1
|
Qiao X, Shah W, Gao X, Gong Y, Li Y, Gao Y, Li J. Understanding how the immune system environment is controlled in high myopia cases. Int Immunopharmacol 2024; 143:113138. [PMID: 39362012 DOI: 10.1016/j.intimp.2024.113138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 10/05/2024]
Abstract
High myopia (HM) is characterized by a significant extension of the eye axis; it has emerged as a serious global public health issue recently. In addition to causing severe visual impairment, HM is associated with several problems that may compromise an individual's vision. Although genetic and environmental factors in HM have been extensively investigated, increasing evidence implicates the immune system and its microenvironment in its pathogenesis. In this review, we explore the complex interactions between cytokines, immune cells, and the eye environment to elucidate the complex processes controlling the immune response in HM. Furthermore, we investigated treatments modulating the immune response and alleviating the progression of HM and its complications. Through a review of the current relevant studies, we highlight the critical functions of the immune system in the multifactorial development of HM. With the evolving understanding of the immune system's involvement in HM, this review provides a valuable resource to clinicians and researchers to develop targeted interventions and personalized treatments for individuals with this vision-threatening condition.
Collapse
Affiliation(s)
- Xin Qiao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China
| | - Wahid Shah
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China; Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaoqin Gao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China
| | - Yuxing Gong
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China
| | - Yanan Li
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China
| | - Yuan Gao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China; Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China.
| | - Junhong Li
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China.
| |
Collapse
|
2
|
Belete GT, Zhou L, Li KK, So PK, Do CW, Lam TC. Metabolomics studies in common multifactorial eye disorders: a review of biomarker discovery for age-related macular degeneration, glaucoma, diabetic retinopathy and myopia. Front Mol Biosci 2024; 11:1403844. [PMID: 39193222 PMCID: PMC11347317 DOI: 10.3389/fmolb.2024.1403844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Multifactorial Eye disorders are a significant public health concern and have a huge impact on quality of life. The pathophysiological mechanisms underlying these eye disorders were not completely understood since functional and low-throughput biological tests were used. By identifying biomarkers linked to eye disorders, metabolomics enables early identification, tracking of the course of the disease, and personalized treatment. Methods The electronic databases of PubMed, Scopus, PsycINFO, and Web of Science were searched for research related to Age-Related macular degeneration (AMD), glaucoma, myopia, and diabetic retinopathy (DR). The search was conducted in August 2023. The number of cases and controls, the study's design, the analytical methods used, and the results of the metabolomics analysis were all extracted. Using the QUADOMICS tool, the quality of the studies included was evaluated, and metabolic pathways were examined for distinct metabolic profiles. We used MetaboAnalyst 5.0 to undertake pathway analysis of differential metabolites. Results Metabolomics studies included in this review consisted of 36 human studies (5 Age-related macular degeneration, 10 Glaucoma, 13 Diabetic retinopathy, and 8 Myopia). The most networked metabolites in AMD include glycine and adenosine monophosphate, while methionine, lysine, alanine, glyoxylic acid, and cysteine were identified in glaucoma. Furthermore, in myopia, glycerol, glutamic acid, pyruvic acid, glycine, cysteine, and oxoglutaric acid constituted significant metabolites, while glycerol, glutamic acid, lysine, citric acid, alanine, and serotonin are highly networked metabolites in cases of diabetic retinopathy. The common top metabolic pathways significantly enriched and associated with AMD, glaucoma, DR, and myopia were arginine and proline metabolism, methionine metabolism, glycine and serine metabolism, urea cycle metabolism, and purine metabolism. Conclusion This review recapitulates potential metabolic biomarkers, networks and pathways in AMD, glaucoma, DR, and myopia, providing new clues to elucidate disease mechanisms and therapeutic targets. The emergence of advanced metabolomics techniques has significantly enhanced the capability of metabolic profiling and provides novel perspectives on the metabolism and underlying pathogenesis of these multifactorial eye conditions. The advancement of metabolomics is anticipated to foster a deeper comprehension of disease etiology, facilitate the identification of novel therapeutic targets, and usher in an era of personalized medicine in eye research.
Collapse
Affiliation(s)
- Gizachew Tilahun Belete
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Lei Zhou
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Centre for Eye and Vision Research (CEVR), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - King-Kit Li
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Pui-Kin So
- University Research Facility in Life Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Chi-Wai Do
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Centre for Eye and Vision Research (CEVR), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for Chinese Medicine Innovation (RCMI), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Thomas Chuen Lam
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Centre for Eye and Vision Research (CEVR), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for Chinese Medicine Innovation (RCMI), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
3
|
Stone RA, Tobias JW, Wei W, Carlstedt X, Zhang L, Iuvone PM, Nickla DL. Diurnal gene expression patterns in retina and choroid distinguish myopia progression from myopia onset. PLoS One 2024; 19:e0307091. [PMID: 39028695 PMCID: PMC11259283 DOI: 10.1371/journal.pone.0307091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/30/2024] [Indexed: 07/21/2024] Open
Abstract
The world-wide prevalence of myopia (nearsightedness) is increasing, but its pathogenesis is incompletely understood. Among many putative mechanisms, laboratory and clinical findings have implicated circadian biology in the etiology of myopia. Consistent with a circadian hypothesis, we recently reported a marked variability in diurnal patterns of gene expression in two crucial tissues controlling post-natal refractive development - the retina and choroid-at the onset of form-deprivation myopia in chick, a widely studied and validated model. To extend these observations, we assayed gene expression by RNA-Seq in retina and choroid during the progression of established unilateral form-deprivation myopia of chick. We assayed gene expression every 4 hours during a single day from myopic and contralateral control eyes. Retinal and choroidal gene expression in myopic vs. control eyes during myopia progression differed strikingly at discrete times during the day. Very few differentially expressed genes occurred at more than one time in either tissue during progressing myopia. Similarly, Gene Set Enrichment Analysis pathways varied markedly by time during the day. Some of the differentially expressed genes in progressing myopia coincided with candidate genes for human myopia, but only partially corresponded with genes previously identified at myopia onset. Considering other laboratory findings and human genetics and epidemiology, these results further link circadian biology to the pathogenesis of myopia; but they also point to important mechanistic differences between the onset of myopia and the progression of established myopia. Future laboratory and clinical investigations should systematically incorporate circadian mechanisms in studying the etiology of myopia and in seeking more effective treatments to normalize eye growth in children.
Collapse
Affiliation(s)
- Richard A. Stone
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John W. Tobias
- Penn Genomics and Sequencing Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Wenjie Wei
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Xia Carlstedt
- Department of Biomedical Sciences and Disease, New England College of Optometry, Boston, Massachusetts, United States of America
| | - Lixin Zhang
- Department of Biomedical Sciences and Disease, New England College of Optometry, Boston, Massachusetts, United States of America
| | - P. Michael Iuvone
- Department of Ophthalmology & Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Debora L. Nickla
- Department of Biomedical Sciences and Disease, New England College of Optometry, Boston, Massachusetts, United States of America
| |
Collapse
|
4
|
Stone RA, Tobias JW, Wei W, Schug J, Wang X, Zhang L, Iuvone PM, Nickla DL. Diurnal retinal and choroidal gene expression patterns support a role for circadian biology in myopia pathogenesis. Sci Rep 2024; 14:533. [PMID: 38177383 PMCID: PMC10767138 DOI: 10.1038/s41598-023-50684-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 12/22/2023] [Indexed: 01/06/2024] Open
Abstract
The prevalence of myopia (nearsightedness) is increasing to alarming levels, but its etiology remains poorly understood. Because both laboratory and clinical findings suggest an etiologic role for circadian rhythms in myopia development, we assayed gene expression by RNA-Seq in retina and choroid at the onset of unilateral experimental myopia in chick, isolating tissues every 4 h during a single 24-h period from myopic and contralateral control eyes. Occluded versus open eye gene expression differences varied considerably over the 24-h sampling period, with some occurring at multiple times of day but with others showing differences at only a single investigated timepoint. Some of the genes identified in retina or choroid of chick myopia were previously identified as candidate genes for common human myopia. Like differentially expressed genes, pathways identified by Gene Set Enrichment Analysis also varied dramatically by sampling time. Considered with other laboratory data, human genetic and epidemiology data, these findings further implicate circadian events in myopia pathogenesis. The present results emphasize a need to include time of day in mechanistic studies of myopia and to assess circadian biology directly in trying to understand better the origin of myopia and to develop more effective therapies.
Collapse
Affiliation(s)
- Richard A Stone
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - John W Tobias
- Penn Genomics and Sequencing Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenjie Wei
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Schug
- Penn Genomics and Sequencing Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xia Wang
- Department of Biomedical Sciences and Disease, New England College of Optometry, Boston, MA, USA
| | - Lixin Zhang
- Department of Biomedical Sciences and Disease, New England College of Optometry, Boston, MA, USA
| | - P Michael Iuvone
- Department of Ophthalmology and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Debora L Nickla
- Department of Biomedical Sciences and Disease, New England College of Optometry, Boston, MA, USA
| |
Collapse
|
5
|
Kim H, Lee W, Kim YA, Yu S, Jeong J, Choi Y, Lee Y, Park YH, Kang MS, Kim MS, Kim TG. RNA-Sequencing Analysis Reveals the Role of Mitochondrial Energy Metabolism Alterations and Immune Cell Activation in Form-Deprivation and Lens-Induced Myopia in Mice. Genes (Basel) 2023; 14:2163. [PMID: 38136985 PMCID: PMC10743199 DOI: 10.3390/genes14122163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Myopia is a substantial global public health concern primarily linked to the elongation of the axial length of the eyeball. While numerous animal models have been employed to investigate myopia, the specific contributions of genetic factors and the intricate signaling pathways involved remain incompletely understood. In this study, we conducted RNA-seq analysis to explore genes and pathways in two distinct myopia-inducing mouse models: form-deprivation myopia (FDM) and lens-induced myopia (LIM). Comparative analysis with a control group revealed significant differential expression of 2362 genes in FDM and 503 genes in LIM. Gene Set Enrichment Analysis (GSEA) identified a common immune-associated pathway between LIM and FDM, with LIM exhibiting more extensive interactions. Notably, downregulation was observed in OxPhos complex III of FDM and complex IV of LIM. Subunit A of complex I was downregulated in LIM but upregulated in FDM. Additionally, complex V was upregulated in LIM but downregulated in FDM. These findings suggest a connection between alterations in energy metabolism and immune cell activation, shedding light on a novel avenue for understanding myopia's pathophysiology. Our research underscores the necessity for a comprehensive approach to comprehending myopia development, which integrates insights from energy metabolism, oxidative stress, and immune response pathways.
Collapse
Affiliation(s)
- Hojung Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
| | - Wonmin Lee
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
- Department of Medicine, Kyung Hee University College of Medicine, Seoul 02453, Republic of Korea
| | - Ye-Ah Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Sanghyeon Yu
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Jisu Jeong
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yueun Choi
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yoonsung Lee
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
| | - Yong Hwan Park
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Min Seok Kang
- Department of Ophthalmology, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul 02447, Republic of Korea;
| | - Man S. Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (H.K.); (W.L.); (Y.-A.K.); (S.Y.); (J.J.); (Y.C.); (Y.L.)
| | - Tae Gi Kim
- Department of Ophthalmology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea
| |
Collapse
|
6
|
Feng J, Zhang X, Li R, Zhao P, Han X, Wu Q, Tian Q, Tang G, Song J, Bi H. Widespread Involvement of Acetylation in the Retinal Metabolism of Form-Deprivation Myopia in Guinea Pigs. ACS OMEGA 2023; 8:23825-23839. [PMID: 37426266 PMCID: PMC10324097 DOI: 10.1021/acsomega.3c02219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023]
Abstract
Myopia has become the major cause of visual impairment worldwide. Although the pathogenesis of myopia remains controversial, proteomic studies suggest that dysregulation of retinal metabolism is potentially involved in the pathology of myopia. Lysine acetylation of proteins plays a key role in regulating cellular metabolism, but little is known about its role in the form-deprived myopic retina. Hence, a comprehensive analysis of proteomic and acetylomic changes in the retinas of guinea pigs with form-deprivation myopia was performed. In total, 85 significantly differential proteins and 314 significantly differentially acetylated proteins were identified. Notably, the differentially acetylated proteins were markedly enriched in metabolic pathways such as glycolysis/gluconeogenesis, the pentose phosphate pathway, retinol metabolism, and the HIF-1 signaling pathway. HK2, HKDC1, PKM, LDH, GAPDH, and ENO1 were the key enzymes in these metabolic pathways with decreased acetylation levels in the form-deprivation myopia group. Altered lysine acetylation of key enzymes in the form-deprived myopic retina might affect the dynamic balance of metabolism in the retinal microenvironment by altering their activity. In conclusion, as the first report on the myopic retinal acetylome, this study provides a reliable basis for further studies on myopic retinal acetylation.
Collapse
Affiliation(s)
- Jiaojiao Feng
- Shandong
University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Xiuyan Zhang
- Affiliated
Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong, China
- Shandong
Provincial Key Laboratory of Integrated Traditional Chinese and Western
Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Jinan 250002, Shandong, China
| | - Runkuan Li
- Shandong
University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Ping Zhao
- Affiliated
Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong, China
- Shandong
Provincial Key Laboratory of Integrated Traditional Chinese and Western
Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Jinan 250002, Shandong, China
| | - Xudong Han
- School
of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Qiuxin Wu
- Affiliated
Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong, China
- Shandong
Provincial Key Laboratory of Integrated Traditional Chinese and Western
Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Jinan 250002, Shandong, China
| | - Qingmei Tian
- Affiliated
Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong, China
- Shandong
Provincial Key Laboratory of Integrated Traditional Chinese and Western
Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Jinan 250002, Shandong, China
| | - Guodong Tang
- Affiliated
Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong, China
- Shandong
Provincial Key Laboratory of Integrated Traditional Chinese and Western
Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Jinan 250002, Shandong, China
| | - Jike Song
- Shandong
University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
- Affiliated
Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong, China
| | - Hongsheng Bi
- Affiliated
Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong, China
- Shandong
Provincial Key Laboratory of Integrated Traditional Chinese and Western
Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Jinan 250002, Shandong, China
| |
Collapse
|
7
|
Applications of Genomics and Transcriptomics in Precision Medicine for Myopia Control or Prevention. Biomolecules 2023; 13:biom13030494. [PMID: 36979429 PMCID: PMC10046175 DOI: 10.3390/biom13030494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/18/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Myopia is a globally emerging concern accompanied by multiple medical and socio-economic burdens with no well-established causal treatment to control thus far. The study of the genomics and transcriptomics of myopia treatment is crucial to delineate disease pathways and provide valuable insights for the design of precise and effective therapeutics. A strong understanding of altered biochemical pathways and underlying pathogenesis leading to myopia may facilitate early diagnosis and treatment of myopia, ultimately leading to the development of more effective preventive and therapeutic measures. In this review, we summarize current data about the genomics and transcriptomics of myopia in human and animal models. We also discuss the potential applicability of these findings to precision medicine for myopia treatment.
Collapse
|
8
|
Zeitz C, Roger JE, Audo I, Michiels C, Sánchez-Farías N, Varin J, Frederiksen H, Wilmet B, Callebert J, Gimenez ML, Bouzidi N, Blond F, Guilllonneau X, Fouquet S, Léveillard T, Smirnov V, Vincent A, Héon E, Sahel JA, Kloeckener-Gruissem B, Sennlaub F, Morgans CW, Duvoisin RM, Tkatchenko AV, Picaud S. Shedding light on myopia by studying complete congenital stationary night blindness. Prog Retin Eye Res 2023; 93:101155. [PMID: 36669906 DOI: 10.1016/j.preteyeres.2022.101155] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023]
Abstract
Myopia is the most common eye disorder, caused by heterogeneous genetic and environmental factors. Rare progressive and stationary inherited retinal disorders are often associated with high myopia. Genes implicated in myopia encode proteins involved in a variety of biological processes including eye morphogenesis, extracellular matrix organization, visual perception, circadian rhythms, and retinal signaling. Differentially expressed genes (DEGs) identified in animal models mimicking myopia are helpful in suggesting candidate genes implicated in human myopia. Complete congenital stationary night blindness (cCSNB) in humans and animal models represents an ON-bipolar cell signal transmission defect and is also associated with high myopia. Thus, it represents also an interesting model to identify myopia-related genes, as well as disease mechanisms. While the origin of night blindness is molecularly well established, further research is needed to elucidate the mechanisms of myopia development in subjects with cCSNB. Using whole transcriptome analysis on three different mouse models of cCSNB (in Gpr179-/-, Lrit3-/- and Grm6-/-), we identified novel actors of the retinal signaling cascade, which are also novel candidate genes for myopia. Meta-analysis of our transcriptomic data with published transcriptomic databases and genome-wide association studies from myopia cases led us to propose new biological/cellular processes/mechanisms potentially at the origin of myopia in cCSNB subjects. The results provide a foundation to guide the development of pharmacological myopia therapies.
Collapse
Affiliation(s)
- Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.
| | - Jérome E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, Saclay, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France
| | | | | | - Juliette Varin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, APHP, Paris, France
| | | | - Nassima Bouzidi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Frederic Blond
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Stéphane Fouquet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Vasily Smirnov
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Ajoy Vincent
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Elise Héon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France; Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Robert M Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Andrei V Tkatchenko
- Oujiang Laboratory, Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health, Wenzhou, China; Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, New York, NY, USA
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
9
|
Retinal Gene Expression of Selective Genes and Histological Stages of Embryonic and Post-Hatch Chickens (Gallus gallus). Genes (Basel) 2022; 13:genes13112048. [DOI: 10.3390/genes13112048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/17/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Chickens are excellent models for the study of retinal development and function. Gene expression at the correct time is crucial to retinal development and function. The present study aimed to investigate retinal gene expression and morphology in locally grown chickens at various developmental stages. RNA was extracted from the retina at the embryonic and post-hatch stages, and the retinal layers were stained with haematoxylin and eosin (H&E). RT-PCR and RT-qPCR were used for gene expression analysis of 14 selected genes. The results showed that all the retinal genes were expressed at different developmental stages. However, there were slight noticeable variations in expression patterns. At the morphological level, all retinal layers were well observed, except for the outer plexiform layer that became visible in the fifteen-day chick embryo. The current study provides a baseline for standard retinal gene expression of 14 genes and retinal histological staining. The selected genes have different roles in retinal development and function, and most of these genes are associated with retinal diseases. The results obtained here can be applied to molecular retinal research and retinal diseases with genetic factors in retina animal models or human diseases.
Collapse
|
10
|
Li Y, Lu Y, Du K, Yin Y, Hu T, Fu Q, Zhang Y, Wen D, Wu X, Xia X. RNA-sequencing analysis reveals the long noncoding RNA profile in the mouse myopic retina. Front Genet 2022; 13:1014031. [PMID: 36313450 PMCID: PMC9606684 DOI: 10.3389/fgene.2022.1014031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2023] Open
Abstract
Aim: Myopia is a prevalent public health problem. The long noncoding RNA (lncRNA) mechanisms for dysregulated retinal signaling in the myopic eye have remained elusive. The aim of this study was to analyze the expression profiles and possible pathogenic roles of lncRNAs in mouse form-deprived myopia (FDM) retinas. Methods: A mouse FDM model was induced and retinas from the FDM right eyes and the contralateral eyes were collected for RNA sequencing. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and lncRNA-mRNA coexpression network analyses were conducted to explore the biological functions of the differentially expressed lncRNAs. In addition, the levels of differentially expressed lncRNAs in the myopic retinas were validated by quantitative real-time PCR (qRT-PCR). Fluorescence in situ hybridization (FISH) was used to detect the localization of lncRNAs in mouse retinas. Results: FDM eyes exhibited reduced refraction and increased ocular axial length compared to control fellow eyes. RNA sequencing revealed that there were 655 differentially expressed lncRNAs between the FDM and control retinas. Functional enrichment analysis indicated that the differentially expressed RNAs were mostly enriched in cellular processes, cytokine-cytokine receptor interactions, retinol metabolism, and rhythmic processes. Differentially expressed lncRNAs were validated by qRT-PCR. Additionally, RNA FISH showed that XR_384718.4 (Gm35369) localized in the ganglion cell (GCL) and inner nuclear layers (INL). Conclusion: This study identified the differential expression profiles of lncRNAs in myopic mouse retinas. Our results provide scientific evidence for investigations of myopia and the development of putative interventions in the future.
Collapse
Affiliation(s)
- Yuanjun Li
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Lu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kaixuan Du
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yewei Yin
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tu Hu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuman Fu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yanni Zhang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Dan Wen
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoying Wu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Ji S, Ye L, Zhang L, Xu D, Dai J. Retinal neurodegeneration in a mouse model of green-light-induced myopia. Exp Eye Res 2022; 223:109208. [DOI: 10.1016/j.exer.2022.109208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/23/2022] [Accepted: 07/31/2022] [Indexed: 11/15/2022]
|
12
|
Zeng L, Li X, Liu J, Liu H, Xu H, Yang Z. RNA-Seq Analysis Reveals an Essential Role of the Tyrosine Metabolic Pathway and Inflammation in Myopia-Induced Retinal Degeneration in Guinea Pigs. Int J Mol Sci 2021; 22:ijms222212598. [PMID: 34830490 PMCID: PMC8618104 DOI: 10.3390/ijms222212598] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Myopia is the second leading cause of visual impairment globally. Myopia can induce sight-threatening retinal degeneration and the underlying mechanism remains poorly defined. We generated a model of myopia-induced early-stage retinal degeneration in guinea pigs and investigated the mechanism of action. Methods: The form-deprivation-induced myopia (FDM) was induced in the right eyes of 2~3-week-old guinea pigs using a translucent balloon for 15 weeks. The left eye remained untreated and served as a self-control. Another group of untreated age-matched animals was used as naïve controls. The refractive error and ocular biometrics were measured at 3, 7, 9, 12 and 15 weeks post-FDM induction. Visual function was evaluated by electroretinography. Retinal neurons and synaptic structures were examined by confocal microscopy of immunolabelled retinal sections. The total RNAs were extracted from the retinas and processed for RNA sequencing analysis. Results: The FDM eyes presented a progressive axial length elongation and refractive error development. After 15 weeks of intervention, the average refractive power was -3.40 ± 1.85 D in the FDM eyes, +2.94 ± 0.59 D and +2.69 ± 0.56 D in the self-control and naïve control eyes, respectively. The a-wave amplitude was significantly lower in FDM eyes and these eyes had a significantly lower number of rods, secretagogin+ bipolar cells, and GABAergic amacrine cells in selected retinal areas. RNA-seq analysis showed that 288 genes were upregulated and 119 genes were downregulated in FDM retinas compared to naïve control retinas. In addition, 152 genes were upregulated and 12 were downregulated in FDM retinas compared to self-control retinas. The KEGG enrichment analysis showed that tyrosine metabolism, ABC transporters and inflammatory pathways were upregulated, whereas tight junction, lipid and glycosaminoglycan biosynthesis were downregulated in FDM eyes. Conclusions: The long-term (15-week) FDM in the guinea pig models induced an early-stage retinal degeneration. The dysregulation of the tyrosine metabolism and inflammatory pathways may contribute to the pathogenesis of myopia-induced retinal degeneration.
Collapse
Affiliation(s)
- Ling Zeng
- Aier School of Ophthalmology, Central South University, Changsha 410000, China;
| | - Xiaoning Li
- Aier Eye Hospital, Changsha 410000, China;
- Aier School of Optometry and Vision Science, Hubei University of Science and Technology, Xian-ning 437100, China
- Aier Institute of Optometry and Vision Science, Changsha 410000, China; (J.L.); (H.L.)
| | - Jian Liu
- Aier Institute of Optometry and Vision Science, Changsha 410000, China; (J.L.); (H.L.)
| | - Hong Liu
- Aier Institute of Optometry and Vision Science, Changsha 410000, China; (J.L.); (H.L.)
| | - Heping Xu
- Aier School of Ophthalmology, Central South University, Changsha 410000, China;
- Aier Institute of Optometry and Vision Science, Changsha 410000, China; (J.L.); (H.L.)
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK
- Correspondence: (H.X.); (Z.Y.); Tel.: +44-(0)28909-76463 (H.X.); +86-(0)13380071988 (Z.Y.)
| | - Zhikuan Yang
- Aier School of Ophthalmology, Central South University, Changsha 410000, China;
- Aier School of Optometry and Vision Science, Hubei University of Science and Technology, Xian-ning 437100, China
- Aier Institute of Optometry and Vision Science, Changsha 410000, China; (J.L.); (H.L.)
- Correspondence: (H.X.); (Z.Y.); Tel.: +44-(0)28909-76463 (H.X.); +86-(0)13380071988 (Z.Y.)
| |
Collapse
|
13
|
Karouta C, Kucharski R, Hardy K, Thomson K, Maleszka R, Morgan I, Ashby R. Transcriptome-based insights into gene networks controlling myopia prevention. FASEB J 2021; 35:e21846. [PMID: 34405458 DOI: 10.1096/fj.202100350rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/11/2022]
Abstract
Myopia (short-sightedness), usually caused by excessive elongation of the eye during development, has reached epidemic proportions worldwide. In animal systems including the chicken model, several treatments have been shown to inhibit ocular elongation and experimental myopia. Although diverse in their apparent mechanism of action, each one leads to a reduction in the rate of ocular growth. We hypothesize that a defined set of retinal molecular changes may underlie growth inhibition, irrespective of the treatment agent used. Accordingly, across five well-established but diverse methods of inhibiting myopia, significant overlap is seen in the retinal transcriptome profile (transcript levels and alternative splicing events) in chicks when analyzed by RNA-seq. Within the two major pathway networks enriched during growth inhibition, that of cell signaling and circadian entrainment, transcription factors form the largest functional grouping. Importantly, a large percentage of those genes forming the defined retinal response are downstream targets of the transcription factor EGR1 which itself shows a universal response to all five growth-inhibitory treatments. This supports EGR1's previously implicated role in ocular growth regulation. Finally, by contrasting our data with human linkage and GWAS studies on refractive error, we confirm the applicability of our study to the human condition. Together, these findings suggest that a universal set of transcriptome changes, which sit within a well-defined retinal network that cannot be bypassed, is fundamental to growth regulation, thus paving a way for designing novel targets for myopia therapies.
Collapse
Affiliation(s)
- Cindy Karouta
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Robert Kucharski
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.,Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Kristine Hardy
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Kate Thomson
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Ryszard Maleszka
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Ian Morgan
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Regan Ashby
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.,Research School of Biology, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
14
|
Riddell N, Crewther SG, Murphy MJ, Tani Y. Long-Wavelength-Filtered Light Transiently Inhibits Negative Lens-Induced Axial Eye Growth in the Chick Myopia Model. Transl Vis Sci Technol 2021; 10:38. [PMID: 34459859 PMCID: PMC8411858 DOI: 10.1167/tvst.10.9.38] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Eye growth and myopia development in chicks, and some other animal models, can be suppressed by rearing under near-monochromatic, short-wavelength blue light. We aimed to determine whether similar effects could be achieved using glass filters that transmit a broader range of short and middle wavelengths. Methods On day 6 or 7 post-hatch, 169 chicks were assigned to one of three monocular lens conditions (−10 D, +10 D, plano) and reared for 7 or 10 days under one of four 201-lux lighting conditions: (1) B410 long-wavelength–filtered light, (2) B460 long-wavelength–filtered light, (3) Y48 short-wavelength–filtered light, or (4) HA50 broadband light. Results At 7 days, B410 (but not B460) long-wavelength–filtered light had significantly inhibited negative lens induced axial growth relative to Y48 short-wavelength–filtered light (mean difference in experimental eye = −0.249 mm; P = 0.006) and HA50 broadband light (mean difference = −0.139 mm; P = 0.038). B410 filters also inhibited the negative lens-induced increase in vitreous chamber depth relative to all other filter conditions. Corresponding changes in refraction did not occur, and biometric measurements in a separate cohort of chicks suggested that the axial dimension changes were transient and not maintained at 10 days. Conclusions Chromatic effects on eye growth can be achieved using filters that transmit a broad range of wavelengths even in the presence of strong cues for myopia development. Translational Relevance Broad-wavelength filters that provide a more “naturalistic” visual experience relative to monochromatic light have potential to alter myopia development, although the effects shown here were modest and transient and require exploration in further species.
Collapse
Affiliation(s)
- Nina Riddell
- Department of Psychology and Counselling, La Trobe University, Melbourne, Australia
| | - Sheila G Crewther
- Department of Psychology and Counselling, La Trobe University, Melbourne, Australia
| | - Melanie J Murphy
- Department of Psychology and Counselling, La Trobe University, Melbourne, Australia
| | - Yuki Tani
- Technical Research & Development Department, Vision Care Section, HOYA Corporation, Tokyo, Japan
| |
Collapse
|
15
|
Summers JA, Schaeffel F, Marcos S, Wu H, Tkatchenko AV. Functional integration of eye tissues and refractive eye development: Mechanisms and pathways. Exp Eye Res 2021; 209:108693. [PMID: 34228967 PMCID: PMC11697408 DOI: 10.1016/j.exer.2021.108693] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/16/2022]
Abstract
Refractive eye development is a tightly coordinated developmental process. The general layout of the eye and its various components are established during embryonic development, which involves a complex cross-tissue signaling. The eye then undergoes a refinement process during the postnatal emmetropization process, which relies heavily on the integration of environmental and genetic factors and is controlled by an elaborate genetic network. This genetic network encodes a multilayered signaling cascade, which converts visual stimuli into molecular signals that guide the postnatal growth of the eye. The signaling cascade underlying refractive eye development spans across all ocular tissues and comprises multiple signaling pathways. Notably, tissue-tissue interaction plays a key role in both embryonic eye development and postnatal eye emmetropization. Recent advances in eye biometry, physiological optics and systems genetics of refractive error have significantly advanced our understanding of the biological processes involved in refractive eye development and provided a framework for the development of new treatment options for myopia. In this review, we summarize the recent data on the mechanisms and signaling pathways underlying refractive eye development and discuss new evidence suggesting a wide-spread signal integration across different tissues and ocular components involved in visually guided eye growth.
Collapse
Affiliation(s)
- Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany; Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
| | - Susana Marcos
- Instituto de Óptica "Daza de Valdés", Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Hao Wu
- Department of Ophthalmology, Columbia University, New York, USA
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, USA; Department of Pathology and Cell Biology, Columbia University, New York, USA.
| |
Collapse
|
16
|
Tkatchenko TV, Tkatchenko AV. Genetic network regulating visual acuity makes limited contribution to visually guided eye emmetropization. Genomics 2021; 113:2780-2792. [PMID: 34147636 DOI: 10.1016/j.ygeno.2021.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/25/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
During postnatal development, the eye undergoes a refinement process whereby optical defocus guides eye growth towards sharp vision in a process of emmetropization. Optical defocus activates a signaling cascade originating in the retina and propagating across the back of the eye to the sclera. Several observations suggest that visual acuity might be important for optical defocus detection and processing in the retina; however, direct experimental evidence supporting or refuting the role of visual acuity in refractive eye development is lacking. Here, we used genome-wide transcriptomics to determine the relative contribution of the retinal genetic network regulating visual acuity to the signaling cascade underlying visually guided eye emmetropization. Our results provide evidence that visual acuity is regulated at the level of molecular signaling in the retina by an extensive genetic network. The genetic network regulating visual acuity makes relatively small contribution to the signaling cascade underlying refractive eye development. This genetic network primarily affects baseline refractive eye development and this influence is primarily facilitated by the biological processes related to melatonin signaling, nitric oxide signaling, phototransduction, synaptic transmission, and dopamine signaling. We also observed that the visual-acuity-related genes associated with the development of human myopia are chiefly involved in light perception and phototransduction. Our results suggest that the visual-acuity-related genetic network primarily contributes to the signaling underlying baseline refractive eye development, whereas its impact on visually guided eye emmetropization is modest.
Collapse
Affiliation(s)
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
17
|
Tkatchenko TV, Tkatchenko AV. Genome-wide analysis of retinal transcriptome reveals common genetic network underlying perception of contrast and optical defocus detection. BMC Med Genomics 2021; 14:153. [PMID: 34107987 PMCID: PMC8190860 DOI: 10.1186/s12920-021-01005-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Refractive eye development is regulated by optical defocus in a process of emmetropization. Excessive exposure to negative optical defocus often leads to the development of myopia. However, it is still largely unknown how optical defocus is detected by the retina. METHODS Here, we used genome-wide RNA-sequencing to conduct analysis of the retinal gene expression network underlying contrast perception and refractive eye development. RESULTS We report that the genetic network subserving contrast perception plays an important role in optical defocus detection and emmetropization. Our results demonstrate an interaction between contrast perception, the retinal circadian clock pathway and the signaling pathway underlying optical defocus detection. We also observe that the relative majority of genes causing human myopia are involved in the processing of optical defocus. CONCLUSIONS Together, our results support the hypothesis that optical defocus is perceived by the retina using contrast as a proxy and provide new insights into molecular signaling underlying refractive eye development.
Collapse
Affiliation(s)
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY USA
- Edward S. Harkness Eye Institute, Research Annex Room 415, 635 W. 165th Street, New York, NY 10032 USA
| |
Collapse
|
18
|
Riddell N, Murphy MJ, Crewther SG. Electroretinography and Gene Expression Measures Implicate Phototransduction and Metabolic Shifts in Chick Myopia and Hyperopia Models. Life (Basel) 2021; 11:life11060501. [PMID: 34072440 PMCID: PMC8228081 DOI: 10.3390/life11060501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 12/26/2022] Open
Abstract
The Retinal Ion-Driven Fluid Efflux (RIDE) model theorizes that phototransduction-driven changes in trans-retinal ion and fluid transport underlie the development of myopia (short-sightedness). In support of this model, previous functional studies have identified the attenuation of outer retinal contributions to the global flash electroretinogram (gfERG) following weeks of myopia induction in chicks, while discovery-driven transcriptome studies have identified changes to the expression of ATP-driven ion transport and mitochondrial metabolism genes in the retina/RPE/choroid at the mid- to late-induction time-points. Less is known about the early time-points despite biometric analyses demonstrating changes in eye growth by 3 h in the chick lens defocus model. Thus, the present study compared gfERG and transcriptome profiles between 3 h and 3 days of negative lens-induced myopia and positive lens-induced hyperopia in chicks. Photoreceptor (a-wave and d-wave) and bipolar (b-wave and late-stage d-wave) cell responses were suppressed following negative lens-wear, particularly at the 3–4 h and 3-day time-points when active shifts in the rate of ocular growth were expected. Transcriptome measures revealed the up-regulation of oxidative phosphorylation genes following 6 h of negative lens-wear, concordant with previous reports at 2 days in this model. Signal transduction pathways, with core genes involved in glutamate and G-protein coupled receptor signalling, were down-regulated at 6 h. These findings contribute to a growing body of evidence for the dysregulation of phototransduction and mitochondrial metabolism in animal models of myopia.
Collapse
|
19
|
RNA-seq and GSEA identifies suppression of ligand-gated chloride efflux channels as the major gene pathway contributing to form deprivation myopia. Sci Rep 2021; 11:5280. [PMID: 33674625 PMCID: PMC7935918 DOI: 10.1038/s41598-021-84338-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Currently there is no consensus regarding the aetiology of the excessive ocular volume that characterizes high myopia. Thus, we aimed to test whether the gene pathways identified by gene set enrichment analysis of RNA-seq transcriptomics refutes the predictions of the Retinal Ion Driven Efflux (RIDE) hypothesis when applied to the induction of form-deprivation myopia (FDM) and subsequent recovery (post-occluder removal). We found that the induction of profound FDM led to significant suppression in the ligand-gated chloride ion channel transport pathway via suppression of glycine, GABAA and GABAC ionotropic receptors. Post-occluder removal for short term recovery from FDM of 6 h and 24 h, induced significant upregulation of the gene families linked to cone receptor phototransduction, mitochondrial energy, and complement pathways. These findings support a model of form deprivation myopia as a Cl− ion driven adaptive fluid response to the modulation of the visual signal cascade by form deprivation that in turn affects the resultant ionic environment of the outer and inner retinal tissues, axial and vitreal elongation as predicted by the RIDE model. Occluder removal and return to normal light conditions led to return to more normal upregulation of phototransduction, slowed growth rate, refractive recovery and apparent return towards physiological homeostasis.
Collapse
|
20
|
Mérida S, Villar VM, Navea A, Desco C, Sancho-Tello M, Peris C, Bosch-Morell F. Imbalance Between Oxidative Stress and Growth Factors in Human High Myopia. Front Physiol 2020; 11:463. [PMID: 32477165 PMCID: PMC7240122 DOI: 10.3389/fphys.2020.00463] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Myopia is one of the commonest eye pathologies that could affect 2.56 billion people by 2020. Today high myopia is a leading cause of blindness worldwide due to associated ocular illness. Nevertheless, the cellular bases for these diseases to develop are unclear in many areas. We conducted a prospective study of oxidative stress and growth factors in human myopic and non myopic eyes in an attempt to increase our understanding of the underlying physiopathological conditions to adequately early diagnose, prevent and treat the retina problem that derives from myopia. Aqueous humor samples were obtained from 41 patients being operated for cataracts in our hospital. Axial length, refractive status and complete ophthalmologic examination were recorded. The VEGF and HGF levels were determined by an ELISA kit. Total antioxidant capacity and total nitrites/nitrate levels were established with a lab kit. We show for the first time an increase in the total nitrite levels in high myopia. We also propose for the first time the concurrence of three factors: myopia, oxidative stress, and oxidative stress together with growth factors in the same group of patients. In this way, it would not be accurate to envision high myopia as a type of normal myopia, but one with more diopters or longer axial length.
Collapse
Affiliation(s)
- Salvador Mérida
- Departamento de Ciencias Biomédicas, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Vincent M Villar
- Departamento de Ciencias Biomédicas, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Amparo Navea
- Departamento de Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Carmen Desco
- Departamento de Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain.,Department of Medical Ophtalmology, Fundación para el Fomento de la Investigación Sanitaria y Biomédica (FISABIO) de la Comunitat Valenciana, Valencia, Spain
| | | | - Cristina Peris
- Department of Medical Ophtalmology, Fundación para el Fomento de la Investigación Sanitaria y Biomédica (FISABIO) de la Comunitat Valenciana, Valencia, Spain
| | - Francisco Bosch-Morell
- Departamento de Ciencias Biomédicas, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain.,Department of Medical Ophtalmology, Fundación para el Fomento de la Investigación Sanitaria y Biomédica (FISABIO) de la Comunitat Valenciana, Valencia, Spain
| |
Collapse
|
21
|
Stone RA, Wei W, Sarfare S, McGeehan B, Engelhart KC, Khurana TS, Maguire MG, Iuvone PM, Nickla DL. Visual Image Quality Impacts Circadian Rhythm-Related Gene Expression in Retina and in Choroid: A Potential Mechanism for Ametropias. Invest Ophthalmol Vis Sci 2020; 61:13. [PMID: 32396635 PMCID: PMC7405616 DOI: 10.1167/iovs.61.5.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/21/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Stimulated by evidence implicating diurnal/circadian rhythms and light in refractive development, we studied the expression over 24 hours of selected clock and circadian rhythm-related genes in retina/retinal pigment epithelium (RPE) and choroid of experimental ametropias in chicks. Methods Newly hatched chicks, entrained to a 12-hour light/dark cycle for 12 to 14 days, either experienced nonrestricted vision OU (i.e., in both eyes) or received an image-blurring diffuser or a minus 10-diopter (D) or a plus 10-D defocusing lens over one eye. Starting 1 day later and at 4-hour intervals for 24 hours, the retina/RPE and choroid were separately dissected. Without pooling, total RNA was extracted, converted to cDNA, and assayed by quantitative PCR for the expression of the following genes: Opn4m, Clock, Npas2, Per3, Cry1, Arntl, and Mtnr1a. Results The expression of each gene in retina/RPE and in choroid of eyes with nonrestricted vision OU varied over 24 hours, with equal levels OU for most genes and times. Altered visual input influenced gene expression in complex patterns that varied by gene, visual input, time, and eye, affecting experimental eyes with altered vision and also contralateral eyes with nonrestricted vision. Discussion Altering visual input in ways known to induce ametropias alters the retinal/RPE and choroidal expression of circadian rhythm-related genes, further linking circadian biology with eye growth regulation. While further investigations are needed, studying circadian processes may help understand refractive mechanisms and the increasing myopia prevalence in contemporary societies where lighting patterns can desynchronize endogenous rhythms from the natural environmental light/dark cycle.
Collapse
Affiliation(s)
- Richard A. Stone
- Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Wenjie Wei
- Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Shanta Sarfare
- Department of Bioscience, New England College of Optometry, Boston, Massachusetts, United States
| | - Brendan McGeehan
- Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - K. Cameron Engelhart
- Department of Bioscience, New England College of Optometry, Boston, Massachusetts, United States
| | - Tejvir S. Khurana
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Maureen G. Maguire
- Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - P. Michael Iuvone
- Departments of Ophthalmology and Pharmacology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Debora L. Nickla
- Department of Bioscience, New England College of Optometry, Boston, Massachusetts, United States
| |
Collapse
|
22
|
Yu FJ, Lam TC, Sze AYH, Li KK, Chun RKM, Shan SW, To CH. Alteration of retinal metabolism and oxidative stress may implicate myopic eye growth: Evidence from discovery and targeted proteomics in an animal model. J Proteomics 2020; 221:103684. [PMID: 32061809 DOI: 10.1016/j.jprot.2020.103684] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/01/2020] [Accepted: 02/08/2020] [Indexed: 12/15/2022]
Abstract
Myopia, the most common cause of impaired vision, may induce sight- threatening diseases or ocular complications due to axial elongation. The exact mechanisms underlying myopia development have received much attention and understanding of these is necessary for clinical prevention or therapeutics. In this study, quantitative proteomics using Isotope Coded Protein Label (ICPL) was applied to identify differentially regulated proteins in the retinas of myopic chicks and, from their presence, infer the possible pathogenesis of excessive ocular elongation. Newly hatched white leghorn chicks (n = 15) wore -10D and + 10D lenses bilaterally for 3 and 7 days, respectively, to develop progressive lens-induced myopia (LIM) and hyperopia (LIH). Retinal proteins were quantified with nano-liquid chromatography electrospray ionization coupled with tandem mass spectrometry (nanoLC-ESI-MS/MS). Bioinformatics analysis of differentially regulated proteins revealed that the majority originated from the cytoplasmic region and were related to various metabolic, glycolytic, or oxidative processes. The fold changes of four proteins of interest (vimentin, apolipoprotein A1, interphotoreceptor retinoid binding protein, and glutathione S-transferase) were further confirmed by a novel high-resolution multiple reaction monitoring mass spectrometry (MRM-HR) using a label-free approach. SIGNIFICANCE: Discovery of effective protein biomarkers of myopia has been extensively studied to inhibit myopia progression. This study first applied lens-induced hyperopia and myopia in the same chick to maximize the inter-ocular differences, aiming to discover more protein biomarker candidates. The findings provided new evidence that myopia was metabolism related, accompanied by altered energy generation and oxidative stress at retinal protein levels. The results in the retina were also compared to our previous study in vitreous using ICPL quantitative technology. We have now presented the protein changes in these two adjacent tissues, which may provide extra information of protein changes during ocular growth in myopia.
Collapse
Affiliation(s)
- Feng-Juan Yu
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Thomas Chuen Lam
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| | - Andes Ying-Hon Sze
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - King-Kit Li
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Rachel Ka-Man Chun
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Sze-Wan Shan
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Chi-Ho To
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| |
Collapse
|
23
|
Tkatchenko TV, Tkatchenko AV. Pharmacogenomic Approach to Antimyopia Drug Development: Pathways Lead the Way. Trends Pharmacol Sci 2019; 40:833-852. [PMID: 31676152 DOI: 10.1016/j.tips.2019.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 12/29/2022]
Abstract
Myopia is the most common eye disorder in the world which is caused by a mismatch between the optical power of the eye and its excessively long axial length. Recent studies revealed that the regulation of the axial length of the eye occurs via a complex signaling cascade, which originates in the retina and propagates across all ocular tissues to the sclera. The complexity of this regulatory cascade has made it particularly difficult to develop effective antimyopia drugs. The current pharmacological treatment options for myopia are limited to atropine and 7-methylxanthine, which have either significant adverse effects or low efficacy. In this review, we focus on the recent advances in genome-wide studies of the signaling pathways underlying myopia development and discuss the potential of systems genetics and pharmacogenomic approaches for the development of antimyopia drugs.
Collapse
Affiliation(s)
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
24
|
Tkatchenko TV, Shah RL, Nagasaki T, Tkatchenko AV. Analysis of genetic networks regulating refractive eye development in collaborative cross progenitor strain mice reveals new genes and pathways underlying human myopia. BMC Med Genomics 2019; 12:113. [PMID: 31362747 PMCID: PMC6668126 DOI: 10.1186/s12920-019-0560-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Population studies suggest that genetic factors play an important role in refractive error development; however, the precise role of genetic background and the composition of the signaling pathways underlying refractive eye development remain poorly understood. METHODS Here, we analyzed normal refractive development and susceptibility to form-deprivation myopia in the eight progenitor mouse strains of the Collaborative Cross (CC). We used RNA-seq to analyze gene expression in the retinae of these mice and reconstruct genetic networks and signaling pathways underlying refractive eye development. We also utilized genome-wide gene-based association analysis to identify mouse genes and pathways associated with myopia in humans. RESULTS Genetic background strongly influenced both baseline refractive development and susceptibility to environmentally-induced myopia. Baseline refractive errors ranged from - 21.2 diopters (D) in 129S1/svlmj mice to + 22.0 D in CAST/EiJ mice and represented a continuous distribution typical of a quantitative genetic trait. The extent of induced form-deprivation myopia ranged from - 5.6 D in NZO/HILtJ mice to - 20.0 D in CAST/EiJ mice and also followed a continuous distribution. Whole-genome (RNA-seq) gene expression profiling in retinae from CC progenitor strains identified genes whose expression level correlated with either baseline refractive error or susceptibility to myopia. Expression levels of 2,302 genes correlated with the baseline refractive state of the eye, whereas 1,917 genes correlated with susceptibility to induced myopia. Genome-wide gene-based association analysis in the CREAM and UK Biobank human cohorts revealed that 985 of the above genes were associated with myopia in humans, including 847 genes which were implicated in the development of human myopia for the first time. Although the gene sets controlling baseline refractive development and those regulating susceptibility to myopia overlapped, these two processes appeared to be controlled by largely distinct sets of genes. CONCLUSIONS Comparison with data for other animal models of myopia revealed that the genes identified in this study comprise a well-defined set of retinal signaling pathways, which are highly conserved across different vertebrate species. These results identify major signaling pathways involved in refractive eye development and provide attractive targets for the development of anti-myopia drugs.
Collapse
Affiliation(s)
| | - Rupal L. Shah
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, UK
| | | | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY USA
| |
Collapse
|
25
|
Zhang Y, Phan E, Wildsoet CF. Retinal Defocus and Form-Deprivation Exposure Duration Affects RPE BMP Gene Expression. Sci Rep 2019; 9:7332. [PMID: 31089149 PMCID: PMC6517395 DOI: 10.1038/s41598-019-43574-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/23/2019] [Indexed: 11/09/2022] Open
Abstract
In the context of ocular development and eye growth regulation, retinal defocus and/or image contrast appear key variables although the nature of the signal(s) relayed from the retina to the sclera remains poorly understood. Nonetheless, under optimal visual conditions, eye length is brought into alignment with its optical power to achieve approximate emmetropia, through appropriate adjustment to eye growth. The retinal pigment epithelium (RPE), which lies between the retina and choroid/sclera, appears to play a crucial role in this process. In the investigations reported here, we used a chick model system to assess the threshold duration of exposure to lens-imposed defocus and form-deprivation necessary for conversion of evoked retinal signals into changes in BMP gene expression in the RPE. Our study provides evidence for the following: 1) close-loop, optical defocus-guided (negative and positive lenses) bidirectional BMP gene expression regulation, 2) open-loop, form-deprivation (diffusers)-induced down-regulation of BMP gene expression, and 3) early, transient up-regulation of BMP gene expression in response to both types of lens and diffuser applications. The critical exposure for accurately encoding retinal images as biological signals at the level of the RPE is in the order of minutes to hours, depending on the nature of the visual manipulations.
Collapse
Affiliation(s)
- Yan Zhang
- School of Optometry, University of California, Berkeley, Berkeley, CA, USA.
| | - Eileen Phan
- School of Optometry, University of California, Berkeley, Berkeley, CA, USA
| | | |
Collapse
|
26
|
Troilo D, Smith EL, Nickla DL, Ashby R, Tkatchenko AV, Ostrin LA, Gawne TJ, Pardue MT, Summers JA, Kee CS, Schroedl F, Wahl S, Jones L. IMI - Report on Experimental Models of Emmetropization and Myopia. Invest Ophthalmol Vis Sci 2019; 60:M31-M88. [PMID: 30817827 PMCID: PMC6738517 DOI: 10.1167/iovs.18-25967] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 10/20/2018] [Indexed: 11/24/2022] Open
Abstract
The results of many studies in a variety of species have significantly advanced our understanding of the role of visual experience and the mechanisms of postnatal eye growth, and the development of myopia. This paper surveys and reviews the major contributions that experimental studies using animal models have made to our thinking about emmetropization and development of myopia. These studies established important concepts informing our knowledge of the visual regulation of eye growth and refractive development and have transformed treatment strategies for myopia. Several major findings have come from studies of experimental animal models. These include the eye's ability to detect the sign of retinal defocus and undergo compensatory growth, the local retinal control of eye growth, regulatory changes in choroidal thickness, and the identification of components in the biochemistry of eye growth leading to the characterization of signal cascades regulating eye growth and refractive state. Several of these findings provided the proofs of concepts that form the scientific basis of new and effective clinical treatments for controlling myopia progression in humans. Experimental animal models continue to provide new insights into the cellular and molecular mechanisms of eye growth control, including the identification of potential new targets for drug development and future treatments needed to stem the increasing prevalence of myopia and the vision-threatening conditions associated with this disease.
Collapse
Affiliation(s)
- David Troilo
- SUNY College of Optometry, State University of New York, New York, New York, United States
| | - Earl L. Smith
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Debora L. Nickla
- Biomedical Sciences and Disease, New England College of Optometry, Boston, Massachusetts, United States
| | - Regan Ashby
- Health Research Institute, University of Canberra, Canberra, Australia
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Department of Pathology and Cell Biology, Columbia University, New York, New York, United States
| | - Lisa A. Ostrin
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Timothy J. Gawne
- School of Optometry, University of Alabama Birmingham, Birmingham, Alabama, United States
| | - Machelle T. Pardue
- Biomedical Engineering, Georgia Tech College of Engineering, Atlanta, Georgia, United States31
| | - Jody A. Summers
- College of Medicine, University of Oklahoma, Oklahoma City, Oklahoma, United States
| | - Chea-su Kee
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Falk Schroedl
- Departments of Ophthalmology and Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Siegfried Wahl
- Institute for Ophthalmic Research, University of Tuebingen, Zeiss Vision Science Laboratory, Tuebingen, Germany
| | - Lyndon Jones
- CORE, School of Optometry and Vision Science, University of Waterloo, Ontario, Canada
| |
Collapse
|
27
|
Tkatchenko TV, Troilo D, Benavente-Perez A, Tkatchenko AV. Gene expression in response to optical defocus of opposite signs reveals bidirectional mechanism of visually guided eye growth. PLoS Biol 2018; 16:e2006021. [PMID: 30300342 PMCID: PMC6177118 DOI: 10.1371/journal.pbio.2006021] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/30/2018] [Indexed: 01/14/2023] Open
Abstract
Myopia (nearsightedness) is the most common eye disorder, which is rapidly becoming one of the leading causes of vision loss in several parts of the world because of a recent sharp increase in prevalence. Nearwork, which produces hyperopic optical defocus on the retina, has been implicated as one of the environmental risk factors causing myopia in humans. Experimental studies have shown that hyperopic defocus imposed by negative power lenses placed in front of the eye accelerates eye growth and causes myopia, whereas myopic defocus imposed by positive lenses slows eye growth and produces a compensatory hyperopic shift in refractive state. The balance between these two optical signals is thought to regulate refractive eye development; however, the ability of the retina to recognize the sign of optical defocus and the composition of molecular signaling pathways guiding emmetropization are the subjects of intense investigation and debate. We found that the retina can readily distinguish between imposed myopic and hyperopic defocus, and identified key signaling pathways underlying retinal response to the defocus of different signs. Comparison of retinal transcriptomes in common marmosets exposed to either myopic or hyperopic defocus for 10 days or 5 weeks revealed that the primate retina responds to defocus of different signs by activation or suppression of largely distinct pathways. We also found that 29 genes differentially expressed in the marmoset retina in response to imposed defocus are localized within human myopia quantitative trait loci (QTLs), suggesting functional overlap between genes differentially expressed in the marmoset retina upon exposure to optical defocus and genes causing myopia in humans. These findings identify retinal pathways involved in the development of myopia, as well as potential new strategies for its treatment. The worldwide prevalence of myopia is predicted to increase from the current 23% to about 50% in the next three decades. Although much effort has been directed towards elucidating the mechanisms underlying refractive eye development and myopia, treatment options for myopia are mostly limited to optical correction, which does not prevent progression of myopia nor the pathological blinding complications often associated with the disease. Several experimental optics-based treatments have had only limited effect on myopia progression, and currently available drug treatments are limited and the mechanisms of action are not well understood. The development of safe and effective pharmacological treatments for myopia is urgently needed to prevent the impending myopia epidemic. The main obstacles that prevent the development of anti-myopia drugs are the uncertainties regarding the mechanisms controlling eye growth and optical development, including the molecular signaling pathways underlying it. In this study, we show that, contrary to the conventional thinking that myopic and hyperopic defocus trigger opposite changes in the same genes and pathways to guide postnatal eye growth, defocus of opposite signs affect eye growth via largely distinct retinal pathways. Knowing that myopic and hyperopic defocus signals drive eye growth in opposite directions and propagate via different pathways provides a framework for the development of new anti-myopia drugs. Myopia can be controlled pharmacologically by stimulating pathways underlying the retinal response to positive lenses and/or by suppressing pathways underlying the retinal response to negative lenses.
Collapse
Affiliation(s)
- Tatiana V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, New York, United States of America
| | - David Troilo
- College of Optometry, State University of New York, New York, New York, United States of America
| | | | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
28
|
Riddell N, Faou P, Crewther SG. Short term optical defocus perturbs normal developmental shifts in retina/RPE protein abundance. BMC DEVELOPMENTAL BIOLOGY 2018; 18:18. [PMID: 30157773 PMCID: PMC6116556 DOI: 10.1186/s12861-018-0177-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 08/16/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Myopia (short-sightedness) affects approximately 1.4 billion people worldwide, and prevalence is increasing. Animal models induced by defocusing lenses show striking similarity with human myopia in terms of morphology and the implicated genetic pathways. Less is known about proteome changes in animals. Thus, the present study aimed to improve understanding of protein pathway responses to lens defocus, with an emphasis on relating expression changes to no lens control development and identifying bidirectional and/or distinct pathways across myopia and hyperopia (long-sightedness) models. RESULTS Quantitative label-free proteomics and gene set enrichment analysis (GSEA) were used to examine protein pathway expression in the retina/RPE of chicks following 6 h and 48 h of myopia induction with - 10 dioptre (D) lenses, hyperopia induction with +10D lenses, or normal no lens rearing. Seventy-one pathways linked to cell development and neuronal maturation were differentially enriched between 6 and 48 h in no lens chicks. The majority of these normal developmental changes were disrupted by lens-wear (47 of 71 pathways), however, only 11 pathways displayed distinct expression profiles across the lens conditions. Most notably, negative lens-wear induced up-regulation of proteins involved in ATP-driven ion transport, calcium homeostasis, and GABA signalling between 6 and 48 h, while the same proteins were down-regulated over time in normally developing chicks. Glutamate and bicarbonate/chloride transporters were also down-regulated over time in normally developing chicks, and positive lens-wear inhibited this down-regulation. CONCLUSIONS The chick retina/RPE proteome undergoes extensive pathway expression shifts during normal development. Most of these pathways are further disrupted by lens-wear. The identified expression patterns suggest close interactions between neurotransmission (as exemplified by increased GABA receptor and synaptic protein expression), cellular ion homeostasis, and associated energy resources during myopia induction. We have also provided novel evidence for changes to SLC-mediated transmembrane transport during hyperopia induction, with potential implications for signalling at the photoreceptor-bipolar synapse. These findings reflect a key role for perturbed neurotransmission and ionic homeostasis in optically-induced refractive errors, and are predicted by our Retinal Ion Driven Efflux (RIDE) model.
Collapse
Affiliation(s)
- Nina Riddell
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Plenty Rd., Bundoora, Melbourne, VIC, 3083, Australia.
| | - Pierre Faou
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Sheila G Crewther
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Plenty Rd., Bundoora, Melbourne, VIC, 3083, Australia
| |
Collapse
|
29
|
Giummarra L, Crewther SG, Riddell N, Murphy MJ, Crewther DP. Pathway analysis identifies altered mitochondrial metabolism, neurotransmission, structural pathways and complement cascade in retina/RPE/ choroid in chick model of form-deprivation myopia. PeerJ 2018; 6:e5048. [PMID: 29967729 PMCID: PMC6026464 DOI: 10.7717/peerj.5048] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/31/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose RNA sequencing analysis has demonstrated bidirectional changes in metabolism, structural and immune pathways during early induction of defocus induced myopia. Thus, the aim of this study was to investigate whether similar gene pathways are also related to the more excessive axial growth, ultrastructural and elemental microanalytic changes seen during the induction and recovery from form-deprivation myopia (FDM) in chicks and predicted by the RIDE model of myopia. Methods Archived genomic transcriptome data from the first three days of induction of monocularly occluded form deprived myopia (FDMI) in chicks was obtained from the GEO database (accession # GSE6543) while data from chicks monocularly occluded for 10 days and then given up to 24 h of normal visual recovery (FDMR) were collected. Gene set enrichment analysis (GSEA) software was used to determine enriched pathways during the induction (FDMI) and recovery (FDMR) from FD. Curated gene-sets were obtained from open access sources. Results Clusters of significant changes in mitochondrial energy metabolism, neurotransmission, ion channel transport, G protein coupled receptor signalling, complement cascades and neuron structure and growth were identified during the 10 days of induction of profound myopia and were found to correlate well with change in axial dimensions. Bile acid and bile salt metabolism pathways (cholesterol/lipid metabolism and sodium channel activation) were significantly upregulated during the first 24 h of recovery from 10 days of FDM. Conclusions The gene pathways altered during induction of FDM are similar to those reported in defocus induced myopia and are established indicators of oxidative stress, osmoregulatory and associated structural changes. These findings are also consistent with the choroidal thinning, axial elongation and hyperosmotic ion distribution patterns across the retina and choroid previously reported in FDM and predicted by RIDE.
Collapse
Affiliation(s)
- Loretta Giummarra
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Sheila G Crewther
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Nina Riddell
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Melanie J Murphy
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - David P Crewther
- Centre for Psychopharmacology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
30
|
Chakraborty R, Ostrin LA, Nickla DL, Iuvone PM, Pardue MT, Stone RA. Circadian rhythms, refractive development, and myopia. Ophthalmic Physiol Opt 2018; 38:217-245. [PMID: 29691928 PMCID: PMC6038122 DOI: 10.1111/opo.12453] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/11/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Despite extensive research, mechanisms regulating postnatal eye growth and those responsible for ametropias are poorly understood. With the marked recent increases in myopia prevalence, robust and biologically-based clinical therapies to normalize refractive development in childhood are needed. Here, we review classic and contemporary literature about how circadian biology might provide clues to develop a framework to improve the understanding of myopia etiology, and possibly lead to rational approaches to ameliorate refractive errors developing in children. RECENT FINDINGS Increasing evidence implicates diurnal and circadian rhythms in eye growth and refractive error development. In both humans and animals, ocular length and other anatomical and physiological features of the eye undergo diurnal oscillations. Systemically, such rhythms are primarily generated by the 'master clock' in the surpachiasmatic nucleus, which receives input from the intrinsically photosensitive retinal ganglion cells (ipRGCs) through the activation of the photopigment melanopsin. The retina also has an endogenous circadian clock. In laboratory animals developing experimental myopia, oscillations of ocular parameters are perturbed. Retinal signaling is now believed to influence refractive development; dopamine, an important neurotransmitter found in the retina, not only entrains intrinsic retinal rhythms to the light:dark cycle, but it also modulates refractive development. Circadian clocks comprise a transcription/translation feedback control mechanism utilizing so-called clock genes that have now been associated with experimental ametropias. Contemporary clinical research is also reviving ideas first proposed in the nineteenth century that light exposures might impact refraction in children. As a result, properties of ambient lighting are being investigated in refractive development. In other areas of medical science, circadian dysregulation is now thought to impact many non-ocular disorders, likely because the patterns of modern artificial lighting exert adverse physiological effects on circadian pacemakers. How, or if, such modern light exposures and circadian dysregulation contribute to refractive development is not known. SUMMARY The premise of this review is that circadian biology could be a productive area worthy of increased investigation, which might lead to the improved understanding of refractive development and improved therapeutic interventions.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- College of Nursing and Health Sciences, Flinders University, Adelaide, Australia
| | | | | | | | - Machelle T. Pardue
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur
| | - Richard A. Stone
- University of Pennsylvania School of Medicine, Philadelphia, USA
| |
Collapse
|
31
|
Riddell N, Faou P, Murphy M, Giummarra L, Downs RA, Rajapaksha H, Crewther SG. The retina/RPE proteome in chick myopia and hyperopia models: Commonalities with inherited and age-related ocular pathologies. Mol Vis 2017; 23:872-888. [PMID: 29259393 PMCID: PMC5723150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 12/03/2017] [Indexed: 11/14/2022] Open
Abstract
Purpose Microarray and RNA sequencing studies in the chick model of early optically induced refractive error have implicated thousands of genes, many of which have also been linked to ocular pathologies in humans, including age-related macular degeneration (AMD), choroidal neovascularization, glaucoma, and cataract. These findings highlight the potential relevance of the chick model to understanding both refractive error development and the progression to secondary pathological complications. The present study aimed to determine whether proteomic responses to early optical defocus in the chick share similarities with these transcriptome-level changes, particularly in terms of dysregulation of pathology-related molecular processes. Methods Chicks were assigned to a lens condition (monocular +10 D [diopters] to induce hyperopia, -10 D to induce myopia, or no lens) on post-hatch day 5. Biometric measures were collected following a further 6 h and 48 h of rearing. The retina/RPE was then removed and prepared for liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) on an LTQ-Orbitrap Elite. Raw data were processed using MaxQuant, and differentially abundant proteins were identified using moderated t tests (fold change ≥1.5, Benjamini-Hochberg adjusted p<0.05). These differentially abundant proteins were compared with the genes and proteins implicated in previous exploratory transcriptome and proteomic studies of refractive error, as well as the genes and proteins linked to the ocular pathologies listed above for which myopia or hyperopia are risk factors. Finally, gene set enrichment analysis (GSEA) was used to assess whether gene sets from the Human Phenotype Ontology database were enriched in the lens groups relative to the no lens groups, and at the top or bottom of the protein data ranked by Spearman's correlation with refraction at 6 and 48 h. Results Refractive errors of -2.63 D ± 0.31 D (mean ± standard error, SE) and 3.90 D ± 0.37 D were evident in the negative and positive lens groups, respectively, at 6 h. By 48 h, refractive compensation to both lens types was almost complete (negative lens -9.70 D ± 0.41 D, positive lens 7.70 D ± 0.44 D). More than 140 differentially abundant proteins were identified in each lens group relative to the no lens controls at both time points. No proteins were differentially abundant between the negative and positive lens groups at 6 h, and 13 were differentially abundant at 48 h. As there was substantial overlap in the proteins implicated across the six comparisons, a total of 390 differentially abundant proteins were identified. Sixty-five of these 390 proteins had previously been implicated in transcriptome studies of refractive error animal models, and 42 had previously been associated with AMD, choroidal neovascularization, glaucoma, and/or cataract in humans. The overlap of differentially abundant proteins with AMD-associated genes and proteins was statistically significant for all conditions (Benjamini-Hochberg adjusted p<0.05), with over-representation analysis implicating ontologies related to oxidative stress, cholesterol homeostasis, and melanin biosynthesis. GSEA identified significant enrichment of genes associated with abnormal electroretinogram, photophobia, and nyctalopia phenotypes in the proteins negatively correlated with ocular refraction across the lens groups at 6 h. The implicated proteins were primarily linked to photoreceptor dystrophies and mitochondrial disorders in humans. Conclusions Optical defocus in the chicks induces rapid changes in the abundance of many proteins in the retina/RPE that have previously been linked to inherited and age-related ocular pathologies in humans. Similar changes have been identified in a meta-analysis of chick refractive error transcriptome studies, highlighting the chick as a model for the study of optically induced stress with possible relevance to understanding the development of a range of pathological states in humans.
Collapse
Affiliation(s)
- Nina Riddell
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Pierre Faou
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Melanie Murphy
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Loretta Giummarra
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Rachael A. Downs
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Harinda Rajapaksha
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Sheila G. Crewther
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
32
|
Yu FJ, Lam TC, Liu LQ, Chun RKM, Cheung JKW, Li KK, To CH. Isotope-coded protein label based quantitative proteomic analysis reveals significant up-regulation of apolipoprotein A1 and ovotransferrin in the myopic chick vitreous. Sci Rep 2017; 7:12649. [PMID: 28978931 PMCID: PMC5627271 DOI: 10.1038/s41598-017-12650-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/13/2017] [Indexed: 02/05/2023] Open
Abstract
This study used isotope-coded protein label (ICPL) quantitative proteomics and bioinformatics analysis to examine changes in vitreous protein content and associated pathways during lens-induced eye growth. First, the vitreous protein profile of normal 7-day old chicks was characterized by nano-liquid chromatography electrospray ionization tandem mass spectrometry. A total of 341 unique proteins were identified. Next, myopia and hyperopia were induced in the same chick by attaching -10D lenses to the right eye and +10D lenses to the left eye, for 3 and 7 days. Protein expression in lens-induced ametropic eyes was analyzed using the ICPL approach coupled to LCMS. Four proteins (cystatin, apolipoprotein A1, ovotransferrin, and purpurin) were significantly up-regulated in the vitreous after 3 days of wearing -10D lenses relative to +10D lens contralateral eyes. The differences in protein expression were less pronounced after 7 days when the eyes approached full compensation. In a different group of chicks, western blot confirmed the up-regulation of apolipoprotein A1 and ovotransferrin in the myopic vitreous relative to both contralateral lens-free eyes and hyperopic eyes in separate animals wearing +10D lenses. Bioinformatics analysis suggested oxidative stress and lipid metabolism as pathways involved in compensated ocular elongation.
Collapse
Affiliation(s)
- Feng-Juan Yu
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Thomas Chuen Lam
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| | - Long-Qian Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Rachel Ka-Man Chun
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Jimmy Ka-Wai Cheung
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - King-Kit Li
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Chi-Ho To
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
33
|
Riddell N, Crewther SG. Novel evidence for complement system activation in chick myopia and hyperopia models: a meta-analysis of transcriptome datasets. Sci Rep 2017; 7:9719. [PMID: 28852117 PMCID: PMC5574905 DOI: 10.1038/s41598-017-10277-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/21/2017] [Indexed: 12/27/2022] Open
Abstract
Myopia (short-sightedness) and hyperopia (long-sightedness) occur when the eye grows too long or short, respectively, for its refractive power. There are currently approximately 1.45 billion myopes worldwide and prevalence is rising dramatically. Although high myopia significantly increases the risk of developing a range of sight-threatening disorders, the molecular mechanisms underlying ocular growth regulation and its relationship to these secondary complications remain poorly understood. Thus, this study meta-analyzed transcriptome datasets collected in the commonly used chick model of optically-induced refractive error. Fifteen datasets (collected across five previous studies) were obtained from GEO, preprocessed in Bioconductor, and divided into 4 conditions representing early (≤1 day) and late (>1 day) myopia and hyperopia induction. Differentially expressed genes in each condition were then identified using Rank Product meta-analysis. The results provide novel evidence for transcriptional activation of the complement system during both myopia and hyperopia induction, and confirm existing literature implicating cell signaling, mitochondrial, and structural processes in refractive error. Further comparisons demonstrated that the meta-analysis results also significantly improve concordance with broader omics data types (i.e., human genetic association and animal proteomics studies) relative to previous transcriptome studies, and show extensive similarities with the genes linked to age-related macular degeneration, choroidal neovascularization, and cataract.
Collapse
Affiliation(s)
- Nina Riddell
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Sheila G Crewther
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, 3086, Australia.
| |
Collapse
|