging. Conclusions This study sheds light on the molecular mechanisms underlying retinal aging by identifying distinct metabolic profiles and pathways. These findings provide a valuable foundation for developing future clinical applications in diagnosing, identifying, and treating age-related retinal degeneration. Translational Relevance This study sheds light on novel metabolic profiles and biomarkers in aging retinas, potentially paving the way for targeted interventions in preventing, diagnosing, and treating age-related retinal degeneration and other retinal diseases.
Collapse
Affiliation(s)
- Wan Mu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Han
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Ming Tong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Shuai Ben
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mudi Yao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Xia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Ren
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Chang Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Duo Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Künzel SE, Frentzel DP, Flesch LTM, Knecht VA, Rübsam A, Dreher F, Schütte M, Dubrac A, Lange B, Yaspo ML, Lehrach H, Joussen AM, Zeitz O. AI-driven discovery of blood xenobiotic biomarkers in neovascular age-related macular degeneration using iterative random forests. Graefes Arch Clin Exp Ophthalmol 2024:10.1007/s00417-024-06538-2. [PMID: 38842593 DOI: 10.1007/s00417-024-06538-2] [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: 03/04/2024] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024] Open
Abstract
PURPOSE To investigate the xenobiotic profiles of patients with neovascular age-related macular degeneration (nAMD) undergoing anti-vascular endothelial growth factor (anti-VEGF) intravitreal therapy (IVT) to identify biomarkers indicative of clinical phenotypes through advanced AI methodologies. METHODS In this cross-sectional observational study, we analyzed 156 peripheral blood xenobiotic features in a cohort of 46 nAMD patients stratified by choroidal neovascularization (CNV) control under anti-VEGF IVT. We employed Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) for measurement and leveraged an AI-driven iterative Random Forests (iRF) approach for robust pattern recognition and feature selection, aligning molecular profiles with clinical phenotypes. RESULTS AI-augmented iRF models effectively refined the metabolite spectrum by discarding non-predictive elements. Perfluorooctanesulfonate (PFOS) and Ethyl β-glucopyranoside were identified as significant biomarkers through this process, associated with various clinically relevant phenotypes. Unlike single metabolite classes, drug metabolites were distinctly correlated with subretinal fluid presence. CONCLUSIONS This study underscores the enhanced capability of AI, particularly iRF, in dissecting complex metabolomic data to elucidate the xenobiotic landscape of nAMD and environmental impact on the disease. The preliminary biomarkers discovered offer promising directions for personalized treatment strategies, although further validation in broader cohorts is essential for clinical application.
Collapse
Affiliation(s)
- Steffen E Künzel
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Dominik P Frentzel
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203, Berlin, Germany
| | - Leonie T M Flesch
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203, Berlin, Germany
| | - Vitus A Knecht
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203, Berlin, Germany
| | - Anne Rübsam
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203, Berlin, Germany
| | - Felix Dreher
- Alacris Theranostics, Max-Planck-Straße 3, 12489, Berlin, Germany
| | - Moritz Schütte
- Alacris Theranostics, Max-Planck-Straße 3, 12489, Berlin, Germany
| | - Alexandre Dubrac
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, H3C 3J7, Canada
| | - Bodo Lange
- Alacris Theranostics, Max-Planck-Straße 3, 12489, Berlin, Germany
| | - Marie-Laure Yaspo
- Max-Planck-Institute for Molecular Genetics, Ihnestrasse 63-73, 14195, Berlin, Germany
| | - Hans Lehrach
- Max-Planck-Institute for Molecular Genetics, Ihnestrasse 63-73, 14195, Berlin, Germany
| | - Antonia M Joussen
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203, Berlin, Germany
| | - Oliver Zeitz
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Campus Benjamin Franklin (CBF), Hindenburgdamm 30, 12203, Berlin, Germany
| |
Collapse
|
7
|
Tang H, Luo N, Zhang X, Huang J, Yang Q, Lin H, Zhang X. Association between biological aging and diabetic retinopathy. Sci Rep 2024; 14:10123. [PMID: 38698194 PMCID: PMC11065862 DOI: 10.1038/s41598-024-60913-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: 12/27/2023] [Accepted: 04/29/2024] [Indexed: 05/05/2024] Open
Abstract
The impact of aging on diabetic retinopathy (DR) remains underestimated. The current study aimed to investigate the association between biological aging and DR, in contrast to chronological age (CA). Using the National Health and Nutrition Survey data from 2005 to 2008. Biological aging was evaluated through the biological age (BA) and phenotypic age (PA), which were calculated from clinical markers. DR was identified in participants with diabetes mellitus (DM) when they exhibited one or more retinal microaneurysms or retinal blot hemorrhages under retinal imaging, with or without the presence of more severe lesions. Survey-weighted multivariable logistic regression was performed, and the regression model was further fitted using restricted cubic splines. The discriminatory capability and clinical utility of the model were evaluated using receiver operating characteristic (ROC) curves and decision curve analysis (DCA). Based on weighted analyses, of the 3100 participants included in this study, of which 162 had DR. In the adjusted model, BA (odds ratio [OR] = 1.12, 95% CI, 1.06-1.18) and PA (OR = 1.11, 95% CI, 1.07-1.14) were associated with DR, while CA was not significantly (OR = 1.01, 95% CI, 0.99-1.03). Narrowing the analysis to DM participants and adjusting for factors like insulin showed similar results. ROC and DCA analyses indicate that BA/PA predicted DR better than CA and offer greater clinical utility. The positive association between BA/PA and DR was consistent across subgroups despite potential interactions. Biological aging heightens DR risk, with BA/PA showing a stronger association than CA. Our findings underscored the importance of timely anti-aging interventions for preventing DR.
Collapse
Affiliation(s)
- Haoxian Tang
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Nan Luo
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Psychiatry, Shantou University Mental Health Center, Shantou, Guangdong, China
| | - Xuan Zhang
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jingtao Huang
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Qinglong Yang
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Urology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hanyuan Lin
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Urology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xinyi Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, 515041, Guangdong, China.
| |
Collapse
|
8
|
Mérida S, Návea A, Desco C, Celda B, Pardo-Tendero M, Morales-Tatay JM, Bosch-Morell F. Glutathione and a Pool of Metabolites Partly Related to Oxidative Stress Are Associated with Low and High Myopia in an Altered Bioenergetic Environment. Antioxidants (Basel) 2024; 13:539. [PMID: 38790644 PMCID: PMC11117864 DOI: 10.3390/antiox13050539] [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: 03/29/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Oxidative stress forms part of the molecular basis contributing to the development and manifestation of myopia, a refractive error with associated pathology that is increasingly prevalent worldwide and that subsequently leads to an upsurge in degenerative visual impairment due to conditions that are especially associated with high myopia. The purpose of our study was to examine the interrelation of potential oxidative-stress-related metabolites found in the aqueous humor of high-myopic, low-myopic, and non-myopic patients within a clinical study. We conducted a cross-sectional study, selecting two sets of patients undergoing cataract surgery. The first set, which was used to analyze metabolites through an NMR assay, comprised 116 patients. A total of 59 metabolites were assigned and quantified. The PLS-DA score plot clearly showed a separation with minimal overlap between the HM and control samples. The PLS-DA model allowed us to determine 31 major metabolite differences in the aqueous humor of the study groups. Complementary statistical analysis of the data allowed us to determine six metabolites that presented significant differences among the experimental groups (p < 005). A significant number of these metabolites were discovered to have a direct or indirect connection to oxidative stress linked with conditions of myopic eyes. Notably, we identified metabolites associated with bioenergetic pathways and metabolites that have undergone methylation, along with choline and its derivatives. The second set consisted of 73 patients who underwent a glutathione assay. Here, we showed significant variations in both reduced and oxidized glutathione in aqueous humor among all patient groups (p < 0.01) for the first time. Axial length, refractive status, and complete ophthalmologic examination were also recorded, and interrelations among metabolic and clinical parameters were evaluated.
Collapse
Affiliation(s)
- Salvador Mérida
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, Alfara del Patriarca, 46115 Valencia, Spain; (S.M.); (C.D.)
| | - Amparo Návea
- Instituto de la Retina y Enfermedades Oculares, 46005 Valencia, Spain;
| | - Carmen Desco
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, Alfara del Patriarca, 46115 Valencia, Spain; (S.M.); (C.D.)
- Instituto de la Retina y Enfermedades Oculares, 46005 Valencia, Spain;
- FOM, Fundación de Oftalmología Médica de la Comunidad Valenciana, 46015 Valencia, Spain
| | - Bernardo Celda
- Physical Chemistry Department, University of Valencia, 46100 Valencia, Spain;
| | - Mercedes Pardo-Tendero
- Department of Pathology, Medicine and Odontology Faculty, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - José Manuel Morales-Tatay
- Department of Pathology, Medicine and Odontology Faculty, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Francisco Bosch-Morell
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, Alfara del Patriarca, 46115 Valencia, Spain; (S.M.); (C.D.)
| |
Collapse
|
9
|
Xia Y, Chen K, Yang Q, Chen Z, Jin L, Zhang L, Yu X, Wang L, Xie C, Zhao Y, Shen Y, Tong J. Methylation in cornea and corneal diseases: a systematic review. Cell Death Discov 2024; 10:169. [PMID: 38589350 PMCID: PMC11002037 DOI: 10.1038/s41420-024-01935-2] [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: 11/30/2023] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/10/2024] Open
Abstract
Corneal diseases are among the primary causes of blindness and vision loss worldwide. However, the pathogenesis of corneal diseases remains elusive, and diagnostic and therapeutic tools are limited. Thus, identifying new targets for the diagnosis and treatment of corneal diseases has gained great interest. Methylation, a type of epigenetic modification, modulates various cellular processes at both nucleic acid and protein levels. Growing evidence shows that methylation is a key regulator in the pathogenesis of corneal diseases, including inflammation, fibrosis, and neovascularization, making it an attractive potential therapeutic target. In this review, we discuss the major alterations of methylation and demethylation at the DNA, RNA, and protein levels in corneal diseases and how these dynamics contribute to the pathogenesis of corneal diseases. Also, we provide insights into identifying potential biomarkers of methylation that may improve the diagnosis and treatment of corneal diseases.
Collapse
Affiliation(s)
- Yutong Xia
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Kuangqi Chen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Qianjie Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Zhitong Chen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Le Jin
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Liyue Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Xin Yu
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Liyin Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Chen Xie
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Yuan Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China.
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
10
|
Sun R, Feng J, Wang J. Underlying Mechanisms and Treatment of Cellular Senescence-Induced Biological Barrier Interruption and Related Diseases. Aging Dis 2024; 15:612-639. [PMID: 37450933 PMCID: PMC10917536 DOI: 10.14336/ad.2023.0621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Given its increasing prevalence, aging is of great concern to researchers worldwide. Cellular senescence is a physiological or pathological cellular state caused by aging and a prominent risk factor for the interruption of the integrity and functionality of human biological barriers. Health barriers play an important role in maintaining microenvironmental homeostasis within the body. The senescence of barrier cells leads to barrier dysfunction and age-related diseases. Cellular senescence has been reported to be a key target for the prevention of age-related barrier diseases, including Alzheimer's disease, Parkinson's disease, age-related macular degeneration, diabetic retinopathy, and preeclampsia. Drugs such as metformin, dasatinib, quercetin, BCL-2 inhibitors, and rapamycin have been shown to intervene in cellular senescence and age-related diseases. In this review, we conclude that cellular senescence is involved in age-related biological barrier impairment. We further outline the cellular pathways and mechanisms underlying barrier impairment caused by cellular senescence and describe age-related barrier diseases associated with senescent cells. Finally, we summarize the currently used anti-senescence pharmacological interventions and discuss their therapeutic potential for preventing age-related barrier diseases.
Collapse
Affiliation(s)
- Ruize Sun
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Wang K, Pu Y, Chen L, Hoshino M, Uesugi K, Yagi N, Chen X, Usui Y, Hanashima A, Hashimoto K, Mohri S, Pierscionek BK. Optical development in the murine eye lens of accelerated senescence-prone SAMP8 and senescence-resistant SAMR1 strains. Exp Eye Res 2024; 241:109858. [PMID: 38467176 DOI: 10.1016/j.exer.2024.109858] [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: 01/07/2024] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
The eye lens is responsible for focusing objects at various distances onto the retina and its refractive power is determined by its surface curvature as well as its internal gradient refractive index (GRIN). The lens continues to grow with age resulting in changes to the shape and to the GRIN profile. The present study aims to investigate how the ageing process may influence lens optical development. Murine lenses of accelerated senescence-prone strain (SAMP8) aged from 4 to 50 weeks; senescence-resistant strain (SAMR1) aged from 5 to 52 weeks as well as AKR strain (served as control) aged from 6 to 70 weeks were measured using the X-ray interferometer at the SPring-8 synchrotron Japan within three consecutive years from 2020 to 2022. Three dimensional distributions of the lens GRIN were reconstructed using the measured data and the lens shapes were determined using image segmentation in MatLab. Variations in the parameters describing the lens shape and the GRIN profile with age were compared amongst three mouse strains. With advancing age, both the lens anterior and posterior surface flattens and the lens sagittal thickness increase in all three mouse strains (Anterior radius of curvature increase at 0.008 mm/week, 0.007 mm/week and 0.002 mm/week while posterior radius of curvature increase at 0.002 mm/week, 0.007 mm/week and 0.003 mm/week respectively in AKR, SAMP8 and SAMR1 lenses). Compared with the AKR strain, the SAMP8 samples demonstrate a higher rate of increase in the posterior curvature radius (0.007 mm/week) and the thickness (0.015 mm/week), whilst the SAMR1 samples show slower increases in the anterior curvature radius (0.002 mm/week) and its thickness (0.013 mm/week). There are similar age-related trends in GRIN shape in the radial direction (in all three types of murine lenses nr2 and nr6 increase with age while nr4 decrease with age consistently) but not in the axial direction amongst three mouse strains (nz1 of AKR lens decrease while of SAMP8 and SAMR1 increase with age; nz2 of all three models increase with age; nz3 of AKR lens increase while of SAMP8 and SAMR1 decrease with age). The ageing process can influence the speed of lens shape change and affect the GRIN profile mainly in the axial direction, contributing to an accelerated decline rate of the optical power in the senescence-prone strain (3.5 D/week compared to 2.3 D/week in the AKR control model) but a retardatory decrease in the senescence-resistant strain (2.1 D/week compared to the 2.3D/week in the AKR control model).
Collapse
Affiliation(s)
- Kehao Wang
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yutian Pu
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Leran Chen
- Peking University First Hospital, Beijing, China.
| | - Masato Hoshino
- Japan Synchrotron Radiation Research Institute (Spring-8), 1-1-1, Kouto, Sayo-cho, Sayo-gun, Hyogo, 679-5198, Japan.
| | - Kentaro Uesugi
- Japan Synchrotron Radiation Research Institute (Spring-8), 1-1-1, Kouto, Sayo-cho, Sayo-gun, Hyogo, 679-5198, Japan.
| | - Naoto Yagi
- Japan Synchrotron Radiation Research Institute (Spring-8), 1-1-1, Kouto, Sayo-cho, Sayo-gun, Hyogo, 679-5198, Japan.
| | - Xiaoyong Chen
- Department of Ophthalmology, Peking University Third Hospital, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, China.
| | - Yuu Usui
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Akira Hanashima
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Ken Hashimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Barbara K Pierscionek
- Faculty of Health, Education, Medicine and Social Care, Medical Technology Research Centre, Anglia Ruskin University, Bishops Hall Lane, Chelmsford, United Kingdom.
| |
Collapse
|
12
|
Oganov AC, Seddon I, Zein M, Yazdanpanah G, Fonoudi H, Jabbehdari S. Composition of the gut microbiome, role of diet, lifestyle, and antioxidant therapies in diabetes mellitus and diabetic retinopathy. Eur J Ophthalmol 2024; 34:367-383. [PMID: 37150930 DOI: 10.1177/11206721231174490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The gut microbiome is a complex ecosystem in the gastrointestinal tract composed of trillions of bacteria, viruses, fungi, and protozoa. Disruption of this delicate ecosystem, formally called "dysbiosis", has been linked to a variety of metabolic and inflammatory pathologies. Several studies have focused on abnormal microbiome composition and correlated these findings with the development of type 2 diabetes mellitus (T2DM) and diabetic retinopathy (DR). However, given the complexity of this ecosystem, the current studies are narrow in design and present variable findings. Composition of the gut microbiome in patients with DR significantly differs from patients with diabetes without retinopathy as well as from healthy controls. Additionally, the gut microbiome has been shown to modify effects of medication, diet, exercise, and antioxidant use on the development and progression of DR. In this paper, we present a comprehensive review of literature on the effect of oxidative stress, antioxidant therapies, and dysbiosis on DR.
Collapse
Affiliation(s)
- Anthony C Oganov
- Department of Ophthalmology, Renaissance School of Medicine, Stony Brook, NY, USA
| | - Ian Seddon
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Mike Zein
- Department of Ophthalmology, Cook County Health, Chicago, IL, USA
| | - Ghasem Yazdanpanah
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Hossein Fonoudi
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Sayena Jabbehdari
- Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
13
|
Shelton DA, Gefke I, Summers V, Kim YK, Yu H, Getz Y, Ferdous S, Donaldson K, Liao K, Papania JT, Chrenek MA, Boatright JH, Nickerson JM. Age-Related RPE changes in Wildtype C57BL/6J Mice between 2 and 32 Months. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.574142. [PMID: 38352604 PMCID: PMC10862734 DOI: 10.1101/2024.01.30.574142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Purpose This study provides a systematic evaluation of age-related changes in RPE cell structure and function using a morphometric approach. We aim to better capture nuanced predictive changes in cell heterogeneity that reflect loss of RPE integrity during normal aging. Using C57BL6/J mice ranging from P60-P730, we sought to evaluate how regional changes in RPE shape reflect incremental losses in RPE cell function with advancing age. We hypothesize that tracking global morphological changes in RPE is predictive of functional defects over time. Methods We tested three groups of C57BL/6J mice (young: P60-180; Middle-aged: P365-729; aged: 730+) for function and structural defects using electroretinograms, immunofluorescence, and phagocytosis assays. Results The largest changes in RPE morphology were evident between the young and aged groups, while the middle-aged group exhibited smaller but notable region-specific differences. We observed a 1.9-fold increase in cytoplasmic alpha-catenin expression specifically in the central-medial region of the eye between the young and aged group. There was an 8-fold increase in subretinal, IBA-1-positive immune cell recruitment and a significant decrease in visual function in aged mice compared to young mice. Functional defects in the RPE corroborated by changes in RPE phagocytotic capacity. Conclusions The marked increase of cytoplasmic alpha-catenin expression and subretinal immune cell deposition, and decreased visual output coincide with regional changes in RPE cell morphometrics when stratified by age. These cumulative changes in the RPE morphology showed predictive regional patterns of stress associated with loss of RPE integrity.
Collapse
Affiliation(s)
- Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Isabelle Gefke
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Vivian Summers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Yong-Kyu Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Hanyi Yu
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Computer Science, Emory University, Atlanta, Georgia, United States
| | - Yana Getz
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Salma Ferdous
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Kevin Donaldson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Kristie Liao
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jack T. Papania
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
14
|
Sbornova I, van der Sande E, Milosavljevic S, Amurrio E, Burbano SD, Das PK, Do HH, Fisher JL, Kargbo P, Patel J, Porcher L, De Zeeuw CI, Meester-Smoor MA, Winkelman BHJ, Klaver CCW, Pocivavsek A, Kelly MP. The Sleep Quality- and Myopia-Linked PDE11A-Y727C Variant Impacts Neural Physiology by Reducing Catalytic Activity and Altering Subcellular Compartmentalization of the Enzyme. Cells 2023; 12:2839. [PMID: 38132157 PMCID: PMC10742168 DOI: 10.3390/cells12242839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Recently, a Y727C variant in the dual-specific 3',5'-cyclic nucleotide phosphodiesterase 11A (PDE11A-Y727C) was linked to increased sleep quality and reduced myopia risk in humans. Given the well-established role that the PDE11 substrates cAMP and cGMP play in eye physiology and sleep, we determined if (1) PDE11A protein is expressed in the retina or other eye segments in mice, (2) PDE11A-Y7272C affects catalytic activity and/or subcellular compartmentalization more so than the nearby suicide-associated PDE11A-M878V variant, and (3) Pde11a deletion alters eye growth or sleep quality in male and female mice. Western blots show distinct protein expression of PDE11A4, but not PDE11A1-3, in eyes of Pde11a WT, but not KO mice, that vary by eye segment and age. In HT22 and COS-1 cells, PDE11A4-Y727C reduces PDE11A4 catalytic activity far more than PDE11A4-M878V, with both variants reducing PDE11A4-cAMP more so than PDE11A4-cGMP activity. Despite this, Pde11a deletion does not alter age-related changes in retinal or lens thickness or axial length, nor vitreous or anterior chamber depth. Further, Pde11a deletion only minimally changes refractive error and sleep quality. That said, both variants also dramatically alter the subcellular compartmentalization of human and mouse PDE11A4, an effect occurring independently of dephosphorylating PDE11A4-S117/S124 or phosphorylating PDE11A4-S162. Rather, re-compartmentalization of PDE11A4-Y727C is due to the loss of the tyrosine changing how PDE11A4 is packaged/repackaged via the trans-Golgi network. Therefore, the protective impact of the Y727C variant may reflect a gain-of-function (e.g., PDE11A4 displacing another PDE) that warrants further investigation in the context of reversing/preventing sleep disturbances or myopia.
Collapse
Affiliation(s)
- Irina Sbornova
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Emilie van der Sande
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, 1105 AZ Amsterdam, The Netherlands
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd., Columbia, SC 29209, USA
| | - Elvis Amurrio
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Steven D. Burbano
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Prosun K. Das
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Helen H. Do
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Janet L. Fisher
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd., Columbia, SC 29209, USA
| | - Porschderek Kargbo
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Janvi Patel
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Latarsha Porcher
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Chris I. De Zeeuw
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, 1105 AZ Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
| | - Magda A. Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
| | - Beerend H. J. Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, 1105 AZ Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Mittlere Strasse 91, 4070 Basel, Switzerland
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd., Columbia, SC 29209, USA
| | - Michy P. Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
- Center for Research on Aging, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA
| |
Collapse
|
15
|
Chauhan P, Kho AM, Srinivasan VJ. From Soma to Synapse: Imaging Age-Related Rod Photoreceptor Changes in the Mouse with Visible Light OCT. OPHTHALMOLOGY SCIENCE 2023; 3:100321. [PMID: 37388138 PMCID: PMC10302163 DOI: 10.1016/j.xops.2023.100321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/05/2023] [Accepted: 04/24/2023] [Indexed: 07/01/2023]
Abstract
Purpose Although the outer nuclear layer (ONL) and outer plexiform layer (OPL) each exhibit a complex internal organization, near-infrared OCT depicts both as monolithic bands. Here, using visible light OCT in the C57BL/6J mouse retina, sublaminar age-related changes in photoreceptor features were imaged and interpreted. These features were (1) oscillations in reflectivity, or striations, in the ONL and (2) a moderately reflective subband in the OPL. Design Cross-sectional study. Participants Pigmented mice (C57BL/6J, n = 14). Methods A 1.0-μm axial resolution visible light spectral/Fourier domain OCT system was used for in vivo retinal imaging. Light and electron microscopy were performed ex vivo. Linear mixed effects models or regression were employed for statistical analysis. Main Outcome Measures Comparison of OCT subbands with corresponding histological features, as well as quantification of subband thickness and reflectivity. Results Corresponding histological comparisons confirm that striations in the ONL arise from the rowlike arrangement of photoreceptor nuclei and reveal that the moderately reflective OPL subband arises from rod spherules. Compression of outer ONL striations with age suggests changes in soma organization. Thinning of the moderately reflective OPL subband with age supports a reduction of synapses in the OPL. Critically, the ONL somas are tightly correlated with the purported spherule layer but not with the rest of the OPL. Conclusions Visible light OCT imaging of the mouse OPL resolves postsynaptic and synaptic differences. Visible light OCT can study rod photoreceptor changes from the soma to the synapse in the living mouse retina. Financial Disclosures Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Pooja Chauhan
- Department of Radiology, NYU Langone Health, New York, New York
| | - Aaron M. Kho
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Vivek J. Srinivasan
- Department of Radiology, NYU Langone Health, New York, New York
- Department of Biomedical Engineering, University of California Davis, Davis, California
- Department of Ophthalmology, NYU Langone Health, New York, New York
| |
Collapse
|
16
|
Sato K, Saigusa D, Kokubun T, Fujioka A, Feng Q, Saito R, Uruno A, Matsukawa N, Ohno-Oishi M, Kunikata H, Yokoyama Y, Yasuda M, Himori N, Omodaka K, Tsuda S, Maekawa S, Yamamoto M, Nakazawa T. Reduced glutathione level in the aqueous humor of patients with primary open-angle glaucoma and normal-tension glaucoma. NPJ AGING 2023; 9:28. [PMID: 37990002 PMCID: PMC10663551 DOI: 10.1038/s41514-023-00124-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/22/2023] [Indexed: 11/23/2023]
Abstract
Glaucoma is a leading cause of blindness worldwide in older people. Profiling the aqueous humor, including the metabolites it contains, is useful to understand physiological and pathological conditions in the eye. In the current study, we used mass spectrometry (MS) to characterize the aqueous humor metabolomic profile and biological features of patients with glaucoma. Aqueous humor samples were collected during trabeculectomy surgery or cataract surgery and analyzed with global metabolomics. We included 40 patients with glaucoma (32 with POAG, 8 with NTG) and 37 control subjects in a discovery study. VIP analysis revealed five metabolites that were elevated and three metabolites that were reduced in the glaucoma patients. The identified metabolomic profile had an area under the receiver operating characteristic curve of 0.953. Among eight selected metabolites, the glutathione level was significantly decreased in association with visual field defects. Moreover, in a validation study to confirm the reproducibility of our findings, the glutathione level was reduced in NTG and POAG patients compared with a cataract control group. Our findings demonstrate that aqueous humor profiling can help to diagnose glaucoma and that various aqueous humor metabolites are correlated with clinical parameters in glaucoma patients. In addition, glutathione is clearly reduced in the aqueous humor of glaucoma patients with both IOP-dependent and IOP-independent disease subtypes. These findings indicate that antioxidant agents in the aqueous humor reflect glaucomatous optic nerve damage and that excessive oxidative stress may be involved in the pathogenesis of glaucoma.
Collapse
Affiliation(s)
- Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Taiki Kokubun
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Amane Fujioka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Qiwei Feng
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ritsumi Saito
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Naomi Matsukawa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Michiko Ohno-Oishi
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroshi Kunikata
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yu Yokoyama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masayuki Yasuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Noriko Himori
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Aging Vision Healthcare, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Satoru Tsuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shigeto Maekawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
- Department of Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
| |
Collapse
|
17
|
Sbornova I, van der Sande E, Milosavljevic S, Amurrio E, Burbano SD, Das P, Do H, Fisher JL, Kargbo P, Patel J, Porcher L, De Zeeuw CI, Meester-Smoor MA, Winkelman BH, Klaver CC, Pocivavsek A, Kelly MP. The sleep quality- and myopia-linked PDE11A-Y727C variant impacts neural physiology by reducing catalytic activity and altering subcellular compartmentalization of the enzyme. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567422. [PMID: 38014312 PMCID: PMC10680747 DOI: 10.1101/2023.11.16.567422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Recently, a Y727C variant in the dual-specific 3',5'-cyclic nucleotide phosphodiesterase 11A (PDE11A-Y727C) was linked to increased sleep quality and reduced myopia risk in humans. Given the well-established role that the PDE11 substrates cAMP and cGMP play in eye physiology and sleep, we determined if 1) PDE11A protein is expressed in the retina or other eye segments in mouse, 2) PDE11A-Y7272C affects catalytic activity and/or subcellular compartmentalization more so than the nearby suicide-associated PDE11A-M878V variant, and 3) Pde11a deletion alters eye growth or sleep quality in male and female mice. Western blots show distinct protein expression of PDE11A4, but not PDE11A1-3, in eyes of Pde11a WT-but not KO mice-that vary by eye segment and age. In HT22 and COS-1 cells, PDE11A4-Y727C reduces PDE11A4 catalytic activity far more than PDE11A4-M878V, with both variants reducing PDE11A4-cAMP more so than PDE11A4-cGMP activity. Despite this, Pde11a deletion does not alter age-related changes in retinal or lens thickness, axial length, nor vitreous or anterior chamber depth. Further, Pde11a deletion only minimally changes refractive error and sleep quality. That said, both variants also dramatically alter the subcellular compartmentalization of human and mouse PDE11A4, an effect occurring independently of dephosphorylating PDE11A4-S117/S124 or phosphorylating PDE11A4-S162. Rather, re-compartmentalization of PDE11A4-Y727C is due to the loss of the tyrosine changing how PDE11A4 is packaged/repackaged via the trans-Golgi network. Therefore, the protective impact of the Y727C variant may reflect a gain-of-function (e.g., PDE11A4 displacing another PDE) that warrants further investigation in the context of reversing/preventing sleep disturbances or myopia.
Collapse
Affiliation(s)
- Irina Sbornova
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Emilie van der Sande
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, Amsterdam, The Netherlands
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd, Columbia, SC
| | - Elvis Amurrio
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Steven D. Burbano
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Prosun Das
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Helen Do
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Janet L. Fisher
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd, Columbia, SC
| | - Porschderek Kargbo
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Janvi Patel
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Latarsha Porcher
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Chris I. De Zeeuw
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
| | - Beerend H.J. Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
| | - Caroline C.W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Mittlere Strasse 91, Basel, Switzerland
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd, Columbia, SC
| | - Michy P. Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
- Center for Research on Aging, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| |
Collapse
|
18
|
Zhu S, Xu R, Engel AL, Wang Y, McNeel R, Hurley JB, Chao JR, Du J. Proline provides a nitrogen source in the retinal pigment epithelium to synthesize and export amino acids for the neural retina. J Biol Chem 2023; 299:105275. [PMID: 37741457 PMCID: PMC10616405 DOI: 10.1016/j.jbc.2023.105275] [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: 04/19/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
It is known that metabolic defects in the retinal pigment epithelium (RPE) can cause degeneration of its neighboring photoreceptors in the retina, leading to retinal degenerative diseases such as age-related macular degeneration. However, how RPE metabolism supports the health of the neural retina remains unclear. The retina requires exogenous nitrogen sources for protein synthesis, neurotransmission, and energy metabolism. Using 15N tracing coupled with mass spectrometry, we found human RPE can utilize the nitrogen in proline to produce and export 13 amino acids, including glutamate, aspartate, glutamine, alanine, and serine. Similarly, we found this proline nitrogen utilization in the mouse RPE/choroid but not in the neural retina of explant cultures. Coculture of human RPE with the retina showed that the retina can take up the amino acids, especially glutamate, aspartate, and glutamine, generated from proline nitrogen in the RPE. Intravenous delivery of 15N proline in vivo demonstrated 15N-derived amino acids appear earlier in the RPE before the retina. We also found proline dehydrogenase, the key enzyme in proline catabolism is highly enriched in the RPE but not the retina. The deletion of proline dehydrogenase blocks proline nitrogen utilization in RPE and the import of proline nitrogen-derived amino acids in the retina. Our findings highlight the importance of RPE metabolism in supporting nitrogen sources for the retina, providing insight into understanding the mechanisms of the retinal metabolic ecosystem and RPE-initiated retinal degenerative diseases.
Collapse
Affiliation(s)
- Siyan Zhu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA; Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, USA; Department of Pharmaceutical and Pharmacological Sciences, West Virginia University, Morgantown, West Virginia, USA
| | - Rong Xu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA; Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Abbi L Engel
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA; Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Rachel McNeel
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA; Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - James B Hurley
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA; Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Jennifer R Chao
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA; Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, USA.
| |
Collapse
|
19
|
Bejarano E, Whitcomb EA, Pfeiffer RL, Rose KL, Asensio MJ, Rodríguez-Navarro JA, Ponce-Mora A, Canto A, Almansa I, Schey KL, Jones BW, Taylor A, Rowan S. Unbalanced redox status network as an early pathological event in congenital cataracts. Redox Biol 2023; 66:102869. [PMID: 37677999 PMCID: PMC10495660 DOI: 10.1016/j.redox.2023.102869] [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: 06/21/2023] [Revised: 08/08/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
The lens proteome undergoes dramatic composition changes during development and maturation. A defective developmental process leads to congenital cataracts that account for about 30% of cases of childhood blindness. Gene mutations are associated with approximately 50% of early-onset forms of lens opacity, with the remainder being of unknown etiology. To gain a better understanding of cataractogenesis, we utilized a transgenic mouse model expressing a mutant ubiquitin protein in the lens (K6W-Ub) that recapitulates most of the early pathological changes seen in human congenital cataracts. We performed mass spectrometry-based tandem-mass-tag quantitative proteomics in E15, P1, and P30 control or K6W-Ub lenses. Our analysis identified targets that are required for early normal differentiation steps and altered in cataractous lenses, particularly metabolic pathways involving glutathione and amino acids. Computational molecular phenotyping revealed that glutathione and taurine were spatially altered in the K6W-Ub cataractous lens. High-performance liquid chromatography revealed that both taurine and the ratio of reduced glutathione to oxidized glutathione, two indicators of redox status, were differentially compromised in lens biology. In sum, our research documents that dynamic proteome changes in a mouse model of congenital cataracts impact redox biology in lens. Our findings shed light on the molecular mechanisms associated with congenital cataracts and point out that unbalanced redox status due to reduced levels of taurine and glutathione, metabolites already linked to age-related cataract, could be a major underlying mechanism behind lens opacities that appear early in life.
Collapse
Affiliation(s)
- Eloy Bejarano
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Elizabeth A Whitcomb
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Rebecca L Pfeiffer
- Moran Eye Center, The University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Maria José Asensio
- Servicio de Neurobiología, Departamento de Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| | - José Antonio Rodríguez-Navarro
- Servicio de Neurobiología, Departamento de Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain; Department of Cell Biology, Complutense University of Madrid, Madrid, Spain
| | - Alejandro Ponce-Mora
- School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Antolín Canto
- School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Inma Almansa
- School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Kevin L Schey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Bryan W Jones
- Moran Eye Center, The University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Allen Taylor
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; Friedman School of Nutrition and Science Policy, Tufts University, Boston, MA, USA.
| | - Sheldon Rowan
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; Friedman School of Nutrition and Science Policy, Tufts University, Boston, MA, USA.
| |
Collapse
|
20
|
Liu J, Si H, Huang D, Lu D, Zou S, Qi D, Pei X, Huang S, Li Z. Mechanisms of Extraorbital Lacrimal Gland Aging in Mice: An Integrative Analysis of the Temporal Transcriptome. Invest Ophthalmol Vis Sci 2023; 64:18. [PMID: 37695604 PMCID: PMC10501490 DOI: 10.1167/iovs.64.12.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 08/16/2023] [Indexed: 09/12/2023] Open
Abstract
Purpose This study used high-throughput RNA sequencing (RNA-Seq) and bioinformatics analysis to investigate the altered transcriptome profile of aging lacrimal glands in mice that occurs over the course of a 24-hour cycle. Methods Male C57BL/6J mice aged 12 weeks (young) and 20 months (aging) were housed in a pathogen-free setting with a 12-hour light/12-hour dark cycle. Throughout a 24-hour cycle, mouse extraorbital lacrimal glands (ELGs) were collected at eight time points at three-hour intervals. To prepare for the high-throughput RNA-Seq, whole mRNA was extracted. Differentially expressed genes (DEGs) in the young and aging groups were subjected to bioinformatic analysis based on diurnal patterns. Furthermore, the cell populations in which significant DEGs express and signaling pathways occur were validated at the single-cell RNA sequencing (scRNA-seq) level. Results The total transcriptome composition was significantly altered in aging ELGs compared with that in young mouse ELGs at eight time points during the 24-hour cycle, with 864 upregulated and 228 downregulated DEGs, which were primarily enriched in inflammatory pathways. Further comparative analysis of the point-to-point transcriptome revealed that aging ELGs underwent alterations in the temporal transcriptome profile in several pathways, including the inflammation-related, metabolism-related, mitochondrial bioenergetic function-associated, synaptome neural activity-associated, cell processes-associated, DNA processing-associated and fibrosis-associated pathways. Most of these pathways occurred separately in distinct cell populations. Conclusions Transcriptome profiles of aging lacrimal glands undergo considerable diurnal time-dependent changes; this finding offers a comprehensive source of information to better understand the pathophysiology of lacrimal gland aging and its underlying mechanisms.
Collapse
Affiliation(s)
- Jiangman Liu
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Sen Zou
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
21
|
Fang W, Chen S, Jin X, Liu S, Cao X, Liu B. Metabolomics in aging research: aging markers from organs. Front Cell Dev Biol 2023; 11:1198794. [PMID: 37397261 PMCID: PMC10313136 DOI: 10.3389/fcell.2023.1198794] [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: 04/02/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Metabolism plays an important role in regulating aging at several levels, and metabolic reprogramming is the main driving force of aging. Due to the different metabolic needs of different tissues, the change trend of metabolites during aging in different organs and the influence of different levels of metabolites on organ function are also different, which makes the relationship between the change of metabolite level and aging more complex. However, not all of these changes lead to aging. The development of metabonomics research has opened a door for people to understand the overall changes in the metabolic level in the aging process of organisms. The omics-based "aging clock" of organisms has been established at the level of gene, protein and epigenetic modifications, but there is still no systematic summary at the level of metabolism. Here, we reviewed the relevant research published in the last decade on aging and organ metabolomic changes, discussed several metabolites with high repetition rate, and explained their role in vivo, hoping to find a group of metabolites that can be used as metabolic markers of aging. This information should provide valuable information for future diagnosis or clinical intervention of aging and age-related diseases.
Collapse
Affiliation(s)
- Weicheng Fang
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Shuxin Chen
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Xuejiao Jin
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Shenkui Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Xiuling Cao
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Beidong Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Zhu S, Xu R, Engel AL, Wang Y, McNeel R, Hurley JB, Chao JR, Du J. Proline provides a nitrogen source in the retinal pigment epithelium to synthesize and export amino acids for the neural retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537355. [PMID: 37131780 PMCID: PMC10153141 DOI: 10.1101/2023.04.18.537355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
It is known that metabolic defects in the retinal pigment epithelium (RPE) can cause degeneration of its neighboring photoreceptors in the retina, leading to retinal degenerative diseases such as age-related macular degeneration. However, how RPE metabolism supports the health of the neural retina remains unclear. The retina requires exogenous nitrogen sources for protein synthesis, neurotransmission, and energy metabolism. Using 15N tracing coupled with mass spectrometry, we found human RPE can utilize the nitrogen in proline to produce and export 13 amino acids, including glutamate, aspartate, glutamine, alanine and serine. Similarly, we found this proline nitrogen utilization in the mouse RPE/choroid but not in the neural retina of explant cultures. Co-culture of human RPE with the retina showed that the retina can take up the amino acids, especially glutamate, aspartate and glutamine, generated from proline nitrogen in the RPE. Intravenous delivery of 15N proline in vivo demonstrated 15N-derived amino acids appear earlier in the RPE before the retina. We also found proline dehydrogenase (PRODH), the key enzyme in proline catabolism is highly enriched in the RPE but not the retina. The deletion of PRODH blocks proline nitrogen utilization in RPE and the import of proline nitrogen-derived amino acids in the retina. Our findings highlight the importance of RPE metabolism in supporting nitrogen sources for the retina, providing insight into understanding the mechanisms of the retinal metabolic ecosystem and RPE-initiated retinal degenerative diseases.
Collapse
Affiliation(s)
- Siyan Zhu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
- Department of Pharmaceutical and Pharmacological Sciences, West Virginia University, Morgantown, WV 26506
| | - Rong Xu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
| | - Abbi L. Engel
- Department of Ophthalmology, University of Washington, Seattle, WA 98109
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
| | - Rachel McNeel
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
| | - James B. Hurley
- Department of Ophthalmology, University of Washington, Seattle, WA 98109
- Department of Biochemistry, University of Washington, Seattle, WA 98109
| | - Jennifer R. Chao
- Department of Ophthalmology, University of Washington, Seattle, WA 98109
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
| |
Collapse
|
23
|
Shan H, Liu W, Li Y, Pang K. The Autoimmune Rheumatic Disease Related Dry Eye and Its Association with Retinopathy. Biomolecules 2023; 13:724. [PMID: 37238594 PMCID: PMC10216215 DOI: 10.3390/biom13050724] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Dry eye disease is a chronic disease of the ocular surface characterized by abnormal tear film composition, tear film instability, and ocular surface inflammation, affecting 5% to 50% of the population worldwide. Autoimmune rheumatic diseases (ARDs) are systemic disorders with multi-organ involvement, including the eye, and play a significant role in dry eye. To date, most studies have focused on Sjögren's syndrome (one of the ARDs) since it manifests as two of the most common symptoms-dry eyes and a dry mouth-and attracts physicians to explore the relationship between dry eye and ARDs. Many patients complained of dry eye related symptoms before they were diagnosed with ARDs, and ocular surface malaise is a sensitive indicator of the severity of ARDs. In addition, ARD related dry eye is also associated with some retinal diseases directly or indirectly, which are described in this review. This review also summarizes the incidence, epidemiological characteristics, pathogenesis, and accompanying ocular lesions of ARD's related dry eye, emphasizing the potential role of dry eye in recognition and monitoring among ARDs patients.
Collapse
Affiliation(s)
| | | | | | - Kunpeng Pang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
24
|
Duan H, Song W, Guo J, Yan W. Taurine: A Source and Application for the Relief of Visual Fatigue. Nutrients 2023; 15:nu15081843. [PMID: 37111062 PMCID: PMC10142897 DOI: 10.3390/nu15081843] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/03/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
According to reports, supplementation with appropriate doses of taurine may help to reduce visual fatigue. Presently, some progress has been made in research related to taurine in eye health, but the lack of systematic summaries has led to the neglect of its application in the relief of visual fatigue. This paper, therefore, provides a systematic review of the sources of taurine, including the endogenous metabolic and exogenous dietary pathways, as well as a detailed review of the distribution and production of exogenous taurine. The physiological mechanisms underlying the production of visual fatigue are summarized and the research progress of taurine in relieving visual fatigue is reviewed, including the safety of consumption and the mechanism of action in relieving visual fatigue, in order to provide some reference basis and inspiration for the development and application of taurine in functional foods for relieving visual fatigue.
Collapse
Affiliation(s)
- Hao Duan
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China
| | - Wei Song
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China
| | - Jinhong Guo
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China
| | - Wenjie Yan
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China
| |
Collapse
|
25
|
Dhingra A, Tobias JW, Philp NJ, Boesze-Battaglia K. Transcriptomic Changes Predict Metabolic Alterations in LC3 Associated Phagocytosis in Aged Mice. Int J Mol Sci 2023; 24:6716. [PMID: 37047689 PMCID: PMC10095460 DOI: 10.3390/ijms24076716] [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: 02/24/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
LC3b (Map1lc3b) plays an essential role in canonical autophagy and is one of several components of the autophagy machinery that mediates non-canonical autophagic functions. Phagosomes are often associated with lipidated LC3b to promote phagosome maturation in a process called LC3-associated phagocytosis (LAP). Specialized phagocytes, such as mammary epithelial cells, retinal pigment epithelial (RPE) cells, and sertoli cells, utilize LAP for optimal degradation of phagocytosed material, including debris. In the visual system, LAP is critical to maintain retinal function, lipid homeostasis, and neuroprotection. In a mouse model of retinal lipid steatosis-mice lacking LC3b (LC3b-/-), we observed increased lipid deposition, metabolic dysregulation, and enhanced inflammation. Herein, we present a non-biased approach to determine if loss of LAP mediated processes modulate the expression of various genes related to metabolic homeostasis, lipid handling, and inflammation. A comparison of the RPE transcriptome of WT and LC3b-/- mice revealed 1533 DEGs, with ~73% upregulated and 27% downregulated. Enriched gene ontology (GO) terms included inflammatory response (upregulated DEGs), fatty acid metabolism, and vascular transport (downregulated DEGs). Gene set enrichment analysis (GSEA) identified 34 pathways; 28 were upregulated (dominated by inflammation/related pathways) and 6 were downregulated (dominated by metabolic pathways). Analysis of additional gene families identified significant differences for genes in the solute carrier family, RPE signature genes, and genes with a potential role in age-related macular degeneration. These data indicate that loss of LC3b induces robust changes in the RPE transcriptome contributing to lipid dysregulation and metabolic imbalance, RPE atrophy, inflammation, and disease pathophysiology.
Collapse
Affiliation(s)
- Anuradha Dhingra
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John W. Tobias
- Penn Genomics and Sequencing Core, Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy J. Philp
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
26
|
Dhingra A, Tobias JW, Philp NJ, Boesze-Battaglia K. Transcriptomic changes predict metabolic alterations in LC3 associated phagocytosis in aged mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532586. [PMID: 36993501 PMCID: PMC10054970 DOI: 10.1101/2023.03.14.532586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
LC3b ( Map1lc3b ) plays an essential role in canonical autophagy and is one of several components of the autophagy machinery that mediates non-canonical autophagic functions. Phagosomes are often associated with lipidated LC3b, to pro-mote phagosome maturation in a process called LC3-associated phagocytosis (LAP). Specialized phagocytes such as mammary epithelial cells, retinal pigment epithelial (RPE) cells, and sertoli cells utilize LAP for optimal degradation of phagocytosed material, including debris. In the visual system, LAP is critical to maintain retinal function, lipid homeostasis and neuroprotection. In a mouse model of retinal lipid steatosis - mice lacking LC3b ( LC3b -/- ), we observed increased lipid deposition, metabolic dysregulation and enhanced inflammation. Herein we present a non-biased approach to determine if loss of LAP mediated processes modulate the expression of various genes related to metabolic homeostasis, lipid handling, and inflammation. A comparison of the RPE transcriptome of WT and LC3b -/- mice revealed 1533 DEGs, with ~73% upregulated and 27% down-regulated. Enriched gene ontology (GO) terms included inflammatory response (upregulated DEGs), fatty acid metabolism and vascular transport (downregulated DEGs). Gene set enrichment analysis (GSEA) identified 34 pathways; 28 were upregulated (dominated by inflammation/related pathways) and 6 were downregulated (dominated by metabolic pathways). Analysis of additional gene families identified significant differences for genes in the solute carrier family, RPE signature genes, and genes with potential role in age-related macular degeneration. These data indicate that loss of LC3b induces robust changes in the RPE transcriptome contributing to lipid dysregulation and metabolic imbalance, RPE atrophy, inflammation, and disease pathophysiology.
Collapse
|
27
|
Saravanan M, Xu R, Roby O, Wang Y, Zhu S, Lu A, Du J. Tissue-Specific Sex Difference in Mouse Eye and Brain Metabolome Under Fed and Fasted States. Invest Ophthalmol Vis Sci 2023; 64:18. [PMID: 36892534 PMCID: PMC10010444 DOI: 10.1167/iovs.64.3.18] [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: 01/09/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023] Open
Abstract
Purpose Visual physiology and various ocular diseases demonstrate sexual dimorphisms; however, how sex influences metabolism in different eye tissues remains undetermined. This study aims to address common and tissue-specific sex differences in metabolism in the retina, RPE, lens, and brain under fed and fasted conditions. Methods After ad libitum fed or being deprived of food for 18 hours, mouse eye tissues (retina, RPE/choroid, and lens), brain, and plasma were harvested for targeted metabolomics. The data were analyzed with both partial least squares-discriminant analysis and volcano plot analysis. Results Among 133 metabolites that cover major metabolic pathways, we found 9 to 45 metabolites that are sex different in different tissues under the fed state and 6 to 18 metabolites under the fasted state. Among these sex-different metabolites, 33 were changed in 2 or more tissues, and 64 were tissue specific. Pantothenic acid, hypotaurine, and 4-hydroxyproline were the top commonly changed metabolites. The lens and the retina had the most tissue-specific, sex-different metabolites enriched in the metabolism of amino acid, nucleotide, lipids, and tricarboxylic acid cycle. The lens and the brain had more similar sex-different metabolites than other ocular tissues. The female RPE and female brain were more sensitive to fasting with more decreased metabolites in amino acid metabolism, tricarboxylic acid cycles, and glycolysis. The plasma had the fewest sex-different metabolites, with very few overlapping changes with tissues. Conclusions Sex has a strong influence on eye and brain metabolism in tissue-specific and metabolic state-specific manners. Our findings may implicate the sexual dimorphisms in eye physiology and susceptibility to ocular diseases.
Collapse
Affiliation(s)
- Meghashri Saravanan
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Rong Xu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Olivia Roby
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Siyan Zhu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Amy Lu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| |
Collapse
|
28
|
Lindovsky J, Palkova M, Symkina V, Raishbrook MJ, Prochazka J, Sedlacek R. OCT and ERG Techniques in High-Throughput Phenotyping of Mouse Vision. Genes (Basel) 2023; 14:genes14020294. [PMID: 36833221 PMCID: PMC9956909 DOI: 10.3390/genes14020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
The purpose of the study is to demonstrate coherent optical tomography and electroretinography techniques adopted from the human clinical practice to assess the morphology and function of the mouse retina in a high-throughput phenotyping environment. We present the normal range of wild-type C57Bl/6NCrl retinal parameters in six age groups between 10 and 100 weeks as well as examples of mild and severe pathologies resulting from knocking out a single protein-coding gene. We also show example data obtained by more detailed analysis or additional methods useful in eye research, for example, the angiography of a superficial and deep vascular complex. We discuss the feasibility of these techniques in conditions demanding a high-throughput approach such as the systemic phenotyping carried out by the International Mouse Phenotyping Consortium.
Collapse
|
29
|
Yang Y, Wu J, Lu W, Dai Y, Zhang Y, Sun X. Mitochondria-associated endoplasmic reticulum membranes dysfunction contributes to PARP-1-dependent cell death under oxidative stress in retinal precursor cells. J Biochem Mol Toxicol 2023; 37:e23303. [PMID: 36639873 DOI: 10.1002/jbt.23303] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/20/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Persistent poly (ADP-ribose) polymerase 1 (PARP-1) activation has proven detrimental and can lead to PARP-1-dependent cell death. Mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) serve as essential hubs for many biological pathways, such as autophagy and mitochondria fission and fusion. This study aimed to alleviate the effects of hydrogen peroxide (H2 O2 )-induced persistent PARP-1 activation and MAM dysregulation by the usage of a PARP-1 inhibitor. Results showed that receptor-interacting protein kinase (RIPK) 1 inhibitor (necrostatin-1) and PARP-1 inhibitor (olaparib) protected retinal precursor cells from H2 O2 -induced death, while a pan-caspase inhibitor (Z-VAD-FMK) failed to protect R28 cells. Olaparib also alleviated H2 O2 -induced MAM dysregulation, as evidenced by decreased VDAC1/ITPR3 interactions and reduced mitochondrial membrane potential collapse. Additionally, olaparib also inhibited H2 O2 -induced autophagy. Inhibiting autophagic flux increased MAM signaling under both normal and oxidative conditions. Furthermore, H2 O2 treatment caused a reduction in the protein level of mitofusin-2 (MFN2) in a dose- and time-dependent manner. Mfn2 knockdown was found to further magnify MAM dysregulation and mitochondrial dysfunction under normal and oxidative conditions. Mfn2 overexpression surprisingly enhanced H2 O2 -induced MAM signaling and failed to rescue H2 O2 -induced mitochondrial dysfunction. These results indicate that MAMs probably serve as a membrane source for oxidative stress-associated autophagy. MAM dysregulation also contributed to H2 O2 -induced PARP-1-dependent cell death. However, more studies are required to decipher the link between the modulation of Mfn2 expression, changes in MAM integrity, and alterations in mitochondrial performances.
Collapse
Affiliation(s)
- Yuting Yang
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jihong Wu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, China
| | - Wei Lu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiqin Dai
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Youjia Zhang
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Xu R, Wang Y, Du J, Salido EM. Retinal Metabolic Profile on IMPG2 Deficiency Mice with Subretinal Lesions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:457-463. [PMID: 37440072 DOI: 10.1007/978-3-031-27681-1_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The interphotoreceptor matrix (IPM) is the extracellular matrix between the photoreceptors and the retinal pigment epithelium (RPE). The IPM has two proteoglycans: the IPM proteoglycans 1 and 2 (IMPG1 and IMPG2, respectively). Patients with mutations on IMPG2 develop subretinal vitelliform lesions that affect vision. We previously created an IMPG2 knockout (KO) mice model that generates subretinal lesions similar to those found in humans. These subretinal lesions in IMPG2 KO mice retinas are, in part, composed of mislocalized IMPG1. In addition, IMPG2 KO mice show microscopic IMPG1 material accumulation between the RPE and the photoreceptor outer segments. In this work we discuss the possibility that material accumulation on IMPG2 KO mice retinas affects photoreceptor metabolism. To further investigate this idea, we used targeted metabolomics to profile retinal metabolome on IMPG2 KO mice. The metabolite set enrichment analysis showed reduced glutamate metabolism, urea cycle, and galactose metabolism suggesting affected energy metabolism in mice retinas of IMPG2 KO mice with subretinal lesion.
Collapse
Affiliation(s)
- Rong Xu
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Yekai Wang
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Jianhai Du
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Ezequiel M Salido
- Departments of Biochemistry and molecular medicine, and Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
31
|
Lampignano L, Niro A, Castellana F, Bortone I, Zupo R, Tirelli S, Tatoli R, Griseta C, De Nucci S, Sila A, De Pergola G, Conte C, Alessio G, Boscia F, Sborgia G, Giannelli G, Sardone R. Liver fibrosis and retinal features in an older Mediterranean population: Results from the Salus in Apulia study. Front Neurosci 2022; 16:1048375. [PMID: 36590297 PMCID: PMC9798127 DOI: 10.3389/fnins.2022.1048375] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
Background Age is a leading contributor to the liver fibrosis rate and a gradual deterioration of optical function, but this association in older populations is still under-explored. The present study aimed to explore the link between vascular and neural retinal characteristics and the risk of liver fibrosis in 731 older adults from the population-based Salus in Apulia study. Methods Retinal features were obtained using optical coherence tomography (OCT) and OCT-angiography (OCT-A). Liver fibrosis risk was taken as the fibrosis-4 (FIB-4) score. Generalized linear models (logistic regression) were used to estimate the association effect between each unit increase of OCT and OCT-A parameters as independent variables and a FIB-4 ≥ 2.67 score as an outcome. Generalized additive models were used to assess the non-linear association between OCT-A features and the linear FIB-4 score. Results Increased gangliar cell complex (GCC) thickness was inversely associated with a FIB-4 score above the cut-off in both the raw model (OR: 0.98; 95% CI: 0.96-0.99; SE: 0.01) and after adjustment for age, sex, education, hypertension, diabetes, total cholesterol, and triglycerides (OR: 0.98; 95% CI: 0.97-0.99; SE: 0.01). Conclusion Our findings add to the growing volume of scientific literature demonstrating that liver fibrosis is associated with retinal neurodegeneration. This study raises a number of new questions, including whether OCT-A may be used to track the progression of metabolic abnormalities and define exact thresholds for predicting and classifying liver disease.
Collapse
Affiliation(s)
- Luisa Lampignano
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Alfredo Niro
- Eye Clinic, Hospital “SS. Annunziata”, Azienda Sanitaria Locale (ASL) Taranto, Taranto, Italy
| | - Fabio Castellana
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Ilaria Bortone
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Roberta Zupo
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Sarah Tirelli
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Rossella Tatoli
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Chiara Griseta
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Sara De Nucci
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Annamaria Sila
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Giovanni De Pergola
- Unit of Geriatrics and Internal Medicine, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, Bari, Italy
| | - Caterina Conte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy,Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan, Italy
| | - Giovanni Alessio
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Boscia
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Giancarlo Sborgia
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | | | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Rodolfo Sardone
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study”, National Institute of Gastroenterology Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “Saverio de Bellis”, Research Hospital, Castellana Grotte, Italy,*Correspondence: Rodolfo Sardone, ; orcid.org/0000-0003-1383-1850
| |
Collapse
|
32
|
Innate immunity dysregulation in aging eye and therapeutic interventions. Ageing Res Rev 2022; 82:101768. [PMID: 36280210 DOI: 10.1016/j.arr.2022.101768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/29/2022] [Accepted: 10/20/2022] [Indexed: 01/31/2023]
Abstract
The prevalence of eye diseases increases considerably with age, resulting in significant vision impairment. Although the pathobiology of age-related eye diseases has been studied extensively, the contribution of immune-related changes due to aging remains elusive. In the eye, tissue-resident cells and infiltrating immune cells regulate innate responses during injury or infection. But due to aging, these cells lose their protective functions and acquire pathological phenotypes. Thus, dysregulated ocular innate immunity in the elderly increases the susceptibility and severity of eye diseases. Herein, we emphasize the impact of aging on the ocular innate immune system in the pathogenesis of infectious and non-infectious eye diseases. We discuss the role of age-related alterations in cellular metabolism, epigenetics, and cellular senescence as mechanisms underlying altered innate immune functions. Finally, we describe approaches to restore protective innate immune functions in the aging eye. Overall, the review summarizes our current understanding of innate immune functions in eye diseases and their dysregulation during aging.
Collapse
|
33
|
A critical bioenergetic switch is regulated by IGF2 during murine cartilage development. Commun Biol 2022; 5:1230. [PMID: 36369360 PMCID: PMC9652369 DOI: 10.1038/s42003-022-04156-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
Long bone growth requires the precise control of chondrocyte maturation from proliferation to hypertrophy during endochondral ossification, but the bioenergetic program that ensures normal cartilage development is still largely elusive. We show that chondrocytes have unique glucose metabolism signatures in these stages, and they undergo bioenergetic reprogramming from glycolysis to oxidative phosphorylation during maturation, accompanied by an upregulation of the pentose phosphate pathway. Inhibition of either oxidative phosphorylation or the pentose phosphate pathway in murine chondrocytes and bone organ cultures impaired hypertrophic differentiation, suggesting that the appropriate balance of these pathways is required for cartilage development. Insulin-like growth factor 2 (IGF2) deficiency resulted in a profound increase in oxidative phosphorylation in hypertrophic chondrocytes, suggesting that IGF2 is required to prevent overactive glucose metabolism and maintain a proper balance of metabolic pathways. Our results thus provide critical evidence of preference for a bioenergetic pathway in different stages of chondrocytes and highlight its importance as a fundamental mechanism in skeletal development.
Collapse
|
34
|
Wang Y, Zhao J, Chen S, Li D, Yang J, Zhao X, Qin M, Guo M, Chen C, He Z, Zhou Y, Xu L. Let-7 as a Promising Target in Aging and Aging-Related Diseases: A Promise or a Pledge. Biomolecules 2022; 12:1070. [PMID: 36008964 PMCID: PMC9406090 DOI: 10.3390/biom12081070] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/10/2022] Open
Abstract
The abnormal regulation and expression of microRNA (miRNA) are closely related to the aging process and the occurrence and development of aging-related diseases. Lethal-7 (let-7) was discovered in Caenorhabditis elegans (C. elegans) and plays an important role in development by regulating cell fate regulators. Accumulating evidence has shown that let-7 is elevated in aging tissues and participates in multiple pathways that regulate the aging process, including affecting tissue stem cell function, body metabolism, and various aging-related diseases (ARDs). Moreover, recent studies have found that let-7 plays an important role in the senescence of B cells, suggesting that let-7 may also participate in the aging process by regulating immune function. Therefore, these studies show the diversity and complexity of let-7 expression and regulatory functions during aging. In this review, we provide a detailed overview of let-7 expression regulation as well as its role in different tissue aging and aging-related diseases, which may provide new ideas for enriching the complex expression regulation mechanism and pathobiological function of let-7 in aging and related diseases and ultimately provide help for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Ya Wang
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Shipeng Chen
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Dongmei Li
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Jing Yang
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Xu Zhao
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Ming Qin
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Zhixu He
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi 563000, China;
| | - Ya Zhou
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Medical Physics, Zunyi Medical University, Zunyi 563000, China
| | - Lin Xu
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China; (Y.W.); (J.Z.); (S.C.); (D.L.); (J.Y.); (X.Z.); (M.Q.); (M.G.); (C.C.)
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
35
|
Zhu Y, Zhang Y, Qi X, Lian Y, Che H, Jia J, Yang C, Xu Y, Chi X, Jiang W, Li Y, Mi J, Yang Y, Li X, Tian G. GAD1 alleviates injury-induced optic neurodegeneration by inhibiting retinal ganglion cell apoptosis. Exp Eye Res 2022; 223:109201. [DOI: 10.1016/j.exer.2022.109201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/21/2022] [Accepted: 07/18/2022] [Indexed: 11/27/2022]
|
36
|
Tong Y, Zhang Z, Wang S. Role of Mitochondria in Retinal Pigment Epithelial Aging and Degeneration. FRONTIERS IN AGING 2022; 3:926627. [PMID: 35912040 PMCID: PMC9337215 DOI: 10.3389/fragi.2022.926627] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/21/2022] [Indexed: 12/17/2022]
Abstract
Retinal pigment epithelial (RPE) cells form a monolayer between the neuroretina and choroid. It has multiple important functions, including acting as outer blood-retina barrier, maintaining the function of neuroretina and photoreceptors, participating in the visual cycle and regulating retinal immune response. Due to high oxidative stress environment, RPE cells are vulnerable to dysfunction, cellular senescence, and cell death, which underlies RPE aging and age-related diseases, including age-related macular degeneration (AMD). Mitochondria are the powerhouse of cells and a major source of cellular reactive oxygen species (ROS) that contribute to mitochondrial DNA damage, cell death, senescence, and age-related diseases. Mitochondria also undergo dynamic changes including fission/fusion, biogenesis and mitophagy for quality control in response to stresses. The role of mitochondria, especially mitochondrial dynamics, in RPE aging and age-related diseases, is still unclear. In this review, we summarize the current understanding of mitochondrial function, biogenesis and especially dynamics such as morphological changes and mitophagy in RPE aging and age-related RPE diseases, as well as in the biological processes of RPE cellular senescence and cell death. We also discuss the current preclinical and clinical research efforts to prevent or treat RPE degeneration by restoring mitochondrial function and dynamics.
Collapse
Affiliation(s)
- Yao Tong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Zunyi Zhang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
- Department of Ophthalmology, Tulane University, New Orleans, LA, United States
- Tulane Personalized Health Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
37
|
Kitazawa K, Inotmata T, Shih K, Hughes JWB, Bozza N, Tomioka Y, Numa K, Yokoi N, Campisi J, Dana R, Sotozono C. Impact of aging on the pathophysiology of dry eye disease: A systematic review and meta-analysis. Ocul Surf 2022; 25:108-118. [PMID: 35753664 DOI: 10.1016/j.jtos.2022.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE Dry eye disease (DED) is a common age-related ocular surface disease. However, it is unknown how aging influences the ocular surface microenvironment. This systematic review aims to investigate how the aging process changes the ocular surface microenvironment and impacts the development of DED. METHODS An article search was performed in PubMed, EMBASE, and Web of Science. 44 studies reporting on age-related ocular changes and 14 large epidemiological studies involving the prevalence of DED were identified. 8 out of 14 epidemiological studies were further analyzed with meta-analysis. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) reporting guidelines were followed. Study-specific estimates (impact of aging on the prevalence of DED) were combined using one-group meta-analysis in a random-effects model. RESULTS Meta-analysis revealed the prevalence of DED in the elderly aged 60 years old or older was 5519 of 60107 (9.2%) and the odds ratio of aging compared to younger age was 1.313 (95% confidence interval [CI]; 1.107, 1.557). With increasing age, the integrity of the ocular surface and tear film stability decreased. Various inflammatory cells, including senescent-associated T-cells, infiltrated the ocular surface epithelium, lacrimal gland, and meibomian gland, accompanied by senescence-related changes, including accumulation of 8-OHdG and lipofuscin-like inclusions, increased expression of p53 and apoptosis-related genes, and decreased Ki67 positive cells. CONCLUSIONS The aging process greatly impacts the ocular surface microenvironment, consequently leading to DED.
Collapse
Affiliation(s)
- Koji Kitazawa
- Buck Institute for Research on Aging, Novato, CA, 94945, USA; Kyoto Prefectural University of Medicine, Department of Ophthalmology, Kyoto, Japan.
| | - Takenori Inotmata
- Juntendo University Graduate School of Medicine, Department of Ophthalmology, Tokyo, Japan; Juntendo University Graduate School of Medicine, Department of Hospital Administration, Tokyo, Japan; Juntendo University Graduate School of Medicine, Department of Digital Medicine, Tokyo, Japan
| | - Kendric Shih
- Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKUMed), Department of Ophthalmology, Hong Kong, China
| | | | - Niha Bozza
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Yasufumi Tomioka
- Kyoto Prefectural University of Medicine, Department of Ophthalmology, Kyoto, Japan
| | - Kohsaku Numa
- Buck Institute for Research on Aging, Novato, CA, 94945, USA; Kyoto Prefectural University of Medicine, Department of Ophthalmology, Kyoto, Japan
| | - Norihiko Yokoi
- Kyoto Prefectural University of Medicine, Department of Ophthalmology, Kyoto, Japan
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA; Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Reza Dana
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Chie Sotozono
- Kyoto Prefectural University of Medicine, Department of Ophthalmology, Kyoto, Japan
| |
Collapse
|
38
|
Alam NM, Douglas RM, Prusky GT. Treatment of age-related visual impairment with a peptide acting on mitochondria. Dis Model Mech 2022; 15:dmm048256. [PMID: 34766182 PMCID: PMC8891924 DOI: 10.1242/dmm.048256] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 10/27/2021] [Indexed: 12/04/2022] Open
Abstract
Age-related visual decline and disease due to neural dysfunction are major sources of disability that have resisted effective treatment. In light of evidence that visual impairment and mitochondrial dysfunction advance with age, we characterized age-related decline of spatial visual function in mice and investigated whether treatment of aged mice with the mitochondrion-penetrating peptide elamipretide that has been reported to improve mitochondrial function, would improve it. Impaired photopic acuity measured by using a virtual optokinetic system emerged near 18 months and declined to ∼40% below normal by 34 months. Daily application of the synthetic peptide elamipretide, which has high selectivity for mitochondrial membranes that contain cardiolipin and promotes efficient electron transfer, was able to mitigate visual decline from 18 months onwards. Daily application from 24 months onwards, i.e. when acuity had reduced by ∼16%, reversed visual decline and normalized function within 2 months. Recovered function persisted for at least 3 months after treatment was withdrawn and a single treatment at 24 months delayed subsequent visual decline. Elamipretide applied daily from 32 months onwards took longer to take effect, but substantial improvement was found within 2 months. The effects of age and elamipretide treatment on contrast sensitivity were similar to those on acuity, systemic and eye drop applications of elamipretide had comparable effects, scotopic spatial visual function was largely unaffected by age or treatment, and altered function was independent of variation in optical clarity. These data indicate that elamipretide treatment adaptively alters the aging visual system. They also provide a rationale to investigate whether mitochondrial dysfunction is a treatable pathophysiology of human visual aging and age-related visual disease.
Collapse
Affiliation(s)
- N. M. Alam
- Burke Neurological Institute, 785 Mamaroneck Avenue, White Plains, New York, NY 10605, USA
| | - R. M. Douglas
- University of British Columbia, Department of Ophthalmology and Visual Sciences, 2550 Willow Street, Vancouver, BCCanadaV5Z 3N9
| | - G. T. Prusky
- Burke Neurological Institute, 785 Mamaroneck Avenue, White Plains, New York, NY 10605, USA
- Weill Cornell Medicine, Department of Physiology and Biophysics, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
39
|
Seo C, Park S, Kim Y, Ji M, Lee HS, Hwang YH, Choi S, Min J, Oh SJ, Yee ST, Lee W, Paik MJ. Metabolomic analysis of amino acids and organic acids in aging mouse eyes using gas chromatography-tandem mass spectrometry. Biomed Chromatogr 2021; 36:e5298. [PMID: 34913179 DOI: 10.1002/bmc.5298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 11/12/2022]
Abstract
This is a metabolomics study for monitoring of altered amino acid (AA) and organic acid (OA) metabolism of in the eyes from aging mouse model of 8-weeks, 18-weeks, and 18-months. Simultaneous metabolic profiling analysis of OAs and AAs was performed as ethoxycarbonyl/methoxime/tert-butyldimethylsilyl derivatives by gas chromatography-tandem mass spectrometry. A total 42 metabolites of 24 AAs and 18 OAs were determined and their composition values were normalized to the corresponding mean values of 8-week-old mice as control group. Then their normalized values were plotted as star graphs, which were distorted and readily distinguishable for age-related each group. Among 42 metabolites, 18 AAs and 11 OAs were age dependent and significantly different (p < 0.05). Principal component analysis and partial least squares discriminant analysis showed unclear separation between 8- and 18-week-old mice but clear separation between these and 18-month-old mice. Especially, the variables importance in projection (VIP) scores of 4-hydroxyproline, cis-aconitic acid, glycine, isocitric acid, leucine, pipecolic acid and lysine from PLS-DA were higher than 1.3. A heatmap for the classification and visualization of 42 metabolites showed differences in metabolite changes with aging. Altered AA and OA profiles were monitored, which may explain for metabolic disturbance of AA and OA. These findings are related with mitochondrial dysfunctions related with energy metabolism and impaired antioxidant system in the aging eye. Therefore, the present metabolomics results of the association between physiological states and altered metabolism of AA and OA will be useful for understanding aging eye and related diseases.
Collapse
Affiliation(s)
- Chan Seo
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Sehoon Park
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Youngbae Kim
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Moongi Ji
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Hyeon-Seong Lee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea.,Natural Product Informatics Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangwon-do, Republic of Korea
| | - Yun-Ho Hwang
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Subin Choi
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Jeuk Min
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Song-Jin Oh
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Sung-Tae Yee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Wonjae Lee
- College of Pharmacy, Chosun University, Gwangju, Republic of Korea
| | - Man-Jeong Paik
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| |
Collapse
|
40
|
Tsantilas KA, Cleghorn WM, Bisbach CM, Whitson JA, Hass DT, Robbings BM, Sadilek M, Linton JD, Rountree AM, Valencia AP, Sweetwyne MT, Campbell MD, Zhang H, Jankowski CSR, Sweet IR, Marcinek DJ, Rabinovitch PS, Hurley JB. An Analysis of Metabolic Changes in the Retina and Retinal Pigment Epithelium of Aging Mice. Invest Ophthalmol Vis Sci 2021; 62:20. [PMID: 34797906 PMCID: PMC8606884 DOI: 10.1167/iovs.62.14.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose The purpose of this study was to present our hypothesis that aging alters metabolic function in ocular tissues. We tested the hypothesis by measuring metabolism in aged murine tissues alongside retinal responses to light. Methods Scotopic and photopic electroretinogram (ERG) responses in young (3–6 months) and aged (23–26 months) C57Bl/6J mice were recorded. Metabolic flux in retina and eyecup explants was quantified using U-13C-glucose or U-13C-glutamine with gas chromatography-mass spectrometry (GC-MS), O2 consumption rate (OCR) in a perifusion apparatus, and quantifying adenosine triphosphatase (ATP) with a bioluminescence assay. Results Scotopic and photopic ERG responses were reduced in aged mice. Glucose metabolism, glutamine metabolism, OCR, and ATP pools in retinal explants were mostly unaffected in aged mice. In eyecups, glutamine usage in the Krebs Cycle decreased while glucose metabolism, OCR, and ATP pools remained stable. Conclusions Our examination of metabolism showed negligible impact of age on retina and an impairment of glutamine anaplerosis in eyecups. The metabolic stability of these tissues ex vivo suggests age-related metabolic alterations may not be intrinsic. Future experiments should focus on determining whether external factors including nutrient supply, oxygen availability, or structural changes influence ocular metabolism in vivo.
Collapse
Affiliation(s)
- Kristine A Tsantilas
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Whitney M Cleghorn
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Celia M Bisbach
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Jeremy A Whitson
- Department of Biology, Davidson College, Davidson, North Carolina, United States
| | - Daniel T Hass
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Brian M Robbings
- Department of Biochemistry, University of Washington, Seattle, Washington, United States.,UW Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Martin Sadilek
- Department of Chemistry, University of Washington, Seattle, Washington, United States
| | - Jonathan D Linton
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Austin M Rountree
- UW Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Ana P Valencia
- Department of Radiology, University of Washington, Seattle, Washington, United States
| | - Mariya T Sweetwyne
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington, United States
| | - Matthew D Campbell
- Department of Radiology, University of Washington, Seattle, Washington, United States
| | - Huiliang Zhang
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| | - Connor S R Jankowski
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States
| | - Ian R Sweet
- UW Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, United States
| | - Peter S Rabinovitch
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington, United States
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, Washington, United States.,Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| |
Collapse
|
41
|
Quantitative Proteomic and Metabolomic Profiling Reveals Altered Mitochondrial Metabolism and Folate Biosynthesis Pathways in the Aging Drosophila Eye. Mol Cell Proteomics 2021; 20:100127. [PMID: 34332122 PMCID: PMC8385154 DOI: 10.1016/j.mcpro.2021.100127] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/09/2021] [Accepted: 07/21/2021] [Indexed: 11/26/2022] Open
Abstract
Aging is associated with increased risk of ocular disease, suggesting that age-associated molecular changes in the eye increase its vulnerability to damage. Although there are common pathways involved in aging at an organismal level, different tissues and cell types exhibit specific changes in gene expression with advanced age. Drosophila melanogaster is an established model system for studying aging and neurodegenerative disease that also provides a valuable model for studying age-associated ocular disease. Flies, like humans, exhibit decreased visual function and increased risk of retinal degeneration with age. Here, we profiled the aging proteome and metabolome of the Drosophila eye and compared these data with age-associated transcriptomic changes from both eyes and photoreceptors to identify alterations in pathways that could lead to age-related phenotypes in the eye. Of note, the proteomic and metabolomic changes observed in the aging eye are distinct from those observed in the head or whole fly, suggesting that tissue-specific changes in protein abundance and metabolism occur in the aging fly. Our integration of the proteomic, metabolomic, and transcriptomic data reveals that changes in metabolism, potentially due to decreases in availability of B vitamins, together with chronic activation of the immune response, may underpin many of the events observed in the aging Drosophila eye. We propose that targeting these pathways in the genetically tractable Drosophila system may help to identify potential neuroprotective approaches for neurodegenerative and age-related ocular diseases. Data are available via ProteomeXchange with identifier PXD027090. Tissue-specific changes in protein abundance occur in the aging Drosophila eye. Increase in mitochondrial metabolism enzyme abundance in the aging eye. Decrease in corneal lens protein abundance and calcium buffering in the aging eye. Dysregulated metabolism impacts vitamin B and methionine metabolism in the aging eye.
Collapse
|
42
|
Leley SP, Ciulla TA, Bhatwadekar AD. Diabetic Retinopathy in the Aging Population: A Perspective of Pathogenesis and Treatment. Clin Interv Aging 2021; 16:1367-1378. [PMID: 34290499 PMCID: PMC8289197 DOI: 10.2147/cia.s297494] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022] Open
Abstract
The elderly population in the United States is projected to almost double by the year 2050. In addition, the numbers of diabetics are rising, along with its most common complication, diabetic retinopathy (DR). To effectively treat DR within the elderly population, it is essential first to consider the retinal changes that occur due to aging, such as decreased blood flow, retinal thinning, and microglial changes, and understand that these changes can render the retina more vulnerable to oxidative and ischemic damage. Given these considerations, as well as the pathogenesis of DR, specific pathways could play a heightened role in DR progression in elderly patients, such as the polyol pathway and the vascular endothelial growth factor (VEGF) axis. Current ocular treatments include intravitreal corticosteroids, intravitreal anti-VEGF agents, laser photocoagulation and surgical interventions, in addition to better control of underlying diabetes with an expanding range of systemic treatments. While using therapeutics, it is also essential to consider how pharmacokinetics and pharmacodynamics change with aging; oral drug absorption can decrease, and ocular drug metabolism might affect the dosing and delivery methods. Also, elderly patients may more likely be nonadherent to their medication regimen or appointments than younger patients, and undertreatment with anti-VEGF drugs often leads to suboptimal outcomes. With a rising number of elderly DR patients, understanding how aging affects disease progression, pharmacological metabolism, and adherence are crucial to ensuring that this population receives adequate care.
Collapse
Affiliation(s)
- Sameer P Leley
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas A Ciulla
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, IN, USA
- Clearside Biomedical, Inc., Alpharetta, GA, USA
- Midwest Eye Institute, Indianapolis, IN, USA
| | - Ashay D Bhatwadekar
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
43
|
Wan W, Zhu W, Wu Y, Long Y, Liu H, Wan W, Wan G, Yu J. Grape Seed Proanthocyanidin Extract Moderated Retinal Pigment Epithelium Cellular Senescence Through NAMPT/SIRT1/NLRP3 Pathway. J Inflamm Res 2021; 14:3129-3143. [PMID: 34285539 PMCID: PMC8286255 DOI: 10.2147/jir.s306456] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Background Retinal pigment epithelium (RPE) cellular senescence is an important process in degenerative retinal disorders. Grape seed proanthocyanidin extract (GSPE) alleviates senescence-related degenerative disorders; however, the potential effects of GSPE intake on RPE cellular senescence through regulating NAMPT/SIRT1/NLRP3 pathway remain unclear. Methods The effects of GSPE on NAMPT expression and NAD+ contents were detected with Western blot and assay kit in both in-vivo and in-vitro AMD models. Senescence-related biomarkers, including p16, p21 expressions and β-gal staining, were conducted in different groups. The protective effects of GSPE treatment on the mitochondrial homeostasis and barrier function of RPE cells were detected using mtDNA lesions analyses, JC-1 staining, ZO1 staining and trans-epithelial cell resistance (TEER) detection. The expression of senescence-associated secretory phenotype (SASP) in different groups would be conducted with qPCR. To demonstrate the potential effects of NAMPT/SIRT1/NLRP3 pathway after GSPE treatment, the protein levels of relevant key regulators after applications of NAMPT inhibitor, Fk866, and SIRT1 inhibitor, EX-527. Results GSPE significantly improves the NAMPT expression and NAD+ content in aging mice, and thus alleviates the RPE cellular senescence. In advanced in-vitro studies, GSPE significantly up-regulated NAMPT content and thus relieved H2O2 induced NAD+ depression through analyzing the NAD+ contents in different groups. In advanced analyses, it was reported that GSPE could alleviate mitochondrial permeability, mtDNA damage, ZO1 expression and SASP levels in aging RPE cells. Thus, GSPE treatment significantly decreased senescence-related protein p16 and p21, as well as SASP levels in in-vitro aging model, and it was demonstrated that GSPE could illustrate a significant anti-aging effect. The Western blot data in GSPE treatment of aging RPE cells demonstrated that GSPE could significantly improve NAMPT and SIRT1 levels, and thus depressed NLRP3 expression. Conclusion This study indicated that GSPE alleviated RPE cellular senescence through NAMPT/SIRT1/NLRP3 pathway. This study highlighted the potential effects of GSPE on degenerative retinopathy through the crosstalk of NAD+ metabolism, SIRT1 function and NLRP3 activation.
Collapse
Affiliation(s)
- Wencui Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Wei Zhu
- Department of Ophthalmology, Changshu No. 2 People's Hospital, Changshu, People's Republic of China
| | - Yan Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China.,Mois Biotech Company, Shanghai, People's Republic of China
| | - Yang Long
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Hongzhuo Liu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Weiwei Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Guangming Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jing Yu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
44
|
Curative effect and possible mechanism of taurine on early corneal alkali burns. Chin Med J (Engl) 2021; 135:744-746. [PMID: 34172618 PMCID: PMC9276421 DOI: 10.1097/cm9.0000000000001570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Indexed: 11/26/2022] Open
|
45
|
Age-related differences in corneal nerve regeneration after SMILE and the mechanism revealed by metabolomics. Exp Eye Res 2021; 209:108665. [PMID: 34118276 DOI: 10.1016/j.exer.2021.108665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 11/20/2022]
Abstract
PURPOSE To investigate the effect of age on wound healing after small incision lenticule extraction (SMILE) and the underlying metabolomic mechanisms. METHODS This prospective study was conducted on 216 patients in four groups: the 18-20 (n = 38, Group I), 21-30 (n = 84, Group Ⅱ), 31-40 (n = 58, Group Ⅲ), and 41-50 (n = 36, Group IV) age groups. The density of corneal epithelial wing cells, basal cells, corneal stromal cells, endothelial cells and corneal nerves were examined with a laser confocal microscope (HRT III-RCM) before and 1 month, 3 month, 6 month and 1 year after SMILE. The central nerve fiber length (CNFL), the central corneal nerve fibre density (CNFD), and the central corneal nerve branch density (CNBD) were analyzed by Nero J. The corneal stroma lenticules were obtained from SMILE to analyze metabolites by high-performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry (HPLC-QTOF-MS). RESULTS The density of corneal wing epithelial cells and basal epithelial cells have no significant difference among the four groups. The CNFL was 21.90 ± 1.68 mm/mm2 in Group Ⅰ and 21.63 ± 2.09 mm/mm2 in Group Ⅱ after 1 year of SMILE, which represented a return to the preoperative level, whereas the CNFL of Group Ⅲ (19.40 ± 0.98 mm/mm2) and Group Ⅳ (18.94 ± 0.72 mm/mm2) were lower than that preoperation (P ˂0.01). CNFL repair had a negative correlation with age after surgery (Pearson's R = -0.572, P ˂0.01). The CNFD and the CNBD showed the same trend with the CNFL (Pearson's R = -0.602 and -0.531, P ˂0.05). Through screening the significantly different metabolites between the 18-30 age group (including Group I and Group Ⅱ) and other two groups, 6 common remarkably different metabolites were identified. Meanwhile, 5 unique different metabolites were identified only between the 18-30 age group and the 31-40 age group. Six unique different metabolites were identified only between the 18-30 age group and the 41-50 age group. CONCLUSION Corneal nerve repair after SMILE was significantly affected by age. The identified age-associated differences in metabolites were mainly related to inflammation, oxidation, nerve protection and regeneration.
Collapse
|
46
|
Kong W, Xiao Y, Wang B, Zhu Z, Hu L, Tang H, Wang K, Fang H, Shi Y, Long J, Gan L, Wang H, Sun Y, Xia Z. Comorbidities of scars in China: a national study based on hospitalized cases. BURNS & TRAUMA 2021; 9:tkab012. [PMID: 34212062 PMCID: PMC8240520 DOI: 10.1093/burnst/tkab012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Background Scar comorbidities seriously affect the physical and mental health of patients, but few studies have reported the exact epidemiological characteristics of scar comorbidities in China. This study aimed to investigate the prevalence of scar comorbidities in China. Methods The data of 177,586 scar cases between 2013 and 2018 were obtained from the Hospital Quality Monitoring System based on the 10th edition of the International Classification of Diseases coding system. The total distribution of scar comorbidities and their relationship with age, aetiology and body regions were analysed. Results Six comorbidities (contracture, malformation, ocular complications, adhesion, infection and others) were the main focus. In general, male patients outnumbered females and urban areas outnumbered rural areas. The proportion of contractures was the highest at 59,028 (33.24%). Students, workers and farmers made up the majority of the occupation. Han Chinese accounted for the majority of the ethnic. The highest proportion of scar contracture occurred at 1-1.9 years of age (58.97%), after which a significant downward trend was observed. However, starting from 50 years of age, ocular complications increased gradually and significantly, eventually reaching a peak of 34.49% in those aged >80 years. Scar contracture was the most common comorbidity according to aetiology, and the highest proportion was observed in patients who were scalded (29.33%). Contractures were also the most frequent comorbidity in hands (10.30%), lower limbs (6.97%), feet (6.80%) and upper limbs (6.02%). The mean and median hospitalization durations were 12.85 and 8 days, respectively. Conclusions Contractures were the most common comorbidities, and different comorbidities tended to occur at different ages and with different causative factors.
Collapse
Affiliation(s)
- Weishi Kong
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China.,Burn Institute of PLA, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai 200433, P.R.China
| | - Yongqiang Xiao
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China.,Department of Burn and Plastic Surgery, The 970th Hospital of People's Liberation Army, Yantai, Shandong, 264000, China
| | - Baoli Wang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China
| | - Zhe Zhu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China
| | - Lunyang Hu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China
| | - Hongtai Tang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China
| | - Kangan Wang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China
| | - He Fang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China
| | - Ying Shi
- China Standard Medical Information Research Centre, Shenzhen 518000, P.R.China
| | - Jianyan Long
- China Standard Medical Information Research Centre, Shenzhen 518000, P.R.China
| | - Lanxia Gan
- China Standard Medical Information Research Centre, Shenzhen 518000, P.R.China
| | - Haibo Wang
- China Standard Medical Information Research Centre, Shenzhen 518000, P.R.China
| | - Yu Sun
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China.,Burn Institute of PLA, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai 200433, P.R.China
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, P.R.China.,Burn Institute of PLA, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai 200433, P.R.China
| |
Collapse
|
47
|
Abstract
The outer retina is nourished from the choroid, a capillary bed just inside the sclera. O2, glucose, and other nutrients diffuse out of the choroid and then filter through a monolayer of retinal pigment epithelium (RPE) cells to fuel the retina. Recent studies of energy metabolism have revealed striking differences between retinas and RPE cells in the ways that they extract energy from fuels. The purpose of this review is to suggest and evaluate the hypothesis that the retina and RPE have complementary metabolic roles that make them depend on each other for survival and for their abilities to perform essential and specialized functions. Expected final online publication date for the Annual Review of Vision Science, Volume 7 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- James B Hurley
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington 98115, USA;
| |
Collapse
|
48
|
Ferdous S, Liao KL, Gefke ID, Summers VR, Wu W, Donaldson KJ, Kim YK, Sellers JT, Dixon JA, Shelton DA, Markand S, Kim SM, Zhang N, Boatright JH, Nickerson JM. Age-Related Retinal Changes in Wild-Type C57BL/6J Mice Between 2 and 32 Months. Invest Ophthalmol Vis Sci 2021; 62:9. [PMID: 34100889 PMCID: PMC8196434 DOI: 10.1167/iovs.62.7.9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 04/21/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose The purpose of this study was to extend our understanding of how aging affects normal retina function and morphology in wild-type C57BL/6J mice, by analyzing electrophysiological recordings and in vivo and post mortem anatomy. Methods Electroretinograms (ERGs), spectral domain optical coherence tomography (SD-OCT), and confocal scanning laser ophthalmoscope (cSLO) in vivo images were obtained from mice between the ages of 2 and 32 months in four groups: group 1 (<0.5 years), group 2 (1.0-1.5 years), group 3 (1.5-2.0 years), and group 4 (>2.0 years). Afterward, mouse bodies and eyes were weighed. Eyes were stained with hematoxylin and eosin (H&E) and cell nuclei were quantified. Results With aging, mice showed a significant reduction in both a- and b-wave ERG amplitudes in scotopic and photopic conditions. Additionally, total retina and outer nuclear layer (ONL) thickness, as measured by SD-OCT images, were significantly reduced in older groups. The cSLO images showed an increase in auto-fluorescence at the photoreceptor-RPE interface as age increases. H&E cell nuclei quantification showed significant reduction in the ONL in older ages, but no differences in the inner nuclear layer (INL) or ganglion cell layer (GCL). Conclusions By using multiple age groups and extending the upper age limit of our animals to approximately 2.65 years (P970), we found that natural aging causes negative effects on retinal function and morphology in a gradual, rather than abrupt, process. Future studies should investigate the exact mechanisms that contribute to these gradual declines in order to discover pathways that could potentially serve as therapeutic targets.
Collapse
Affiliation(s)
- Salma Ferdous
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Kristie L. Liao
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Isabelle D. Gefke
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Vivian R. Summers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Wenfei Wu
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- The First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shan'xi, China
| | - Kevin J. Donaldson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Yong-Kyu Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Gangdong-gu, Seoul, South Korea
| | - Jana T. Sellers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jendayi A. Dixon
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Shanu Markand
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Somin M. Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Nan Zhang
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
49
|
Dhingra A, Sharp RC, Kim T, Popov AV, Ying GS, Pietrofesa RA, Park K, Christofidou-Solomidou M, Boesze-Battaglia K. Assessment of a Small Molecule Synthetic Lignan in Enhancing Oxidative Balance and Decreasing Lipid Accumulation in Human Retinal Pigment Epithelia. Int J Mol Sci 2021; 22:5764. [PMID: 34071220 PMCID: PMC8198017 DOI: 10.3390/ijms22115764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 01/03/2023] Open
Abstract
Visual function depends on the intimate structural, functional and metabolic interactions between the retinal pigment epithelium (RPE) and the neural retina. The daily phagocytosis of the photoreceptor outer segment tips by the overlaying RPE provides essential nutrients for the RPE itself and photoreceptors through intricate metabolic synergy. Age-related retinal changes are often characterized by metabolic dysregulation contributing to increased lipid accumulation and peroxidation as well as the release of proinflammatory cytokines. LGM2605 is a synthetic lignan secoisolariciresinol diglucoside (SDG) with free radical scavenging, antioxidant and anti-inflammatory properties demonstrated in diverse in vitro and in vivo inflammatory disease models. In these studies, we tested the hypothesis that LGM2605 may be an attractive small-scale therapeutic that protects RPE against inflammation and restores its metabolic capacity under lipid overload. Using an in vitro model in which loss of the autophagy protein, LC3B, results in defective phagosome degradation and metabolic dysregulation, we show that lipid overload results in increased gasdermin cleavage, IL-1 β release, lipid accumulation and decreased oxidative capacity. The addition of LGM2605 resulted in enhanced mitochondrial capacity, decreased lipid accumulation and amelioration of IL-1 β release in a model of defective lipid homeostasis. Collectively, these studies suggest that lipid overload decreases mitochondrial function and increases the inflammatory response, with LGM2605 acting as a protective agent.
Collapse
Affiliation(s)
- Anuradha Dhingra
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (R.C.S.)
| | - Rachel C. Sharp
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (R.C.S.)
| | - Taewan Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Anatoliy V. Popov
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Gui-Shuang Ying
- Center for Preventive Ophthalmology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (G.-S.Y.); (K.P.)
| | - Ralph A. Pietrofesa
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (R.A.P.); (M.C.-S.)
| | - Kyewon Park
- Center for Preventive Ophthalmology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (G.-S.Y.); (K.P.)
| | - Melpo Christofidou-Solomidou
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (R.A.P.); (M.C.-S.)
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (R.C.S.)
| |
Collapse
|
50
|
Shi D, Tan Q, Ruan J, Tian Z, Wang X, Liu J, Liu X, Liu Z, Zhang Y, Sun C, Niu Y. Aging-related markers in rat urine revealed by dynamic metabolic profiling using machine learning. Aging (Albany NY) 2021; 13:14322-14341. [PMID: 34016789 PMCID: PMC8202887 DOI: 10.18632/aging.203046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 04/29/2021] [Indexed: 12/29/2022]
Abstract
The process of aging and metabolism is intimately intertwined; thus, developing biomarkers related to metabolism is critical for delaying aging. However, few studies have identified reliable markers that reflect aging trajectories based on machine learning. We generated metabolomic profiles from rat urine using ultra-performance liquid chromatography/mass spectrometry. This was dynamically collected at four stages of the rat's age (20, 50, 75, and 100 weeks) for both the training and test groups. Partial least squares-discriminant analysis score plots revealed a perfect separation trajectory in one direction with increasing age in the training and test groups. We further screened 25 aging-related biomarkers through the combination of four algorithms (VIP, time-series, LASSO, and SVM-RFE) in the training group. They were validated in the test group with an area under the curve of 1. Finally, six metabolites, known or novel aging-related markers, were identified, including epinephrine, glutarylcarnitine, L-kynurenine, taurine, 3-hydroxydodecanedioic acid, and N-acetylcitrulline. We also found that, except for N-acetylcitrulline (p < 0.05), the identified aging-related metabolites did not differ between tumor-free and tumor-bearing rats at 100 weeks (p > 0.05). Our findings reveal the metabolic trajectories of aging and provide novel biomarkers as potential therapeutic antiaging targets.
Collapse
Affiliation(s)
- Dan Shi
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
- Department of Nutrition and Food Hygiene, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Qilong Tan
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Jingqi Ruan
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Zhen Tian
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Xinyue Wang
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Jinxiao Liu
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Xin Liu
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Zhipeng Liu
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Yuntao Zhang
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Changhao Sun
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Yucun Niu
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, PR China
| |
Collapse
|