1
|
Zhuang Z, Li L, Yu Y, Su X, Lin S, Hu J. Targeting MicroRNA in myopia: Current insights. Exp Eye Res 2024; 243:109905. [PMID: 38642599 DOI: 10.1016/j.exer.2024.109905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Myopia, the most prevalent eye condition, has sparked notable interest regarding its origin and prevention. MicroRNAs (miRNAs) are short, non-coding RNA strands typically consisting of 18-24 nucleotides. They play a central role in post-transcriptional gene regulation and are closely associated with both normal and pathological processes in organisms. Recent advances in next-generation sequencing and bioinformatics have provided novel insights into miRNA expression and its regulatory role in myopia. This review discusses the distinct expression patterns, regulatory functions, and potential pathways of miRNAs involved in the onset and progression of myopia. The primary objective of this review was to provide valuable insights into molecular mechanisms underlying myopia and the contribution of miRNAs. These insights are expected to pave the way for further exploration of the molecular mechanisms, diagnosis, treatment, and clinical applications of myopia.
Collapse
Affiliation(s)
- Zihao Zhuang
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian, China
| | - Licheng Li
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian, China
| | - Yang Yu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian, China
| | - Xuemei Su
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China; Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - Jianmin Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Centre of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, Fujian, China; The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
2
|
Hao J, Yang Z, Zhang R, Ma Z, Liu J, Bi H, Guo D. Crosstalk between heredity and environment in myopia: An overview. Heliyon 2024; 10:e29715. [PMID: 38660258 PMCID: PMC11040123 DOI: 10.1016/j.heliyon.2024.e29715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 03/04/2024] [Accepted: 04/14/2024] [Indexed: 04/26/2024] Open
Abstract
In recent years, the prevalence of myopia has gradually increased, and it has become a significant global public health problem in the 21st century, posing a serious challenge to human eye health. Currently, it is confirmed that the development of myopia is attributed to the combined action of genes and environmental factors. Thus, elucidating the risk factors and pathogenesis of myopia is of great significance for the prevention and control of myopia. To elucidate the impact of gene-environment interaction on myopia, we used the Pubmed database to search for literature related to myopia. Search terms are as follows: myopia, genes, environmental factors, gene-environment interaction, and treatment. This paper reviews the effects of gene and environmental interaction on myopia.
Collapse
Affiliation(s)
- Jiawen Hao
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Zhaohui Yang
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Ruixue Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Zhongyu Ma
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Jinpeng Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China
- Shandong Academy of Eye Disease Prevention and Therapy, Jinan, 250002, China
- Shandong Provincial Clinical Research Center of Ophthalmology and Children Visual Impairment Prevention and Control, Jinan, 250002, China
- Shandong Engineering Technology Research Center of Visual Intelligence, Jinan, 250002, China
- Shandong Academy of Health and Myopia Prevention and Control of Children and Adolescents, Jinan, 250002, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Dadong Guo
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China
- Shandong Academy of Eye Disease Prevention and Therapy, Jinan, 250002, China
- Shandong Provincial Clinical Research Center of Ophthalmology and Children Visual Impairment Prevention and Control, Jinan, 250002, China
- Shandong Engineering Technology Research Center of Visual Intelligence, Jinan, 250002, China
- Shandong Academy of Health and Myopia Prevention and Control of Children and Adolescents, Jinan, 250002, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| |
Collapse
|
3
|
Sbornova I, van der Sande E, Milosavljevic S, Amurrio E, Burbano SD, Das PK, Do HH, Fisher JL, Kargbo P, Patel J, Porcher L, De Zeeuw CI, Meester-Smoor MA, Winkelman BHJ, Klaver CCW, Pocivavsek A, Kelly MP. The Sleep Quality- and Myopia-Linked PDE11A-Y727C Variant Impacts Neural Physiology by Reducing Catalytic Activity and Altering Subcellular Compartmentalization of the Enzyme. Cells 2023; 12:2839. [PMID: 38132157 PMCID: PMC10742168 DOI: 10.3390/cells12242839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Recently, a Y727C variant in the dual-specific 3',5'-cyclic nucleotide phosphodiesterase 11A (PDE11A-Y727C) was linked to increased sleep quality and reduced myopia risk in humans. Given the well-established role that the PDE11 substrates cAMP and cGMP play in eye physiology and sleep, we determined if (1) PDE11A protein is expressed in the retina or other eye segments in mice, (2) PDE11A-Y7272C affects catalytic activity and/or subcellular compartmentalization more so than the nearby suicide-associated PDE11A-M878V variant, and (3) Pde11a deletion alters eye growth or sleep quality in male and female mice. Western blots show distinct protein expression of PDE11A4, but not PDE11A1-3, in eyes of Pde11a WT, but not KO mice, that vary by eye segment and age. In HT22 and COS-1 cells, PDE11A4-Y727C reduces PDE11A4 catalytic activity far more than PDE11A4-M878V, with both variants reducing PDE11A4-cAMP more so than PDE11A4-cGMP activity. Despite this, Pde11a deletion does not alter age-related changes in retinal or lens thickness or axial length, nor vitreous or anterior chamber depth. Further, Pde11a deletion only minimally changes refractive error and sleep quality. That said, both variants also dramatically alter the subcellular compartmentalization of human and mouse PDE11A4, an effect occurring independently of dephosphorylating PDE11A4-S117/S124 or phosphorylating PDE11A4-S162. Rather, re-compartmentalization of PDE11A4-Y727C is due to the loss of the tyrosine changing how PDE11A4 is packaged/repackaged via the trans-Golgi network. Therefore, the protective impact of the Y727C variant may reflect a gain-of-function (e.g., PDE11A4 displacing another PDE) that warrants further investigation in the context of reversing/preventing sleep disturbances or myopia.
Collapse
Affiliation(s)
- Irina Sbornova
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Emilie van der Sande
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, 1105 AZ Amsterdam, The Netherlands
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd., Columbia, SC 29209, USA
| | - Elvis Amurrio
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Steven D. Burbano
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Prosun K. Das
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Helen H. Do
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Janet L. Fisher
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd., Columbia, SC 29209, USA
| | - Porschderek Kargbo
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Janvi Patel
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Latarsha Porcher
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
| | - Chris I. De Zeeuw
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, 1105 AZ Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
| | - Magda A. Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
| | - Beerend H. J. Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, 1105 AZ Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, 3015 CN Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Mittlere Strasse 91, 4070 Basel, Switzerland
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd., Columbia, SC 29209, USA
| | - Michy P. Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA (P.K.D.); (J.P.)
- Center for Research on Aging, University of Maryland School of Medicine, 20 Penn St., Baltimore, MD 21201, USA
| |
Collapse
|
4
|
Sbornova I, van der Sande E, Milosavljevic S, Amurrio E, Burbano SD, Das P, Do H, Fisher JL, Kargbo P, Patel J, Porcher L, De Zeeuw CI, Meester-Smoor MA, Winkelman BH, Klaver CC, Pocivavsek A, Kelly MP. The sleep quality- and myopia-linked PDE11A-Y727C variant impacts neural physiology by reducing catalytic activity and altering subcellular compartmentalization of the enzyme. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567422. [PMID: 38014312 PMCID: PMC10680747 DOI: 10.1101/2023.11.16.567422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Recently, a Y727C variant in the dual-specific 3',5'-cyclic nucleotide phosphodiesterase 11A (PDE11A-Y727C) was linked to increased sleep quality and reduced myopia risk in humans. Given the well-established role that the PDE11 substrates cAMP and cGMP play in eye physiology and sleep, we determined if 1) PDE11A protein is expressed in the retina or other eye segments in mouse, 2) PDE11A-Y7272C affects catalytic activity and/or subcellular compartmentalization more so than the nearby suicide-associated PDE11A-M878V variant, and 3) Pde11a deletion alters eye growth or sleep quality in male and female mice. Western blots show distinct protein expression of PDE11A4, but not PDE11A1-3, in eyes of Pde11a WT-but not KO mice-that vary by eye segment and age. In HT22 and COS-1 cells, PDE11A4-Y727C reduces PDE11A4 catalytic activity far more than PDE11A4-M878V, with both variants reducing PDE11A4-cAMP more so than PDE11A4-cGMP activity. Despite this, Pde11a deletion does not alter age-related changes in retinal or lens thickness, axial length, nor vitreous or anterior chamber depth. Further, Pde11a deletion only minimally changes refractive error and sleep quality. That said, both variants also dramatically alter the subcellular compartmentalization of human and mouse PDE11A4, an effect occurring independently of dephosphorylating PDE11A4-S117/S124 or phosphorylating PDE11A4-S162. Rather, re-compartmentalization of PDE11A4-Y727C is due to the loss of the tyrosine changing how PDE11A4 is packaged/repackaged via the trans-Golgi network. Therefore, the protective impact of the Y727C variant may reflect a gain-of-function (e.g., PDE11A4 displacing another PDE) that warrants further investigation in the context of reversing/preventing sleep disturbances or myopia.
Collapse
Affiliation(s)
- Irina Sbornova
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Emilie van der Sande
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, Amsterdam, The Netherlands
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd, Columbia, SC
| | - Elvis Amurrio
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Steven D. Burbano
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Prosun Das
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Helen Do
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Janet L. Fisher
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd, Columbia, SC
| | - Porschderek Kargbo
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Janvi Patel
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Latarsha Porcher
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| | - Chris I. De Zeeuw
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
| | - Beerend H.J. Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Meibergdreef 47, Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
| | - Caroline C.W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Wytemaweg 40, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Mittlere Strasse 91, Basel, Switzerland
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Garners Ferry Rd, Columbia, SC
| | - Michy P. Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
- Center for Research on Aging, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201
| |
Collapse
|
5
|
He X, Lin C, Zhang F, Zhang S, Kang M, Wei S, Li H, Wang N, Li SM. Outdoor time influences VIPR2 polymorphism rs2071623 to regulate axial length in Han Chinese children. Mol Vis 2023; 29:266-273. [PMID: 38222453 PMCID: PMC10784227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 11/01/2023] [Indexed: 01/16/2024] Open
Abstract
Clinical relevance Identification of individuals with a higher risk of developing refractive error under specific gene and environmental backgrounds, especially myopia, could enable more personalized myopic control advice for patients. Background Refractive error is a common disease that affects visual quality and ocular health worldwide. Its mechanisms have not been elaborated, although both genes and the environment are known to contribute to the process. Interactions between genes and the environment have been shown to exert effects on the onset of refractive error, especially myopia. Axial length elongation is the main characteristic of myopia development and could indicate the severity of myopia. Thus, the purpose of the study was to investigate the interaction between environmental factors and genetic markers of VIPR2 and their impact on spherical equivalence and axial length in a population of Han Chinese children. Methods A total of 1825 children aged 13~15 years in the Anyang Childhood Eye Study (ACES) were measured for cycloplegic autorefraction, axial length, and height. Saliva DNA was extracted for genotyping three single-nucleotide polymorphisms (SNPs) in the candidate gene (VIPR2). The median outdoor time (2 h/day) was used to categorize children into high and low exposure groups, respectively. Genetic quality control and linear and logistic regressions were performed. Generalized multifactor dimensional reduction (GMDR) was used to investigate gene-environment interactions. Results There were 1391 children who passed genetic quality control. Rs2071623 of VIPR2 was associated with axial length (T allele, β=-0.11 se=0.04 p=0.006), while SNP nominally interacted with outdoor time (T allele, β=-0.17 se=0.08 p=0.029). Rs2071623 in children with high outdoor exposure had a significant interaction effect on axial length (p=0.0007, β=-0.19 se=0.056) compared to children with low outdoor exposure. GMDR further suggested the existence of an interaction effect between outdoor time and rs2071623. Conclusions Rs2071623 within VIPR2 could interact with outdoor time in Han Chinese children. More outdoor exposure could enhance the protective effect of the T allele on axial elongation.
Collapse
Affiliation(s)
- Xi He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Caixia Lin
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Fengchuan Zhang
- School of Mathematics Sciences, University of Chinese Academy of Science
| | - Sanguo Zhang
- School of Mathematics Sciences, University of Chinese Academy of Science
| | - Mengtian Kang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Shifei Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - He Li
- Anyang Eye Hospital, Henan Province, China
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Shi-Ming Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| |
Collapse
|
6
|
He X, Li SM. Gene-environment interaction in myopia. Ophthalmic Physiol Opt 2023; 43:1438-1448. [PMID: 37486033 DOI: 10.1111/opo.13206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
Myopia is a health issue that has attracted global attention due to its high prevalence and vision-threatening complications. It is well known that the onset and progression of myopia are related to both genetic and environmental factors: more than 450 common genetic loci have been found to be associated with myopia, while near work and outdoor time are the main environmental risk factors. As for many complex traits, gene-environment interactions are implicated in myopia development. To date, several genetic loci have been found to interact with near work or educational level. Gene-environment interaction research on myopia could yield models that provide more accurate risk predictions, thus improving targeted treatments and preventive strategies. Additionally, such investigations might have the potential to reveal novel genetic information. In this review, we summarised the findings in this field and proposed some topics for future investigations.
Collapse
Affiliation(s)
- Xi He
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| | - Shi-Ming Li
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| |
Collapse
|
7
|
Huang T, Wang Y, Wang Z, Long Q, Li Y, Chen D. Complement-mediated inflammation and mitochondrial energy metabolism in the proteomic profile of myopic human corneas. J Proteomics 2023; 285:104949. [PMID: 37331426 DOI: 10.1016/j.jprot.2023.104949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 06/20/2023]
Abstract
Myopia is one of the most common causes of visual impairment worldwide. To identify proteins related to myopiagenesis, data-independent acquisition proteomic analysis was performed using corneal lenticules of myopic patients who underwent small incision lenticule extraction surgery. A total of 19 lenticules from 19 age and sex-matched patients were analyzed, 10 in high refractive error (HR, spherical equivalent over -6.00 D) group and 9 in low refractive error (LR, spherical equivalent between -3.00 and - 1.00 D) group. Differentially expressed proteins (DEPs) were identified by comparing the corneal proteome between the two groups. Functional analyses were performed to explore the biological pathways and interactions of the DEPs. 107 DEPs (67 upregulated and 40 downregulated in HR group, compared to LR) were identified from 2138 quantified proteins. Functional analyses indicated that upregulated proteins were primarily involved in the complement pathways and extracellular matrix (ECM) remodeling, while downregulated proteins were involved in mitochondrial energy metabolism. Western blot analysis confirmed increased complement C3a and apolipoprotein E in HR samples, further supporting the proteomics data. In conclusion, this proteomic study reveals that proteins associated with the complement system, ECM remodeling, and mitochondrial energy metabolism might be key effectors in myopiagenesis. SIGNIFICANCE: Myopia has become one of the most prevalent causes of visual impairment, especially in Asia. The underlying mechanism of myopia development is still up for debate. This study compares the proteomic profiles of high and low myopic corneas, identifying differentially expressed proteins associated with the complement system, ECM remodeling, and mitochondrial energy metabolism. The findings of this study could provide novel insights into the pathogenesis of myopia. The complement system and mitochondrial energy metabolism may provide potential therapeutic targets in the treatment and prevention of myopia.
Collapse
Affiliation(s)
- Tianze Huang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Yuchen Wang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Zhonghai Wang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Qin Long
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Ying Li
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Di Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
8
|
Yi J, Dai Y, Ma S, Zheng Y, Liang Y, Huang X. Correlation between High Myopia Susceptibility and Polymorphisms of RASGRF1 Gene among College Students in Zhejiang. JOURNAL OF ENVIRONMENTAL AND PUBLIC HEALTH 2023; 2023:6767410. [PMID: 36864897 PMCID: PMC9974243 DOI: 10.1155/2023/6767410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/10/2022] [Accepted: 01/24/2023] [Indexed: 02/23/2023]
Abstract
Objective The aim of the study is to analyze the correlation between high myopia susceptibility and Ras protein-specific guanine nucleotide-releasing factor-1(RASGRF1) gene polymorphism among college students in Zhejiang. Methods A stratified whole-group sampling method was used to select 218 cases of college students in Zhejiang who met the inclusion and exclusion criteria from January, 2019, to December, 2021, and they were divided into 77 cases (154 eyes) in the high myopia group and 141 cases (282 eyes) in the medium-low myopia group according to the degree of myopia, and 109 cases of college volunteers without myopia from the same period of medical examination in the region were included in the control group. The single nucleotide polymorphisms (SNPs) located in functional regions were selected by searching the literature and genetic databases, and the base sequences of rs939658, rs4778879, and rs8033417 loci were obtained by genotyping candidate SNPs using multiplex ligase detection reaction technique. The cardinality test was used to compare the differences in genotype frequency distribution of each locus of the RASGRF1 gene between the high myopia group and the low to moderate myopia group and the control group. Results The genotype frequencies and allele frequencies of the RASGRF1 gene rs939658 locus in the high myopia group compared with the moderate-low myopia group and the control group were not statistically significant (P > 0.05). The genotype frequencies and allele frequencies of the rs4778879 locus of the RASGRF1 gene were compared among the three groups, and the differences were not statistically significant (P > 0.05). The genotype frequency and allele frequency of the rs8033417 locus of the RASGRF1 gene differed significantly among the three groups (P < 0.05). Conclusion The polymorphism of the rs8033417 locus of the RASGRF1 gene was significantly correlated with the susceptibility of high myopia among college students in Zhejiang.
Collapse
Affiliation(s)
- Jipan Yi
- Optometry Technology of Zhejiang Industry and Trade Vocational College, Wenzhou 325000, Zhejiang, China
| | - Yingying Dai
- Optometry Technology of Zhejiang Industry and Trade Vocational College, Wenzhou 325000, Zhejiang, China
| | - Shangsheng Ma
- Optometry Technology of Zhejiang Industry and Trade Vocational College, Wenzhou 325000, Zhejiang, China
| | - Yiyi Zheng
- Optometry Technology of Zhejiang Industry and Trade Vocational College, Wenzhou 325000, Zhejiang, China
| | - Yunjie Liang
- Optometry Technology of Zhejiang Industry and Trade Vocational College, Wenzhou 325000, Zhejiang, China
| | - Xiaojie Huang
- Optometry Technology of Zhejiang Industry and Trade Vocational College, Wenzhou 325000, Zhejiang, China
| |
Collapse
|
9
|
Hendriks WJAJ, van Cruchten RTP, Pulido R. Hereditable variants of classical protein tyrosine phosphatase genes: Will they prove innocent or guilty? Front Cell Dev Biol 2023; 10:1051311. [PMID: 36755664 PMCID: PMC9900141 DOI: 10.3389/fcell.2022.1051311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
Protein tyrosine phosphatases, together with protein tyrosine kinases, control many molecular signaling steps that control life at cellular and organismal levels. Impairing alterations in the genes encoding the involved proteins is expected to profoundly affect the quality of life-if compatible with life at all. Here, we review the current knowledge on the effects of germline variants that have been reported for genes encoding a subset of the protein tyrosine phosphatase superfamily; that of the thirty seven classical members. The conclusion must be that the newest genome research tools produced an avalanche of data that suggest 'guilt by association' for individual genes to specific disorders. Future research should face the challenge to investigate these accusations thoroughly and convincingly, to reach a mature genotype-phenotype map for this intriguing protein family.
Collapse
Affiliation(s)
- Wiljan J. A. J. Hendriks
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen, The Netherlands,*Correspondence: Wiljan J. A. J. Hendriks,
| | | | - Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
10
|
Swierkowska J, Vishweswaraiah S, Mrugacz M, Radhakrishna U, Gajecka M. Differential methylation of microRNA encoding genes may contribute to high myopia. Front Genet 2023; 13:1089784. [PMID: 36685896 PMCID: PMC9847511 DOI: 10.3389/fgene.2022.1089784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: High myopia (HM), an eye disorder with a refractive error ≤-6.0 diopters, has multifactorial etiology with environmental and genetic factors involved. Recent studies confirm the impact of alterations in DNA methylation and microRNAs (miRNAs) on myopia. Here, we studied the combined aspects evaluating to the role of methylation of miRNA encoding genes in HM. Materials and Methods: From the genome-wide DNA methylation data of 18 Polish children with HM and 18 matched controls, we retrieved differentially methylated CG dinucleotides localized in miRNA encoding genes. Putative target genes of the highest-ranked miRNAs were obtained from the miRDB and included in overrepresentation analyses in the ConsensusPathDB. Expression of target genes was assessed using the RNA sequencing data of retinal ARPE-19 cell line. Results: We identified differential methylation of CG dinucleotides in promoter regions of MIR3621, MIR34C, MIR423 (increased methylation level), and MIR1178, MIRLET7A2, MIR885, MIR548I3, MIR6854, MIR675, MIRLET7C, MIR99A (decreased methylation level) genes. Several targets of these miRNAs, e.g. GNAS, TRAM1, CTNNB1, EIF4B, TENM3 and RUNX were previously associated with myopia/HM/refractive error in Europeans in genome-wide association studies. Overrepresentation analyses of miRNAs' targets revealed enrichment in pathways/processes related to eye structure/function, such as axon guidance, transcription, focal adhesion, and signaling pathways of TGF-β, insulin, MAPK and EGF-EGFR. Conclusion: Differential methylation of indicated miRNA encoding genes might influence their expression and contribute to HM pathogenesis via disrupted regulation of transcription of miRNAs' target genes. Methylation of genes encoding miRNAs may be a new direction in research on both the mechanisms determining HM and non-invasive indicators in diagnostics.
Collapse
Affiliation(s)
| | - Sangeetha Vishweswaraiah
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, MI, United States
| | - Malgorzata Mrugacz
- Department of Ophthalmology and Eye Rehabilitation, Medical University of Bialystok, Bialystok, Poland
| | - Uppala Radhakrishna
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, MI, United States
| | - Marzena Gajecka
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland,Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland,*Correspondence: Marzena Gajecka,
| |
Collapse
|
11
|
Fan Y, Li J, Huang L, Wang K, Zhao M. 7-Methylxanthine Influences the Behavior of ADORA2A-DRD2 Heterodimers in Human Retinal Pigment Epithelial Cells. Ophthalmic Res 2022; 65:678-684. [PMID: 35724635 DOI: 10.1159/000525563] [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: 03/28/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The goal of this study was to investigate the presence of ADORA2A-DRD2 heterodimers in human retinal pigment epithelial (RPE) cells; determine if 7-methylxanthine (7-MX), a nonselective adenosine receptor antagonist which was used to control myopia progression, can influence the behavior of RPE cells through the ADORA2A-DRD2 receptor pathway; and assess the changes in the expression of signaling molecules during cellular signal transduction. METHODS Human RPE cells were cultured in vitro in the presence or absence of 7-MX. Cell proliferation was evaluated with the CCK-8 assay. Apoptosis and necrosis rates were determined by annexin V-FITC/propidium iodide staining and flow cytometry. Immunofluorescence and coimmunoprecipitation were used to examine the protein expression and colocalization of ADORA2A and DRD2 in RPE cells. ADORA2A and DRD2 were knocked down with small interfering RNAs (siRNAs). Changes in the protein expression of ERK1/2 and phospho-ERK1/2 (pERK 1/2), which are signaling molecules downstream of dopamine receptors, were evaluated by Western blot analysis. RESULTS Immunofluorescence and coimmunoprecipitation showed that ADORA2A and DRD2 were colocalized in RPE cells. The expression of ADORA2A in RPE cells was inhibited by treatment with 50 µmol/L 7-MX for 48 h, and the expression of DRD2, ERK1/2, and pERK1/2 was increased after treatment with 50 µmol/L 7-MX for 48 h. After siRNA-mediated knockdown of DRD2 in RPE cells and further treatment with 50 µmol/L 7-MX for 48 h, the expression of DRD2 was nearly restored to the level observed in the native control. At the experimental concentrations, 7-MX and siRNAs did not affect the proliferation or apoptosis of human RPE cells. CONCLUSIONS ADORA2A and DRD2 heterodimers were present in RPE cells. 7-MX may affect the behaviors of RPE cells through the ADORA2A-DRD2 receptor pathway. 7-MX is an inhibitor of ADORA2A receptors that can prevent inhibition of the DRD2 receptor pathway and increase DRD2 receptor pathway activity. This phenomenon may explain the mechanism of action through which 7-MX can control myopia progression.
Collapse
Affiliation(s)
- Yuzhuo Fan
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China, .,Department of Ophthalmology & Clinical Center of Optometry, Peking University People's Hospital, Beijing, China, .,College of Optometry, Peking University Health Science Center, Beijing, China, .,Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China, .,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China,
| | - Jiarui Li
- Department of Ophthalmology & Clinical Center of Optometry, Peking University People's Hospital, Beijing, China.,College of Optometry, Peking University Health Science Center, Beijing, China.,Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| | - Lvzhen Huang
- Department of Ophthalmology & Clinical Center of Optometry, Peking University People's Hospital, Beijing, China.,College of Optometry, Peking University Health Science Center, Beijing, China.,Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| | - Kai Wang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.,Department of Ophthalmology & Clinical Center of Optometry, Peking University People's Hospital, Beijing, China.,College of Optometry, Peking University Health Science Center, Beijing, China.,Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| | - Mingwei Zhao
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.,Department of Ophthalmology & Clinical Center of Optometry, Peking University People's Hospital, Beijing, China.,College of Optometry, Peking University Health Science Center, Beijing, China.,Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| |
Collapse
|