1
|
Khan M, Verma L. Crosstalk between signaling pathways (Rho/ROCK, TGF-β and Wnt/β-Catenin Pathways/ PI3K-AKT-mTOR) in Cataract: A Mechanistic Exploration and therapeutic strategy. Gene 2025; 947:149338. [PMID: 39965745 DOI: 10.1016/j.gene.2025.149338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
Cataract are a leading cause of visual impairment that is characterized by clouding or lens opacification of the healthy clear lens of the eye or its capsule. It can be classified based on their etiology and clinical presentation such as congenital, age-related, and secondary cataracts. Clinically, it may be further classified as a cortical or nuclear cataract. Cortical cataracts are responsible for opacification of the lens cortex, while nuclear cataracts cause age-related degeneration of the lens nucleus. This review aims to explore the molecular mechanism associated with various signaling pathways underlying cataract formation. Additionally, explore the potential therapeutic strategies for the management of cataracts. A comprehensive literature search was performed utilizing different keywords such as cataract, pathogenesis, signaling pathways, therapeutic approaches, RNA therapeutics, and surgery. Electronic databases such as PubMed, Google Scholar, Springer Link, and Web of Science were used for the literature search. The cataract formation is responsible for protein aggregation, primarily of γ-crystallin, and causes disruptions in signaling pathways. Key pathways include Rho/ROCK, TGF-β, Wnt/β-catenin, NF-κB, and PI3K-AKT-mTOR. Signaling pathways governing lens epithelial cell differentiation and epithelial-to-mesenchymal transition (EMT) are essential for maintaining lens transparency. Disruptions in these pathways, often caused by genetic mutations in genes like MIP, TDRD7, PAX6, FOXE3, HSF4, MAF, and PITX3 lead to cataract formation. While surgical intervention remains the primary treatment, pharmacological therapies and emerging RNA-based strategies offer promising strategies for the prevention and management of cataracts. A deeper understanding of the underlying molecular mechanisms is essential to develop innovative therapeutic strategies and improve the quality of life for individuals affected by cataracts.
Collapse
Affiliation(s)
- Meraj Khan
- Faculty of Pharmaceutical Sciences, Sagar Institute of Research & Technology-Pharmacy, Sanjeev Agrawal Global Educational University, Bhopal, Madhya.Pradesh 462022, India.
| | - Lokesh Verma
- Faculty of Pharmaceutical Sciences, Sagar Institute of Research & Technology-Pharmacy, Sanjeev Agrawal Global Educational University, Bhopal, Madhya.Pradesh 462022, India.
| |
Collapse
|
2
|
Fu L, Yang Q, Han Y, Sun F, Jin J, Wang J. Slit2 Promotes H 2O 2-Induced Lens Epithelial Cells Oxidative Damage and Age-Related Cataract. Curr Eye Res 2025; 50:41-50. [PMID: 39143744 DOI: 10.1080/02713683.2024.2388698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 05/14/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE To analyze the role of Slit2 in lens epithelial cell oxidative damage and its underlying mechanism. METHODS Human lens epithelial cells (SRA01/04 cells) and rat transparent lens were cultured with H2O2 to establish cell oxidative stress models and rat cataract models. Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot assays were employed to detect Slit2 levels within age-related cataracts(ARC) lens anterior capsule samples, rat cataract models, and cell oxidative stress models. In this study, qRT-PCR and Western blot assays were performed to derermine E-cadherin, N-cadherin, occludens1(ZO-1), α-SMA(α‑smooth muscle actin), Bcl-2, Bax, p-AKT, and AKT levels. In addition, Flow cytometry were performed to examine reactive oxygen species (ROS) and cell apoptosis. Cell viability, invasion, and migration were detected by CCK8, Transwell, and Wound healing. RESULTS Increased expression of Slit2 was found in ARC lens anterior capsule samples, H2O2-induced rat cataract models, and Human lens epithelial cells (HLECs) oxidative stress models. H2O2 significantly increased cell apoptosis and ROS generation, also accelerating cell migration, invasion, and epithelial-mesenchymal transition (EMT). In addition, H2O2 treatment repressed AKT phosphorylation and cell viability. Knock-down of Slit2 promoted cell viability and AKT phosphorylation levels, as well as repressed cell invasion, migration, apoptosis, ROS production and EMT. CONCLUSION Slit2 promoted lens epithelial cells oxidative stress damage via the AKT signalling pathways, providing a novel insight in ARC treatment.
Collapse
Affiliation(s)
- Lingzhi Fu
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Bengbu, Anhui, China
| | - Qing Yang
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Bengbu, Anhui, China
| | - Yuanyuan Han
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Feng Sun
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jiacheng Jin
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui, China
| | - Jianfeng Wang
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
3
|
Lin Q, Wang X, Han X, Peng X, Zhang X, Ran Y, Sun L, Wang Y, Han T, Zhou X. WES and Transcriptome Analysis Identifies FN1 as a Candidate Gene for Anterior Segment Dysgenesis. J Gene Med 2024; 26:e70001. [PMID: 39601165 DOI: 10.1002/jgm.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/29/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Anterior segment dysgenesis (ASD) disorders are phenotypically diverse and have multiple associated genes. This study reports on a Chinese family of three generations with ASD disorders and identifies several associated genes and pathways of the disorders. METHODS The history of illnesses, clinical observations, and blood samples of all family members were collected. Whole exome sequencing (WES) and polymerase chain reaction (PCR) were conducted to detect the genetic variants between patients and control members in the family. Transcriptomic study and quantitative real-time PCR (qRT-PCR) were subsequently performed to validate the differentially expressed genes (DEGs) and investigate the possible mechanisms of ASD caused by the variations. RESULTS The medical examination and illness history identified four members of the family diagnosed with ASD (III:3, II:3, II:2 and I:2). All four patients suffered various degrees of corneal opacity with abnormally thin cornea. Members II:3, II:2, and I:2 also had cataracts and iris hypoplasia and received an intraocular lens implant before the age of 20. We detected a heterozygous missense variation c.6122G > A (p.R2041Q and rs746145647) in fibronectin1 (FN1) in the four patients in this family that was absent in the other healthy members. The transcriptome and RT-PCR analysis revealed that the FN1 mRNA level was significantly upregulated in the blood samples of patients compared to healthy controls. A total of 909 DEGs were identified, including 607 upregulated genes and 302 downregulated genes. GO and KEGG annotation revealed that many DEGs were involved in biological processes closely related to focal adhesion, extracellular matrix-receptor interaction, TGF-β pathway, and the immune system. CONCLUSION This study identified an FN1 mutation associated with ASD patients and probed potential pathways related to it.
Collapse
Affiliation(s)
- Qinghong Lin
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
- Department of Refractive Surgery, Bright Eye Hospital, Fuzhou, China
| | - Xuejun Wang
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
| | - Xiaosong Han
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
| | - Xiaoliao Peng
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
| | - Xiaoyu Zhang
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
| | - Yuwen Ran
- Department of Refractive Surgery, Aier Eye Hospital, Changsha, China
| | - Ling Sun
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
| | - Yan Wang
- Department of Refractive Surgery, Bright Eye Hospital, Fuzhou, China
| | - Tian Han
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
| | - Xingtao Zhou
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, NHC, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Disease, Chinese Academy of Medical Sciences, Shanghai, China
- Ophthalmology and Optometry, Shanghai Research Center, Shanghai, China
- Shanghai Engineering Research Center of Laser and Autostereoscopic 3D for Vision Care (20DZ2255000), Shanghai, China
| |
Collapse
|
4
|
Kellermeyer R, Seidel C, Redwine WB, Moran RL, Bertho S, Ornelas-García CP, Alegre D, Weaver K, Unruh J, Troutwine B, Wang Y, Collins E, Rutkowski J, McGaugh SE, Espinasa L, Rohner N. Long-term hybridization in a karst window reveals the genetic basis of eye loss in cavefish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620266. [PMID: 39554031 PMCID: PMC11565769 DOI: 10.1101/2024.10.25.620266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Eye loss is a hallmark trait of animals inhabiting perpetual darkness, such as caves. The Mexican tetra ( Astyanax mexicanus) provides an unparalleled model for studying the genetic basis of eye loss. There are two interfertile morphs of the Mexican tetra, sighted surface fish and multiple independently evolved eyeless, blind cavefish populations. Despite decades of research on eye loss in this species, our understanding of the precise genetic basis remains sparse. Here, we focused on the unique Caballo Moro cave, where there is a karst window collapse that introduced sunlight and coexistence of both eyed and eyeless cavefish of similar genetic background. This unique genetic mosaic allowed us to pinpoint coding mutations in Connexin 50 (Cx50), also known as gap junction protein alpha 8b (Gja8b), as critical in the genetic basis of eye loss. CRISPR based knockouts of Cx50 in surface fish result in small or absent eyes as young as 48 hours post-fertilization. Further, we identified similar mutations in Cx50 that alter predicted protein structure among other cave-dwelling fish and even subterranean mammals, indicating a conserved evolutionary mechanism of Cx50 mutations. We introduced a mutation (Cx50-S89K) in mice, which resulted in cataracts, smaller eyes, and smaller lenses. Mutations in Cx50 mimic those identified in human congenital cataracts. We additionally leveraged phenotypic variation in a hybrid cavefish population to demonstrate that eyes provide fish with a metabolic advantage, providing a mechanism by which loss of eyes could be favored by selection. This unique hybridization event allowed us to identify novel alleles that contribute to the convergent evolution of eye loss, providing profound insights into the genetic underpinnings of one of nature's most fascinating adaptive traits.
Collapse
|
5
|
Li J, Buonfiglio F, Zeng Y, Pfeiffer N, Gericke A. Oxidative Stress in Cataract Formation: Is There a Treatment Approach on the Horizon? Antioxidants (Basel) 2024; 13:1249. [PMID: 39456502 PMCID: PMC11505147 DOI: 10.3390/antiox13101249] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Cataracts, a leading cause of blindness worldwide, are closely linked to oxidative stress-induced damage to lens epithelial cells (LECs). Key factors contributing to cataract formation include aging, arterial hypertension, and diabetes mellitus. Given the high global prevalence of cataracts, the burden of cataract-related visual impairment is substantial, highlighting the need for pharmacological strategies to supplement surgical interventions. Understanding the molecular pathways involved in oxidative stress during cataract development may offer valuable insights for designing novel therapeutic approaches. This review explores the role of oxidative stress in cataract formation, focusing on critical mechanisms, such as mitochondrial dysfunction, endoplasmic reticulum stress, loss of gap junctions, and various cell death pathways in LECs. Additionally, we discuss emerging therapeutic strategies and potential targeting options, including antioxidant-based treatments.
Collapse
Affiliation(s)
- Jingyan Li
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (Y.Z.); (N.P.)
| | | | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (Y.Z.); (N.P.)
| |
Collapse
|
6
|
Xia CH, Lin W, Li R, Xing X, Shang GJ, Zhang H, Gong X. Altered Cell Clusters and Upregulated Aqp1 in Connexin 50 Knockout Lens Epithelium. Invest Ophthalmol Vis Sci 2024; 65:27. [PMID: 39287589 PMCID: PMC11412383 DOI: 10.1167/iovs.65.11.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Purpose To characterize the heterogeneity and cell clusters of postnatal lens epithelial cells (LECs) and to investigate the downstream targets of connexin 50 (Cx50) in the regulation of lens homeostasis and lens growth. To determine differentially expressed genes (DEGs) in the connexin 50 knockout (Cx50KO) lens epithelial cells that shed light on novel mechanism underlying the cataract and small size of the Cx50KO lenses. Methods Single-cell RNA sequencing (scRNA-seq) of lens epithelial cells isolated from one-month-old Cx50KO and wild-type (WT) mice were performed. Differentially expressed genes were identified, and selected DEGs were further studied by quantitative real-time PCR (RT-qPCR) analysis and Western blot analysis. Results The expression profiles of several thousand genes were identified by scRNA-seq data analysis. In comparison to the WT control, many DEGs were identified in the Cx50KO lens epithelial cells, including growth regulating transcriptional factors and genes encoding water channels. Significantly upregulated aquaporin 1 (Aqp1) gene expression was confirmed by RT-qPCR, and upregulated AQP1 protein expression was confirmed by Western blot analysis and immunostaining both in vivo and in vitro. Conclusions Lens epithelial cells exhibit an intrinsic heterogeneity of different cell clusters in regulating lens homeostasis and lens growth. Upregulated Aqp1 in Cx50KO lens epithelial cells suggests that both connexin 50 and AQP1 likely play important roles in regulating water homeostasis in lens epithelial cells.
Collapse
Affiliation(s)
- Chun-Hong Xia
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - William Lin
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Rachel Li
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Xinfang Xing
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Guangdu Jack Shang
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Haiwei Zhang
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| | - Xiaohua Gong
- Herbert Wertheim School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, California, United States
| |
Collapse
|
7
|
Kulbay M, Wu KY, Nirwal GK, Bélanger P, Tran SD. Oxidative Stress and Cataract Formation: Evaluating the Efficacy of Antioxidant Therapies. Biomolecules 2024; 14:1055. [PMID: 39334822 PMCID: PMC11430732 DOI: 10.3390/biom14091055] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
This comprehensive review investigates the pivotal role of reactive oxygen species (ROS) in cataract formation and evaluates the potential of antioxidant therapies in mitigating this ocular condition. By elucidating the mechanisms of oxidative stress, the article examines how ROS contribute to the deterioration of lens proteins and lipids, leading to the characteristic aggregation, cross-linking, and light scattering observed in cataracts. The review provides a thorough assessment of various antioxidant strategies aimed at preventing and managing cataracts, such as dietary antioxidants (i.e., vitamins C and E, lutein, and zeaxanthin), as well as pharmacological agents with antioxidative properties. Furthermore, the article explores innovative therapeutic approaches, including gene therapy and nanotechnology-based delivery systems, designed to bolster antioxidant defenses in ocular tissues. Concluding with a critical analysis of current research, the review offers evidence-based recommendations for optimizing antioxidant therapies. The current literature on the use of antioxidant therapies to prevent cataract formation is sparse. There is a lack of evidence-based conclusions; further clinical studies are needed to endorse the use of antioxidant strategies in patients to prevent cataractogenesis. However, personalized treatment plans considering individual patient factors and disease stages can be applied. This article serves as a valuable resource, providing insights into the potential of antioxidants to alleviate the burden of cataracts.
Collapse
Affiliation(s)
- Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada
| | - Kevin Y Wu
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J4K 0A8, Canada
| | - Gurleen K Nirwal
- Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Paul Bélanger
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J4K 0A8, Canada
| | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
8
|
Zhou L, Wang G, Hu B, Jiang H, Jiang F, Xu Z. Microphthalmia and anterior segment dysgenesis due to a double gene variant in GJA8 and CRYGC. Eur J Ophthalmol 2024; 34:NP12-NP17. [PMID: 36916241 DOI: 10.1177/11206721231163611] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
INTRODUCTION To report a family with severe ocular disorder caused by double gene variants in causative genes of autosomal dominant cataracts, GJA8 and CRYGC. CASE PRESENTATION A 5-month-old boy with poor vision and enophthalmos was referred to our hospital. Further ocular examination showed horizontal nystagmus, iris abnormalities with pinpoint pupils, and extreme microphthalmia with axial right and left eye lengths of 13.48 mm and 13.75 mm, respectively. Digenic heterozygous variants (c.269T > G, p.Leu90Arg in CRYGC and c.151G > A, p.Asp51Asn in GJA8) have been detected based on the whole exome sequencing. His mother, who carried variant in CRYGC (c.269T > G, p.Leu90Arg), had nuclear cataract, microcornea and nystagmus, while his father, who carried variant in GJA8 (c.151G > A, p.Asp51Asn), showed bilateral membranous cataract, microphthalmia, sclerocornea, glaucoma, and nystagmus. CONCLUSIONS To our knowledge, this is the first report of a patient with variants in two cataract-related genes. Importantly, patient with double heterozygous variants in two dominantly inherited genes may suffer more serious phenotypes than those with heterozygous variant in a single dominantly inherited gene. Whole exome or genome sequencing is necessary for a genetic diagnosis in case of multiple gene variants.
Collapse
Affiliation(s)
- Lin Zhou
- Department of ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ganghua Wang
- Department of ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bin Hu
- Department of ophthalmology, Chengdu tianfu new district people's hospital, Chengdu, 641400, China
| | - Hui Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanwen Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhuping Xu
- Department of ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
9
|
Sun Q, Li J, Ma J, Zheng Y, Ju R, Li X, Ren X, Huang L, Chen R, Tan X, Luo L. JAM-C Is Important for Lens Epithelial Cell Proliferation and Lens Fiber Maturation in Murine Lens Development. Invest Ophthalmol Vis Sci 2023; 64:15. [PMID: 38095908 PMCID: PMC10723223 DOI: 10.1167/iovs.64.15.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
Purpose The underlying mechanism of congenital cataracts caused by deficiency or mutation of junctional adhesion molecule C (JAM-C) gene remains unclear. Our study aims to elucidate the abnormal developmental process in Jamc-/- lenses and reveal the genes related to lens development that JAM-C may regulate. Methods Jamc knockout (Jamc-/-) mouse embryos and pups were generated for in vivo studies. Four key developmental stages from embryonic day (E) 12.5 to postnatal day (P) 0.5 were selected for the following experiments. Hematoxylin and eosin staining was used for histological analysis. The 5-bromo-2'-deoxyuridine (BrdU) incorporation assay and TUNEL staining were performed to label lens epithelial cell (LEC) proliferation and apoptosis, respectively. Immunofluorescence and Western blot were used to analyze the markers of lens epithelium, cell cycle exit, and lens fiber differentiation. Results JAM-C was expressed throughout the process of lens development. Deletion of Jamc resulted in decreased lens size and disorganized lens fibers, which arose from E16.5 and aggravated gradually. The LECs of Jamc-/- lenses showed decreased quantity and proliferation, accompanied with reduction of key transcription factor, FOXE3. The fibers in Jamc-/- lenses were disorganized. Moreover, Jamc-deficient lens fibers showed significantly altered distribution patterns of Cx46 and Cx50. The marker of fiber homeostasis, γ-crystallin, was also decreased in the inner cortex and core fibers of Jamc-/- lenses. Conclusions Deletion of JAM-C exhibits malfunction of LEC proliferation and fiber maturation during murine lens development, which may be related to the downregulation of FOXE3 expression and abnormal localization patterns of Cx46 and Cx50.
Collapse
Affiliation(s)
- Qihang Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jiani Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingyu Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxing Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiangrong Ren
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lijuan Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Rongyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xuhua Tan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
10
|
Takashima M, Taniguchi K, Nagaya M, Yamamura S, Takamura Y, Inatani M, Oki M. Gene profiles and mutations in the development of cataracts in the ICR rat model of hereditary cataracts. Sci Rep 2023; 13:18161. [PMID: 37875594 PMCID: PMC10598066 DOI: 10.1038/s41598-023-45088-1] [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: 06/30/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023] Open
Abstract
Cataracts are opacifications of the lens that cause loss of visual acuity and ultimately of eyesight. Age-related cataract develops in most elderly people, but the mechanisms of cataract onset are incompletely understood. The Ihara Cataract Rat (ICR) is an animal model of hereditary cataracts showing cortical opacity that commonly develops prematurely. We identified putative mechanisms of cataract onset in the ICR rat model by measuring gene expression changes before and after cortical cataract development and conducting point mutation analysis. Genes differentially expressed between 4-week-old animals without cortical cataracts and 8-10-week-old animals with cortical cataracts were selected from microarray analysis. Three connections were identified by STRING analysis: (i) Epithelial-Mesenchymal Transition (EMT), including Col1a2, and Pik3r1. (ii) Lens homeostasis, including Aqp5, and Cpm. (iii) Lipid metabolism, including Scd1, Srebf1, and Pnpla3. Subsequently, mutation points were selected by comparing ICR rats with 12 different rats that do not develop cataracts. The apolipoprotein Apoc3 was mutated in ICR rats. Analyses of gene expression changes and point and mutations suggested that abnormalities in EMT or lipid metabolism could contribute to cataract development in ICR rats.
Collapse
Affiliation(s)
- Masaru Takashima
- Department of Industrial Creation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Kei Taniguchi
- Department of Industrial Creation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Masaya Nagaya
- Department of Industrial Creation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Shunki Yamamura
- Department of Industrial Creation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Yoshihiro Takamura
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masaru Inatani
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masaya Oki
- Department of Industrial Creation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan.
- Life Science Innovation Center, University of Fukui, Fukui, Japan.
| |
Collapse
|
11
|
Sun W, Gu S, Zhang F, Xu M, Chang P, Zhao Y. Congenital cataracts affect the retinal visual cycle and mitochondrial function: A multi-omics study of GJA8 knockout rabbits. J Proteomics 2023; 287:104972. [PMID: 37467890 DOI: 10.1016/j.jprot.2023.104972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/30/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Congenital cataracts are a threat to visual development in children, and the visual impairment persists after surgical treatment; however, the mechanisms involved remain unclear. Previous clinical studies have identified the effect of congenital cataracts on retinal morphology and function. To further understand the molecular mechanisms by which congenital cataracts affect retinal development, we analyzed retina samples from 7-week-old GJA8-knockout rabbits with congenital cataracts and controls by four-dimensional label-free quantification proteomics and untargeted metabolomics. Bioinformatics analysis of proteomic data showed that retinol metabolism, oxidative phosphorylation, and fatty acid degradation pathways were downregulated in the retinas of rabbits with congenital cataracts, indicating that their visual cycle and mitochondrial function were affected. Additional validation of differentially abundant proteins related to the visual cycle and mitochondrial function was performed using Parallel reaction monitoring and western blot experiments. Untargeted metabolome analysis showed significant upregulation of the antioxidant glutathione and ascorbic acid in the retinas of rabbits with congenital cataracts, indicating that their oxidative stress balance was not dysregulated. SIGNIFICANCE: Congenital cataracts in children can alter retinal structure and function, yet the mechanisms are uncertain. Here is the first study to use proteomics and metabolomics approaches to investigate the effects of congenital cataracts on retinal development in the early postnatal period. Our findings suggest that congenital cataracts have an impact on the retinal visual cycle and mitochondrial function. These findings give insight on the molecular pathways behind congenital cataract-induced visual function impairment in the early postnatal period.
Collapse
Affiliation(s)
- Weijie Sun
- Wenzhou Medical University School of Optometry and Ophthalmology, Eye Hospital, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; The State Key Laboratory of Optometry, Ophthalmology and Vision Science, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; National Center for Clinical and Medical Research, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China
| | - Siyi Gu
- Wenzhou Medical University School of Optometry and Ophthalmology, Eye Hospital, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; The State Key Laboratory of Optometry, Ophthalmology and Vision Science, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; National Center for Clinical and Medical Research, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China
| | - Fan Zhang
- Wenzhou Medical University School of Optometry and Ophthalmology, Eye Hospital, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; The State Key Laboratory of Optometry, Ophthalmology and Vision Science, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; National Center for Clinical and Medical Research, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China
| | - Mengxiang Xu
- Wenzhou Medical University School of Optometry and Ophthalmology, Eye Hospital, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; The State Key Laboratory of Optometry, Ophthalmology and Vision Science, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; National Center for Clinical and Medical Research, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China
| | - Pingjun Chang
- Wenzhou Medical University School of Optometry and Ophthalmology, Eye Hospital, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; The State Key Laboratory of Optometry, Ophthalmology and Vision Science, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; National Center for Clinical and Medical Research, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China.
| | - Yune Zhao
- Wenzhou Medical University School of Optometry and Ophthalmology, Eye Hospital, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; The State Key Laboratory of Optometry, Ophthalmology and Vision Science, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; National Center for Clinical and Medical Research, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China.
| |
Collapse
|
12
|
Du Y, Tong Y, Quan Y, Wang G, Cheng H, Gu S, Jiang JX. Protein kinase A activation alleviates cataract formation via increased gap junction intercellular communication. iScience 2023; 26:106114. [PMID: 36852280 PMCID: PMC9958365 DOI: 10.1016/j.isci.2023.106114] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/09/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Cataract is the leading cause of blindness worldwide. Here, we reported a potential, effective therapeutic mean for cataract prevention and treatment. Gap junction communication, an important mechanism in maintaining lens transparency, is increased by protein kinase A (PKA). We found that PKA activation reduced cataracts induced by oxidative stress, increased gap junctions/hemichannels in connexin (Cx) 50, Cx46 or Cx50 and Cx46 co-expressing cells, and decreased reactive oxygen species (ROS) levels. However, ROS reduction was shown in wild-type, Cx46 and Cx50 knockout, but not in Cx46/Cx50 double KO lens. In addition, PKA activation protects lens fiber cell death induced by oxidative stress via hemichannel-mediated glutathione transport. Connexin deletion increased lens opacity induced by oxidative stress associated with reduction of anti-oxidative stress gene expression. Together, our results suggest that PKA activation through increased connexin channels in lens fiber cell decreases ROS levels and cell death, leading to alleviated cataracts.
Collapse
Affiliation(s)
- Yu Du
- Department of Ophthalmology, Lanzhou University Second Hospital; Second Clinical School, Lanzhou University, Lanzhou, Gansu, 730000, China
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Yuxin Tong
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Yumeng Quan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
- Department of Ophthalmology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Guangyan Wang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
- Department of Ophthalmology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hongyun Cheng
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| |
Collapse
|
13
|
Abstract
Gap junctions, comprising connexin proteins, create conduits directly coupling the cytoplasms of adjacent cells. Expressed in essentially all tissues, dynamic gap junction structures enable the exchange of small molecules including ions and second messengers, and are central to maintenance of homeostasis and synchronized excitability. With such diverse and critical roles throughout the body, it is unsurprising that alterations to gap junction and/or connexin expression and function underlie a broad array of age-related pathologies. From neurological dysfunction to cardiac arrhythmia and bone loss, it is hard to identify a human disease state that does not involve reduced, or in some cases inappropriate, intercellular communication to affect organ function. With a complex life cycle encompassing several key regulatory steps, pathological gap junction remodeling during ageing can arise from alterations in gene expression, translation, intracellular trafficking, and posttranslational modification of connexins. Connexin proteins are now known to "moonlight" and perform a variety of non-junctional functions in the cell, independent of gap junctions. Furthermore, connexin "hemichannels" on the cell surface can communicate with the extracellular space without ever coupling to an adjacent cell to form a gap junction channel. This chapter will focus primarily on gap junctions in ageing, but such non-junctional connexin functions will be referred to where appropriate and the full spectrum of connexin biology should be noted as potentially causative/contributing to some findings in connexin knockout animals, for example.
Collapse
Affiliation(s)
- Michael J Zeitz
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.,FBRI Center for Vascular and Heart Research, Roanoke, VA, USA
| | - James W Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA. .,FBRI Center for Vascular and Heart Research, Roanoke, VA, USA. .,Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, USA. .,Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.
| |
Collapse
|
14
|
A Quantitative Assay for Ca2+ Uptake through Normal and Pathological Hemichannels. Int J Mol Sci 2022; 23:ijms23137337. [PMID: 35806342 PMCID: PMC9266989 DOI: 10.3390/ijms23137337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 12/25/2022] Open
Abstract
Connexin (Cx) hemichannels (HCs) are large pore hexameric structures that allow the exchange of ions, metabolites and a variety of other molecules between the cell cytoplasm and extracellular milieu. HC inhibitors are attracting growing interest as drug candidates because deregulated fluxes through HCs have been implicated in a plethora of genetic conditions and other diseases. HC activity has been mainly investigated by electrophysiological methods and/or using HC-permeable dye uptake measurements. Here, we present an all-optical assay based on fluorometric measurements of ionized calcium (Ca2+) uptake with a Ca2+-selective genetically encoded indicator (GCaMP6s) that permits the optical tracking of cytosolic Ca2+ concentration ([Ca2+]cyt) changes with high sensitivity. We exemplify use of the assay in stable pools of HaCaT cells overexpressing human Cx26, Cx46, or the pathological mutant Cx26G45E, under control of a tetracycline (Tet) responsive element (TRE) promoter (Tet-on). We demonstrate the usefulness of the assay for the characterization of new monoclonal antibodies (mAbs) targeting the extracellular domain of the HCs. Although we developed the assay on a spinning disk confocal fluorescence microscope, the same methodology can be extended seamlessly to high-throughput high-content platforms to screen other kinds of inhibitors and/or to probe HCs expressed in primary cells and microtissues.
Collapse
|
15
|
Li Z, Quan Y, Gu S, Jiang JX. Beyond the Channels: Adhesion Functions of Aquaporin 0 and Connexin 50 in Lens Development. Front Cell Dev Biol 2022; 10:866980. [PMID: 35465319 PMCID: PMC9022433 DOI: 10.3389/fcell.2022.866980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 12/04/2022] Open
Abstract
Lens, an avascular tissue involved in light transmission, generates an internal microcirculatory system to promote ion and fluid circulation, thus providing nutrients to internal lens cells and excreting the waste. This unique system makes up for the lack of vasculature and distinctively maintains lens homeostasis and lens fiber cell survival through channels of connexins and other transporters. Aquaporins (AQP) and connexins (Cx) comprise the majority of channels in the lens microcirculation system and are, thus, essential for lens development and transparency. Mutations of AQPs and Cxs result in abnormal channel function and cataract formation. Interestingly, in the last decade or so, increasing evidence has emerged suggesting that in addition to their well-established channel functions, AQP0 and Cx50 play pivotal roles through channel-independent actions in lens development and transparency. Specifically, AQP0 and Cx50 have been shown to have a unique cell adhesion function that mediates lens development and transparency. Precise regulation of cell-matrix and cell-cell adhesion is necessary for cell migration, a critical process during lens development. This review will provide recent advances in basic research of cell adhesion mediated by AQP0 and Cx50.
Collapse
Affiliation(s)
- Zhen Li
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yumeng Quan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| |
Collapse
|