1
|
Jang SY, Yang JY, Park JH, Kim Y, An S, Jung WH, Park JW, Han JW, Kim JH, Park HS, Lyu J, Park TK. The Role of the Mitogen-Activated Protein Kinase Pathway in the Development of Laser-Induced Choroidal Neovascularization. Int J Mol Sci 2025; 26:2585. [PMID: 40141227 PMCID: PMC11942168 DOI: 10.3390/ijms26062585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The role of the mitogen-activated protein kinase (MAPK) pathway in choroidal neovascularization (CNV) remains unclear. This study investigates the involvement of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 pathways in CNV development, as well as the therapeutic potential of sprouty 2 (SPRY2), an MAPK inhibitor, in a laser-induced mouse model. The expressions of ERK, JNK, and p38 proteins were analyzed using Western blotting and immunostaining. Immunofluorescence imaging revealed increased p-ERK and p-JNK expression in the retina, retinal pigment epithelium (RPE), and choroid up to day 7. Co-immunostaining showed p-ERK colocalized with CD31, CD11b, F4/80, cytokeratin, and GFAP in the retina, while p-JNK and p-p38 were associated with angiogenesis and inflammation throughout the retina and choroid. Compared to aflibercept, SPRY2 administration significantly inhibited CNV lesions, endothelial proliferation, fibrosis, and apoptosis, while better-preserving RPE integrity. SPRY2-treated mice showed a stronger reduction in CNV-related inflammation, epithelial-mesenchymal transition, and photoreceptor apoptosis. These results highlight the MAPK pathway's role in CNV pathogenesis, with ERK primarily mediating Müller cell gliosis and JNK, contributing to angiogenesis and inflammation. SPRY2 effectively suppressed CNV lesions, supporting its potential as a therapeutic target for CNV treatment via MAPK pathway modulation.
Collapse
Affiliation(s)
- Sun Young Jang
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (S.Y.J.); (J.H.P.); (W.H.J.); (J.W.H.); (J.H.K.); (H.S.P.)
| | - Jin Young Yang
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| | - Jin Hwan Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (S.Y.J.); (J.H.P.); (W.H.J.); (J.W.H.); (J.H.K.); (H.S.P.)
| | - Yeji Kim
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea; (Y.K.); (S.A.)
| | - Sumin An
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea; (Y.K.); (S.A.)
| | - Wook Hyun Jung
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (S.Y.J.); (J.H.P.); (W.H.J.); (J.W.H.); (J.H.K.); (H.S.P.)
| | - Jong-Whi Park
- Department of Life Sciences, Gachon University, Incheon 21936, Republic of Korea;
| | - Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (S.Y.J.); (J.H.P.); (W.H.J.); (J.W.H.); (J.H.K.); (H.S.P.)
| | - Jin Ha Kim
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (S.Y.J.); (J.H.P.); (W.H.J.); (J.W.H.); (J.H.K.); (H.S.P.)
| | - Hyo Song Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (S.Y.J.); (J.H.P.); (W.H.J.); (J.W.H.); (J.H.K.); (H.S.P.)
| | - Jungmook Lyu
- Department of Medical Science, Konyang University, Daejun 32992, Republic of Korea
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (S.Y.J.); (J.H.P.); (W.H.J.); (J.W.H.); (J.H.K.); (H.S.P.)
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| |
Collapse
|
2
|
Huang H, Zeng J, Yu X, Du H, Wen C, Mao Y, Tang H, Kuang X, Liu W, Yu H, Liu H, Li B, Long C, Yan J, Shen H. Establishing chronic models of age-related macular degeneration via long-term iron ion overload. Am J Physiol Cell Physiol 2024; 326:C1367-C1383. [PMID: 38406826 DOI: 10.1152/ajpcell.00532.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/26/2024] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
Age-related macular degeneration (AMD) is characterized by the degenerative senescence in the retinal pigment epithelium (RPE) and photoreceptors, which is accompanied by the accumulation of iron ions in the aging retina. However, current models of acute oxidative stress are still insufficient to simulate the gradual progression of AMD. To address this, we established chronic injury models by exposing the aRPE-19 cells, 661W cells, and mouse retina to iron ion overload over time. Investigations at the levels of cell biology and molecular biology were performed. It was demonstrated that long-term treatment of excessive iron ions induced senescence-like morphological changes, decreased cell proliferation, and impaired mitochondrial function, contributing to apoptosis. Activation of the mitogen-activated protein kinase (MAPK) pathway and the downstream molecules were confirmed both in the aRPE-19 and 661W cells. Furthermore, iron ion overload resulted in dry AMD-like lesions and decreased visual function in the mouse retina. These findings suggest that chronic exposure to overloading iron ions plays a significant role in the pathogenesis of retinopathy and provide a potential model for future studies on AMD.NEW & NOTEWORTHY To explore the possibility of constructing reliable research carriers on age-related macular degeneration (AMD), iron ion overload was applied to establish models in vitro and in vivo. Subsequent investigations into cellular physiology and molecular biology confirmed the presence of senescence in these models. Through this study, we hope to provide a better option of feasible methods for future researches into AMD.
Collapse
Affiliation(s)
- Hao Huang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, People's Republic of China
| | - Jingshu Zeng
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinyue Yu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Han Du
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chaojuan Wen
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Mao
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Han Tang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xielan Kuang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei Liu
- Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, People's Republic of China
| | - Huan Yu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huijun Liu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Eye Fundus Department, Affiliated Aier Eye Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Bowen Li
- Eye Center of Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chongde Long
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianhua Yan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
3
|
Jia X, Sun Y, Wang T, Zhong L, Deng J, Zhu X. Mechanism of circular RNA-mediated regulation of L-DOPA to improve wet age-related macular degeneration. Gene 2023; 861:147247. [PMID: 36736867 DOI: 10.1016/j.gene.2023.147247] [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: 11/30/2022] [Revised: 01/14/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
This study aimed to investigate the effect and mechanism of levodopa (L-DOPA) in the treatment of age-related macular degeneration (AMD). A wet AMD cell model was created via CoCl2 treatment of ARPE-19 cells. The cytoprotective effects of L-DOPA in the model were determined using CCK-8, flow cytometry, TUNEL, qPCR, and ELISA assays. Subsequently, circRNA sequencing and bioinformatics analysis were used to screen differentially expressed circRNAs, which were overexpressed in ARPE-19 cells, to explore their role in wet AMD. The findings revealed that 200 μM CoCl2 treatment inhibited the cell viability and the production of tyrosinase, melanin, and pigment epithelium-derived growth factor but promoted apoptosis and the expression of vascular endothelial growth factor in ARPE-19 cells. Moreover, 20 μM L-DOPA exerted the best therapeutic effect on the model. qPCR showed that Hsa_circ_0018401 (circ-SGMS1) was significantly differentially expressed in each experimental group, which was consistent with the sequencing results. The overexpression of circ-SGMS1 in ARPE-19 cells reversed the effects of CoCl2. Fluorescence in situ hybridization showed that circ-SGMS1 was expressed more in the nucleus than in the cytoplasm. qPCR assays indicated that circ-SGMS1 overexpression did not have a significant effect on the expressions of VEGFA and KDR but significantly reduced the expressions of HIF-1a and THBS1. Circ-SGMS1 is of immense significance in the AMD treatment mechanism of L-DOPA. Overexpression of circ-SGMS1 may alleviate wet AMD by inhibiting HIF-1a and THBS1 expression.
Collapse
Affiliation(s)
- Xiuhua Jia
- Department of Ophthalmology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yi Sun
- Department of Ophthalmology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Wang
- Department of Ophthalmology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lei Zhong
- Department of Ophthalmology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Juan Deng
- Department of Ophthalmology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xiang Zhu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Pariente A, Pérez-Sala Á, Ochoa R, Bobadilla M, Villanueva-Martínez Á, Peláez R, Larráyoz IM. Identification of 7-Ketocholesterol-Modulated Pathways and Sterculic Acid Protective Effect in Retinal Pigmented Epithelium Cells by Using Genome-Wide Transcriptomic Analysis. Int J Mol Sci 2023; 24:ijms24087459. [PMID: 37108627 PMCID: PMC10144535 DOI: 10.3390/ijms24087459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries. AMD is characterized by the formation of lipidic deposits between the retinal pigment epithelium (RPE) and the choroid called drusen. 7-Ketocholesterol (7KCh), an oxidized-cholesterol derivative, is closely related to AMD as it is one of the main molecules accumulated in drusen. 7KCh induces inflammatory and cytotoxic responses in different cell types, and a better knowledge of the signaling pathways involved in its response would provide a new perspective on the molecular mechanisms that lead to the development of AMD. Furthermore, currently used therapies for AMD are not efficient enough. Sterculic acid (SA) attenuates the 7KCh response in RPE cells and is presented as an alternative to improve these therapies. By using genome-wide transcriptomic analysis in monkey RPE cells, we have provided new insight into 7KCh-induced signaling in RPE cells, as well as the protective capacity of SA. 7KCh modulates the expression of several genes associated with lipid metabolism, endoplasmic reticulum stress, inflammation and cell death and induces a complex response in RPE cells. The addition of SA successfully attenuates the deleterious effect of 7KCh and highlights its potential for the treatment of AMD.
Collapse
Affiliation(s)
- Ana Pariente
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Álvaro Pérez-Sala
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Rodrigo Ochoa
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
- Proteomics Research Core Facility, Aragonese Institute of Health Sciences (IACS), University of Zaragoza, San Juan Bosco 13, 50009 Zaragoza, Spain
| | - Miriam Bobadilla
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Ángela Villanueva-Martínez
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Rafael Peláez
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
| | - Ignacio M Larráyoz
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006 Logroño, Spain
- Biomarkers, Artificial Intelligence and Signaling (BIAS), Department of Nursing, University of La Rioja, Duquesa de la Victoria 88, 26006 Logroño, Spain
| |
Collapse
|
5
|
MAPK Pathways in Ocular Pathophysiology: Potential Therapeutic Drugs and Challenges. Cells 2023; 12:cells12040617. [PMID: 36831285 PMCID: PMC9954064 DOI: 10.3390/cells12040617] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathways represent ubiquitous cellular signal transduction pathways that regulate all aspects of life and are frequently altered in disease. Once activated through phosphorylation, these MAPKs in turn phosphorylate and activate transcription factors present either in the cytoplasm or in the nucleus, leading to the expression of target genes and, as a consequence, they elicit various biological responses. The aim of this work is to provide a comprehensive review focusing on the roles of MAPK signaling pathways in ocular pathophysiology and the potential to influence these for the treatment of eye diseases. We summarize the current knowledge of identified MAPK-targeting compounds in the context of ocular diseases such as macular degeneration, cataract, glaucoma and keratopathy, but also in rare ocular diseases where the cell differentiation, proliferation or migration are defective. Potential therapeutic interventions are also discussed. Additionally, we discuss challenges in overcoming the reported eye toxicity of some MAPK inhibitors.
Collapse
|
6
|
Muraleva NA, Kolosova NG. Alteration of the MEK1/2–ERK1/2 Signaling Pathway in the Retina Associated with Age and Development of AMD-Like Retinopathy. BIOCHEMISTRY (MOSCOW) 2023; 88:179-188. [PMID: 37072329 DOI: 10.1134/s0006297923020025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Age-related macular degeneration (AMD) is a complex neurodegenerative disease and a major cause of irreversible visual impairment in patients in developed countries. Although age is the greatest risk factor in AMD, molecular mechanisms involved in AMD remain unknown. Growing evidence shows that dysregulation of MAPK signaling contributes to aging and neurodegenerative diseases; however, the information on the role of MAPK upregulation in these processes is controversial. ERK1 and ERK2 participate in the maintenance of proteostasis through the regulation of protein aggregation induced by the endoplasmic reticulum stress and other stress-mediated cell responses. To assess the contribution of alterations in the ERK1/2 signaling to the AMD development, we compared age-associated changes in the activity of ERK1/2 signaling pathway in the retina of Wistar rats (control) and OXYS rats that develop AMD-like retinopathy spontaneously. The activity of the ERK1/2 signaling increased during physiological aging in the retina of Wistar rats. The manifestation and progression of the AMD-like pathology in the retina of OXYS rats was accompanied by hyperphosphorylation of ERK1/2 and MEK1/2, the key kinases of the ERK1/2 signaling pathway. The progression of the AMD-like pathology was also associated with the ERK1/2-dependent tau protein hyperphosphorylation and increase in the ERK1/2-dependent phosphorylation of alpha B crystallin at Ser45 in the retina.
Collapse
Affiliation(s)
- Natalia A Muraleva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | - Nataliya G Kolosova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
7
|
Zekavat SM, Raghu VK, Trinder M, Ye Y, Koyama S, Honigberg MC, Yu Z, Pampana A, Urbut S, Haidermota S, O’Regan DP, Zhao H, Ellinor PT, Segrè AV, Elze T, Wiggs JL, Martone J, Adelman RA, Zebardast N, Del Priore L, Wang JC, Natarajan P. Deep Learning of the Retina Enables Phenome- and Genome-Wide Analyses of the Microvasculature. Circulation 2022; 145:134-150. [PMID: 34743558 PMCID: PMC8746912 DOI: 10.1161/circulationaha.121.057709] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The microvasculature, the smallest blood vessels in the body, has key roles in maintenance of organ health and tumorigenesis. The retinal fundus is a window for human in vivo noninvasive assessment of the microvasculature. Large-scale complementary machine learning-based assessment of the retinal vasculature with phenome-wide and genome-wide analyses may yield new insights into human health and disease. METHODS We used 97 895 retinal fundus images from 54 813 UK Biobank participants. Using convolutional neural networks to segment the retinal microvasculature, we calculated vascular density and fractal dimension as a measure of vascular branching complexity. We associated these indices with 1866 incident International Classification of Diseases-based conditions (median 10-year follow-up) and 88 quantitative traits, adjusting for age, sex, smoking status, and ethnicity. RESULTS Low retinal vascular fractal dimension and density were significantly associated with higher risks for incident mortality, hypertension, congestive heart failure, renal failure, type 2 diabetes, sleep apnea, anemia, and multiple ocular conditions, as well as corresponding quantitative traits. Genome-wide association of vascular fractal dimension and density identified 7 and 13 novel loci, respectively, that were enriched for pathways linked to angiogenesis (eg, vascular endothelial growth factor, platelet-derived growth factor receptor, angiopoietin, and WNT signaling pathways) and inflammation (eg, interleukin, cytokine signaling). CONCLUSIONS Our results indicate that the retinal vasculature may serve as a biomarker for future cardiometabolic and ocular disease and provide insights into genes and biological pathways influencing microvascular indices. Moreover, such a framework highlights how deep learning of images can quantify an interpretable phenotype for integration with electronic health record, biomarker, and genetic data to inform risk prediction and risk modification.
Collapse
Affiliation(s)
- Seyedeh Maryam Zekavat
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT (S.M.Z., J.M., R.A.A., L.D.P., J.C.W.)
- Computational Biology & Bioinformatics Program (S.M.Z., Y.Y., H.Z.), Yale University, New Haven, CT
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
| | - Vineet K. Raghu
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
- Cardiovascular Research Center (S.M.Z., V.K.R., M.C.H., S.U., S.H., P.T.E., P.N.), Massachusetts General Hospital, Harvard Medical School, Boston
- Cardiovascular Imaging Research Center (V.K.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mark Trinder
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada (M.T.)
| | - Yixuan Ye
- Computational Biology & Bioinformatics Program (S.M.Z., Y.Y., H.Z.), Yale University, New Haven, CT
| | - Satoshi Koyama
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
| | - Michael C. Honigberg
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
- Cardiovascular Research Center (S.M.Z., V.K.R., M.C.H., S.U., S.H., P.T.E., P.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Zhi Yu
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
| | - Akhil Pampana
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
| | - Sarah Urbut
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
- Cardiovascular Research Center (S.M.Z., V.K.R., M.C.H., S.U., S.H., P.T.E., P.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sara Haidermota
- Cardiovascular Research Center (S.M.Z., V.K.R., M.C.H., S.U., S.H., P.T.E., P.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Declan P. O’Regan
- MRC London Institute of Medical Sciences, Imperial College London, UK (D.P.O.)
| | - Hongyu Zhao
- Computational Biology & Bioinformatics Program (S.M.Z., Y.Y., H.Z.), Yale University, New Haven, CT
- School of Public Health (H.Z.), Yale University, New Haven, CT
| | - Patrick T. Ellinor
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
- Cardiovascular Research Center (S.M.Z., V.K.R., M.C.H., S.U., S.H., P.T.E., P.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Ayellet V. Segrè
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston (A.V.S., T.E., J.L.W., N.Z.)
| | - Tobias Elze
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston (A.V.S., T.E., J.L.W., N.Z.)
| | - Janey L. Wiggs
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston (A.V.S., T.E., J.L.W., N.Z.)
| | - James Martone
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT (S.M.Z., J.M., R.A.A., L.D.P., J.C.W.)
| | - Ron A. Adelman
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT (S.M.Z., J.M., R.A.A., L.D.P., J.C.W.)
| | - Nazlee Zebardast
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston (A.V.S., T.E., J.L.W., N.Z.)
| | - Lucian Del Priore
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT (S.M.Z., J.M., R.A.A., L.D.P., J.C.W.)
| | - Jay C. Wang
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT (S.M.Z., J.M., R.A.A., L.D.P., J.C.W.)
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.M.Z., V.K.R., M.T., S.K., M.C.H., Z.Y., A.P., S.U., P.T.E., P.N.)
- Cardiovascular Research Center (S.M.Z., V.K.R., M.C.H., S.U., S.H., P.T.E., P.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
8
|
APOE2 promotes the development and progression of subretinal neovascularization in age-related macular degeneration via MAPKs signaling pathway. Saudi J Biol Sci 2020; 27:2770-2777. [PMID: 32994736 PMCID: PMC7499293 DOI: 10.1016/j.sjbs.2020.06.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
Background Neovascular age-related macular degeneration (nvAMD) is one of the main pathological features of wet AMD. Apolipoprotein E2 is involved in the formation of nvAMD but the molecular mechanism has not been reported. Methods The APOE alleles in AMD patients were detected by genotyping. Mouse models were divided into 4 groups according to transfection different gene segments and laser-induced treatment. APOE2, VEGF, PDGF-BB, b-FGF and inflammatory cytokines (including p-NF-κB, TNF-α, IL-1β and IL-6) were tested by ELISA in mice retinal lysate. The formation of nvAMD in the indicated treatment groups at 3rd, 7th and 14th day after laser-induced damage were detected by FFA. Besides, qRT-PCR was used to determine the mRNA levels of p38, JNK and ERK in ARPE-19 cells. Finally, the inflammatory cytokines and MAPK proteins (including P38, p-P38, JNK, p-JNK, ERK and p-ERK) were detected by western blot. Results The statistics of APOE alleles showed that APOE2 allele carriers were more likely to nvAMD. VEGF, PDGF-BB, b-FGF and related inflammatory cytokines were up-regulated significantly after treatment with APOE2, which were reduced after silencing the MAPK family genes, however. Further, the expression levels of neovascular growth factors and inflammatory cytokines were highly consistent between mouse models and ARPE-19 cells. Besides, the phosphorylation levels of p38, JNK and ERK were affected by APOE2. Conclusion nvAMD was affected directly by the overexpression of VEGF, PDGF-BB and b-FGF, which were regulated by APOE2 through activating MAPKs pathway.
Collapse
|
9
|
Shen Y, Li M, Liu K, Xu X, Zhu S, Wang N, Guo W, Zhao Q, Lu P, Yu F, Xu X. Integrated bioinformatics analysis of aberrantly-methylated differentially-expressed genes and pathways in age-related macular degeneration. BMC Ophthalmol 2020; 20:119. [PMID: 32209064 PMCID: PMC7092446 DOI: 10.1186/s12886-020-01392-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/13/2020] [Indexed: 11/11/2022] Open
Abstract
Background Age-related macular degeneration (AMD) represents the leading cause of visual impairment in the aging population. The goal of this study was to identify aberrantly-methylated, differentially-expressed genes (MDEGs) in AMD and explore the involved pathways via integrated bioinformatics analysis. Methods Data from expression profile GSE29801 and methylation profile GSE102952 were obtained from the Gene Expression Omnibus database. We analyzed differentially-methylated genes and differentially-expressed genes using R software. Functional enrichment and protein–protein interaction (PPI) network analysis were performed using the R package and Search Tool for the Retrieval of Interacting Genes online database. Hub genes were identified using Cytoscape. Results In total, 827 and 592 genes showed high and low expression, respectively, in GSE29801; 4117 hyper-methylated genes and 511 hypo-methylated genes were detected in GSE102952. Based on overlap, we categorized 153 genes as hyper-methylated, low-expression genes (Hyper-LGs) and 24 genes as hypo-methylated, high-expression genes (Hypo-HGs). Four Hyper-LGs (CKB, PPP3CA, TGFB2, SOCS2) overlapped with AMD risk genes in the Public Health Genomics and Precision Health Knowledge Base. KEGG pathway enrichment analysis indicated that Hypo-HGs were enriched in the calcium signaling pathway, whereas Hyper-LGs were enriched in sphingolipid metabolism. In GO analysis, Hypo-HGs were enriched in fibroblast migration, membrane raft, and coenzyme binding, among others. Hyper-LGs were enriched in mRNA transport, nuclear speck, and DNA binding, among others. In PPI network analysis, 23 nodes and two edges were established from Hypo-HGs, and 151 nodes and 73 edges were established from Hyper-LGs. Hub genes (DHX9, MAPT, PAX6) showed the greatest overlap. Conclusion This study revealed potentially aberrantly MDEGs and pathways in AMD, which might improve the understanding of this disease.
Collapse
Affiliation(s)
- Yinchen Shen
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, People's Republic of China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Mo Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Kun Liu
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, People's Republic of China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Xiaoyin Xu
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, People's Republic of China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Shaopin Zhu
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, People's Republic of China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Ning Wang
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, People's Republic of China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Wenke Guo
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Qianqian Zhao
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Ping Lu
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Fudong Yu
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, People's Republic of China. .,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.
| |
Collapse
|
10
|
PPARG Polymorphisms Are Associated with Unexplained Mild Vision Loss in Patients with Type 2 Diabetes Mellitus. J Ophthalmol 2019; 2019:5284867. [PMID: 31915541 PMCID: PMC6930731 DOI: 10.1155/2019/5284867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 01/17/2023] Open
Abstract
Objectives To investigate whether the presence of peroxisome proliferator-activated receptor gamma (PPARG) gene polymorphisms is associated with unexplained mild visual impairment (UMVI) in patients with type 2 diabetes mellitus (T2DM). Methods A total of 135 T2DM residents with UMVI and 133 with normal vision (NV; best-corrected visual acuity ≥ 20/25 in both eyes) were enrolled. UMVI was defined as best-corrected visual acuity (BCVA) < 20/25 and ≥ 20/63 in both eyes, with no visual impairment-causing diseases found. Four PPARG gene single-nucleotide polymorphisms (SNPs) (rs3856806, rs1801282, rs709158, and rs10865710) were assessed with the HAPLOVIEW 4.0 software to examine the statistical association of PPARG polymorphisms and UMVI in patients with T2DM. Results Four SNPs qualified the Hardy-Weinberg equilibrium (p > 0.05). The frequency of genotype GC at SNP rs10865710 was significantly higher in the UMVI group than in the NV group (p < 0.001; GG + GC versus CC) (OR = 8.94, 95% CI: 4.90-16.31), whereas genotype CC decreased the risk (OR = 0.07, 95% CI: 0.03-0.14). Genotype TT at SNP rs3856806 was strongly associated with UMVI (p < 0.0001, TT + TC versus CC) (OR = 4.74, 95% CI: 2.68-8.54), whereas genotype CC appeared to be protective for UMVI (OR = 0.55, 95% CI: 0.37-0.82). Conclusions Susceptibilities of PPARG variants may lead to differences in PPARG transcription, result in early function loss of retinal photoreceptor cells, and eventually cause UMVI.
Collapse
|
11
|
Cheng Z, Yao W, Zheng J, Ding W, Wang Y, Zhang T, Zhu L, Zhou F. A derivative of betulinic acid protects human Retinal Pigment Epithelial (RPE) cells from cobalt chloride-induced acute hypoxic stress. Exp Eye Res 2018; 180:92-101. [PMID: 30578788 DOI: 10.1016/j.exer.2018.12.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 02/05/2023]
Abstract
The Retinal Pigment Epithelium (RPE) is a monolayer of cells located above the choroid. It mediates human visual cycle and nourishes photoreceptors. Hypoxia-induced oxidative stress to RPE is a vital cause of retinal degeneration such as the Age-related Macular Degeneration. Most of these retinal diseases are irreversible with no efficient treatment, therefore protecting RPE cells from hypoxia stress is an important way to prevent or slow down the progression of retinal degeneration. Betulinic acid (BA) and betulin (BE) are pentacyclic triterpenoids with anti-oxidative property, but little is known about their effect on RPE cells. We investigated the protective effect of BA, BE and their derivatives against cobalt chloride-induced hypoxia stress in RPE cells. Human ARPE-19 cells were exposed to BA, BE and their eighteen derivatives (named as H3H20) that we customized through replacing moieties at C3 and C28 positions. We found that cobalt chloride reduced cell viability, increased Reactive Oxygen Species (ROS) production as well as induced apoptosis and necrosis in ARPE-19 cells. Interestingly, the pretreatment of 3-O-acetyl-glycyl- 28-O-glycyl-betulinic acid effectively protected cells from acute hypoxia stress induced by cobalt chloride. Our immunoblotting results suggested that this derivative attenuated the cobalt chloride-induced activation of Akt, Erk and JNK pathways. All findings were further validated in human primary RPE cells. In summary, this BA derivate has protective effect against the acute hypoxic stress in human RPE cells and may be developed into a candidate agent effective in the prevention of prevalent retinal diseases.
Collapse
Affiliation(s)
- Zhengqi Cheng
- School of Pharmacy, The University of Sydney, NSW, 2006, Australia
| | - Wenjuan Yao
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jian Zheng
- Center for Bioactive Products, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Weimin Ding
- School of Chemical and Environmental Engineering, Harbin University of Science and Technology, Harbin, 150080, Heilongjiang, China
| | - Yang Wang
- Center for Bioactive Products, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Ting Zhang
- Save Sight Institute, The University of Sydney, Sydney, NSW, 2000, Australia; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Zhu
- Save Sight Institute, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Fanfan Zhou
- School of Pharmacy, The University of Sydney, NSW, 2006, Australia.
| |
Collapse
|
12
|
Jiang D, Liu X, Hu J. Topical administration of Esculetin as a potential therapy for experimental dry eye syndrome. Eye (Lond) 2017; 31:1724-1732. [PMID: 28643798 PMCID: PMC5733282 DOI: 10.1038/eye.2017.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 04/27/2017] [Indexed: 01/16/2023] Open
Abstract
PurposeIn this study, we investigated the therapeutic effects of topical Esculetin for dry eye rabbits through the ocular tests, inflammatory factor levels and specific phosphorylated protein expressions of ERK1/2 singnal pathway.Patients and methodsThirty-two healthy adult male New Zealand white rabbits were chosen for the study. DES models were established after removing of the main lacrimal gland, Harderian gland and nictitating membrane in the left eyes and randomly divided into group DES control, group CsA, group Esculetin and group Esculetin combined with CsA (C&E), meanwhile the right eyes served as group Normal control. Schirmer's I tests, fluorescein scores, goblet cell densities, inflammatory cytokines IL-1α,IL-1β,TNF-αlevels were observed by slit-lamp microscope, conjunctival impression cytology and ELISA essay at week 0, 1, 2, 4, 8. Phosphorylated-ERK1/2 expressions were detected in Western blot analysis at week 8.ResultsAfter induction of DES, aqueous tear production and goblet cell density were decreased, FL score was much higher in group DES control throughout the study (P<0.05). Both topical Esculetin and Esculetin combing CsA increased the SIT values (10±1 mm, 14±3 mm, P<0.05) and goblet cell densities (77±12/HP, 92±12/HP, P<0.05), decreased FL scores (7.48±0.33, 5.09±0.24, P<0.05) at week 8. Alternations of IL-1α,IL-1β,TNF-αlevels had similar trend. In Western blot analysis, downregulations of p-ERK1/2 were observed in therapy groups when compared with group DES control and the most decreasing was found in group C&E (P<0.05).ConclusionTopical Esculetin improved DES symptoms, downregulated the inflammatory cytokine expressions, suppressed the ERK1/2 pathway and enhanced the therapeutic effect of CsA.
Collapse
Affiliation(s)
- D Jiang
- Department of Ophthalmology, Longhua Hosptial, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - X Liu
- Department of Ophthalmology, Longhua Hosptial, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - J Hu
- Department of Ophthalmology, Longhua Hosptial, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Kheitan S, Minuchehr Z, Soheili ZS. Exploring the cross talk between ER stress and inflammation in age-related macular degeneration. PLoS One 2017; 12:e0181667. [PMID: 28742151 PMCID: PMC5524348 DOI: 10.1371/journal.pone.0181667] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
Increasing evidence demonstrates that inflammation and endoplasmic reticulum (ER) stress is implicated in the development and progression of age-related macular degeneration (AMD), a multifactorial neurodegenerative disease. However the cross talk between these cellular mechanisms has not been clearly and fully understood. The present study investigates a possible intersection between ER stress and inflammation in AMD. In this study, we recruited two collections of involved protein markers to retrieve their interaction information from IMEx-curated databases, which are the most well- known protein-protein interaction collections, allowing us to design an intersection network for AMD that is unprecedented. In order to find expression activated subnetworks, we utilized AMD expression profiles in our network. In addition, we studied topological characteristics of the most expressed active subnetworks to identify the hubs. With regard to topological quantifications and expressional activity, we reported a list of the most pivotal hubs which are potentially applicable as probable therapeutic targets. Furthermore, we introduced MAPK signaling pathway as a significantly involved pathway in the association between ER stress and inflammation, leading to promising new directions in discovering AMD formation mechanisms and possible treatments.
Collapse
Affiliation(s)
- Samira Kheitan
- Systems Biotechnology Department, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zarrin Minuchehr
- Systems Biotechnology Department, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- * E-mail:
| | - Zahra-Soheila Soheili
- Molecular Medicine Department, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
14
|
Plössl K, Weber BHF, Friedrich U. The X-linked juvenile retinoschisis protein retinoschisin is a novel regulator of mitogen-activated protein kinase signalling and apoptosis in the retina. J Cell Mol Med 2016; 21:768-780. [PMID: 27995734 PMCID: PMC5345684 DOI: 10.1111/jcmm.13019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/26/2016] [Indexed: 02/01/2023] Open
Abstract
X-linked juvenile retinoschisis (XLRS) is a hereditary retinal dystrophy in young males, caused by mutations in the RS1 gene. The function of the encoded protein, termed retinoschisin, and the molecular mechanisms underlying XLRS pathogenesis are still unresolved, although a direct interaction partner of the secreted retinoschisin, the retinal Na/K-ATPase, was recently identified. Earlier gene expression studies in retinoschisin-deficient (Rs1h-/Y ) mice provided a first indication of pathological up-regulation of mitogen-activated protein (MAP) kinase signalling in disease pathogenesis. To further investigate the role for retinoschisin in MAP kinase regulation, we exposed Y-79 cells and murine Rs1h-/Y retinae to recombinant retinoschisin and the XLRS-associated mutant RS1-C59S. Although normal retinoschisin stably bound to retinal cells, RS1-C59S exhibited a strongly reduced binding affinity. Simultaneously, exposure to normal retinoschisin significantly reduced phosphorylation of C-RAF and MAP kinases ERK1/2 in Y-79 cells and murine Rs1h-/Y retinae. Expression of MAP kinase target genes C-FOS and EGR1 was also down-regulated in both model systems. Finally, retinoschisin treatment decreased pro-apoptotic BAX-2 transcript levels in Y-79 cells and Rs1h-/Y retinae. Upon retinoschisin treatment, these cells showed increased resistance against apoptosis, reflected by decreased caspase-3 activity (in Y-79 cells) and increased photoreceptor survival (in Rs1h-/Y retinal explants). RS1-C59S did not influence C-RAF or ERK1/2 activation, C-FOS or EGR1 expression, or apoptosis. Our data imply that retinoschisin is a novel regulator of MAP kinase signalling and exerts an anti-apoptotic effect on retinal cells. We therefore discuss that disturbances of MAP kinase signalling by retinoschisin deficiency could be an initial step in XLRS pathogenesis.
Collapse
Affiliation(s)
- Karolina Plössl
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Ulrike Friedrich
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| |
Collapse
|
15
|
Mishra S, Peterson K, Yin L, Berger A, Fan J, Wistow G. Accumulation of cholesterol and increased demand for zinc in serum-deprived RPE cells. Mol Vis 2016; 22:1387-1404. [PMID: 28003730 PMCID: PMC5166821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/08/2016] [Indexed: 11/03/2022] Open
Abstract
PURPOSE Having observed that confluent ARPE-19 cells (derived from human RPE) survive well in high-glucose serum-free medium (SFM) without further feeding for several days, we investigated the expression profile of RPE cells under the same conditions. METHODS Expression profiles were examined with microarray and quantitative PCR (qPCR) analyses, followed by western blot analysis of key regulated proteins. The effects of low-density lipoprotein (LDL) and zinc supplementation were examined with qPCR. Immunofluorescence was used to localize the LDL receptor and to examine LDL uptake. Cellular cholesterol levels were measured with filipin binding. Expression patterns in primary fetal RPE cells were compared using qPCR. RESULTS Microarray analyses of gene expression in ARPE-19, confirmed with qPCR, showed upregulation of lipid and cholesterol biosynthesis pathways in SFM. At the protein level, the cholesterol synthesis control factor SRBEF2 was activated, and other key lipid synthesis proteins increased. Supplementation of SFM with LDL reversed the upregulation of lipid and cholesterol synthesis genes, but not of cholesterol transport genes. The LDL receptor relocated to the plasma membrane, and LDL uptake was activated by day 5-7 in SFM, suggesting increased demand for cholesterol. Confluent ARPE-19 cells in SFM accumulated intracellular cholesterol, compared with cells supplemented with serum, over 7 days. Over the same time course in SFM, the expression of metallothioneins decreased while the major zinc transporter was upregulated, consistent with a parallel increase in demand for zinc. Supplementation with zinc reversed expression changes for metallothionein genes, but not for other zinc-related genes. Similar patterns of regulation were also seen in primary fetal human RPE cells in SFM. CONCLUSIONS ARPE-19 cells respond to serum deprivation and starvation with upregulation of the lipid and cholesterol pathways, accumulation of intracellular cholesterol, and increased demand for zinc. Similar trends are seen in primary fetal RPE cells. Cholesterol accumulation basal to RPE is a prominent feature of age-related macular degeneration (AMD), while dietary zinc is protective. It is conceivable that accumulating defects in Bruch's membrane and dysfunction of the choriocapillaris could impede transport between RPE and vasculature in AMD. Thus, this pattern of response to serum deprivation in RPE-derived cells may have relevance for some aspects of the progression of AMD.
Collapse
Affiliation(s)
- Sanghamitra Mishra
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Katherine Peterson
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Lili Yin
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Alan Berger
- Lowe Family Genomics Core, Johns Hopkins University - School of Medicine, Baltimore, MD
| | - Jianguo Fan
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Graeme Wistow
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
16
|
Kyosseva SV. Targeting MAPK Signaling in Age-Related Macular Degeneration. OPHTHALMOLOGY AND EYE DISEASES 2016; 8:23-30. [PMID: 27385915 PMCID: PMC4920203 DOI: 10.4137/oed.s32200] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/08/2016] [Accepted: 05/13/2016] [Indexed: 12/26/2022]
Abstract
Age-related macular degeneration (AMD) is a major cause of irreversible blindness affecting elderly people in the world. AMD is a complex multifactorial disease associated with demographic, genetics, and environmental risk factors. It is well established that oxidative stress, inflammation, and apoptosis play critical roles in the pathogenesis of AMD. The mitogen-activated protein kinase (MAPK) signaling pathways are activated by diverse extracellular stimuli, including growth factors, mitogens, hormones, cytokines, and different cellular stressors such as oxidative stress. They regulate cell proliferation, differentiation, survival, and apoptosis. This review addresses the novel findings from human and animal studies on the relationship of MAPK signaling with AMD. The use of specific MAPK inhibitors may represent a potential therapeutic target for the treatment of this debilitating eye disease.
Collapse
Affiliation(s)
- Svetlana V Kyosseva
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
17
|
Simón MV, Agnolazza DL, German OL, Garelli A, Politi LE, Agbaga MP, Anderson RE, Rotstein NP. Synthesis of docosahexaenoic acid from eicosapentaenoic acid in retina neurons protects photoreceptors from oxidative stress. J Neurochem 2016; 136:931-46. [PMID: 26662863 DOI: 10.1111/jnc.13487] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/28/2015] [Accepted: 12/02/2015] [Indexed: 12/27/2022]
Abstract
Oxidative stress is involved in activating photoreceptor death in several retinal degenerations. Docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, protects cultured retina photoreceptors from apoptosis induced by oxidative stress and promotes photoreceptor differentiation. Here, we investigated whether eicosapentaenoic acid (EPA), a metabolic precursor to DHA, had similar effects and whether retinal neurons could metabolize EPA to DHA. Adding EPA to rat retina neuronal cultures increased opsin expression and protected photoreceptors from apoptosis induced by the oxidants paraquat and hydrogen peroxide (H2 O2 ). Palmitic, oleic, and arachidonic acids had no protective effect, showing the specificity for DHA. We found that EPA supplementation significantly increased DHA percentage in retinal neurons, but not EPA percentage. Photoreceptors and glial cells expressed Δ6 desaturase (FADS2), which introduces the last double bond in DHA biosynthetic pathway. Pre-treatment of neuronal cultures with CP-24879 hydrochloride, a Δ5/Δ6 desaturase inhibitor, prevented EPA-induced increase in DHA percentage and completely blocked EPA protection and its effect on photoreceptor differentiation. These results suggest that EPA promoted photoreceptor differentiation and rescued photoreceptors from oxidative stress-induced apoptosis through its elongation and desaturation to DHA. Our data show, for the first time, that isolated retinal neurons can synthesize DHA in culture. Docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in retina photoreceptors, and its precursor, eicosapentaenoic acid (EPA) have multiple beneficial effects. Here, we show that retina neurons in vitro express the desaturase FADS2 and can synthesize DHA from EPA. Moreover, addition of EPA to these cultures protects photoreceptors from oxidative stress and promotes their differentiation through its metabolization to DHA.
Collapse
Affiliation(s)
- María Victoria Simón
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Daniela L Agnolazza
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Olga Lorena German
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Andrés Garelli
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Luis E Politi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| | - Martin-Paul Agbaga
- Cell Biology, Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA.,Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA
| | - Robert E Anderson
- Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA.,Ophthalmology/Cell Biology, Univ of Oklahoma Hlth Sci Ctr, Oklahoma City, Oklahoma, USA
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Department of Biology, Biochemistry and Pharmacy, Univ Nacional del Sur (UNS)-CONICET, Bahía Blanca, Buenos Aires, Argentina
| |
Collapse
|
18
|
SanGiovanni JP, Chen J, Gupta AS, Smith LEH, Sapieha P, Lee PH. Netrin-1 - DCC Signaling Systems and Age-Related Macular Degeneration. PLoS One 2015; 10:e0125548. [PMID: 25950802 PMCID: PMC4423995 DOI: 10.1371/journal.pone.0125548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 03/25/2015] [Indexed: 01/04/2023] Open
Abstract
We conducted a nested candidate gene study and pathway-based enrichment analysis on data from a multi-national 77,000-person project on the molecular genetics of age-related macular degeneration (AMD) to identify AMD-associated DNA-sequence variants in genes encoding constituents of a netrin-1 (NTN1)-based signaling pathway that converges on DNA-binding transcription complexes through a 3'-5'-cyclic adenosine monophosphate-calcineurin (cAMP-CN)-dependent axis. AMD-associated single nucleotide polymorphisms (SNPs) existed in 9 linkage disequilibrium-independent genomic regions; these included loci overlapping NTN1 (rs9899630, P ≤ 9.48 x 10-5), DCC (Deleted in Colorectal Cancer)—the gene encoding a primary NTN1 receptor (rs8097127, P ≤ 3.03 x 10-5), and 6 other netrin-related genes. Analysis of the NTN1-DCC pathway with exact methods demonstrated robust enrichment with AMD-associated SNPs (corrected P-value = 0.038), supporting the idea that processes driven by NTN1-DCC signaling systems operate in advanced AMD. The NTN1-DCC pathway contains targets of FDA-approved drugs and may offer promise for guiding applied clinical research on preventive and therapeutic interventions for AMD.
Collapse
Affiliation(s)
- John Paul SanGiovanni
- National Institute of Alcohol Abuse and Alcoholism, Section on Nutritional Neuroscience, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail:
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA, United States of America
| | - Ankur S. Gupta
- University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Lois E. H. Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA, United States of America
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC, Canada
| | - Phil H. Lee
- Analytic & Translational Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
19
|
SanGiovanni JP, Chew EY. Clinical applications of age-related macular degeneration genetics. Cold Spring Harb Perspect Med 2014; 4:cshperspect.a017228. [PMID: 25125423 DOI: 10.1101/cshperspect.a017228] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Understanding genetic causes of age-related macular degeneration (AMD) will eventually yield effective discoveries and improvements in predictive/prognostic methods. These include, but are not limited to, reliable disease prediction (screening for increased discrimination of clinical risk), differential classification of AMD subtypes with biomarkers (development of risk-linked molecular taxonomies), selection of optimal preventive and therapeutic interventions (guided by a biologically meaningful understanding of treatment response), and drug dosing. In this review, we discuss clinical applications informed by key findings in AMD genetics, and provide commentary on leveraging extant and forthcoming evidence to improve AMD risk prediction, AMD classification, and knowledge on the genetic basis of drug activity and toxicity. Advances in translating AMD genetics findings for AMD risk prediction require development of a genetics-based causality for AMD incidence and progression. Molecular subtyping of AMD phenotypes requires a set of dynamic biomarkers presenting prognostic value; although these have yet to be identified, the formation of multidisciplinary teams and their participation in large-scale consortia may yield promising results. Drugs targeting complement and vascular endothelial growth factor (VEGF) systems are under evaluation, and forthcoming work on rare variants and noncoding DNA in AMD pathogenesis will likely reveal biochemical pathways enriched with AMD-associated genetic variants. Pharmacologic targets in these pathways may inform a rational and effective therapeutic approach to preventing and treating this sight-threatening disease.
Collapse
Affiliation(s)
- John Paul SanGiovanni
- National Eye Institute, Clinical Trials Branch, Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Emily Y Chew
- National Eye Institute, Clinical Trials Branch, Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|