1
|
Lu X, Zhang Y, Wang R, Li Z. Ginkgolide B Inhibits EMT and Promotes Pyroptosis in Gastric Cancer via AKT/mTOR Pathway. Drug Des Devel Ther 2025; 19:2491-2502. [PMID: 40190805 PMCID: PMC11972580 DOI: 10.2147/dddt.s485240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/08/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide, necessitating the exploration of novel therapeutic agents to improve patient outcomes. This study elucidates the anti-cancer properties of Ginkgolide B (GGB), a diterpenoid lactone derived from Ginkgo biloba, in both in vitro and in vivo models of GC. Methods and Results Using AGS and HGC-27 cell lines, we assessed GGB's impact on cellular proliferation, colony formation, migration, invasion, apoptosis, and pyroptosis. GGB exhibited significant dose- and time-dependent inhibition of cell proliferation and colony formation, with no cytotoxicity observed in normal gastric epithelial cells. Furthermore, GGB markedly suppressed migration and invasion, and induced apoptosis and pyroptosis, as evidenced by increased Bax and GSDMD expression and decreased Bcl-2 levels. In vivo, GGB treatment significantly reduced tumor growth in a nude mouse xenograft model and modulated EMT markers, decreasing PCNA and N-cadherin levels while increasing E-cadherin expression. Mechanistically, GGB's anti-cancer effects were mediated through the deactivation of the PI3K/AKT/mTOR signaling pathway. Conclusion These findings underscore the potential of GGB as a promising therapeutic agent for GC, warranting further clinical evaluation.
Collapse
MESH Headings
- Humans
- Lactones/pharmacology
- Lactones/chemistry
- Ginkgolides/pharmacology
- Ginkgolides/chemistry
- TOR Serine-Threonine Kinases/metabolism
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/pathology
- Stomach Neoplasms/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Animals
- Cell Proliferation/drug effects
- Mice
- Mice, Nude
- Epithelial-Mesenchymal Transition/drug effects
- Dose-Response Relationship, Drug
- Pyroptosis/drug effects
- Drug Screening Assays, Antitumor
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/isolation & purification
- Signal Transduction/drug effects
- Cell Movement/drug effects
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Structure-Activity Relationship
- Tumor Cells, Cultured
- Mice, Inbred BALB C
- Molecular Structure
- Apoptosis/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xinxing Lu
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Yan Zhang
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Ran Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Ziyu Li
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Qin Z, Yao K, Fu Q. Generation of Functional Lentoid Bodies from Human-Induced Pluripotent Stem Cells. Methods Mol Biol 2025; 2848:25-36. [PMID: 39240514 DOI: 10.1007/978-1-0716-4087-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The pathological mechanisms of cataract remain largely unknown due to the lack of appropriate in vitro cellular models. We developed a stable in vitro system, namely, a "fried egg" differentiation method to generate functional lentoid bodies (LBs) from induced pluripotent stem cells (iPSCs). The iPSCs-derived LBs exhibited crystalline lens-like morphology and a transparent structure, and expressed lens-specific markers. TEM examination and optical analysis further demonstrated that it has the same cell arrangement structure and magnifying ability as lens.
Collapse
Affiliation(s)
- Zhenwei Qin
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Ke Yao
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Qiuli Fu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Wu H, Mao Y, Wang Q, Yu H, Bouaziz M, Makrides N, Koleske AJ, Radice GL, Zhang X. Abl kinases regulate FGF signaling independent of Crk phosphorylation to prevent Peters anomaly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.619064. [PMID: 39484567 PMCID: PMC11526961 DOI: 10.1101/2024.10.24.619064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Peters anomaly, the most common cause of congenital corneal opacity, stems from corneal-lenticular adhesion. Despite numerous identified mutations, a cohesive molecular framework of the disease's etiology remains elusive. Here, we identified Abl kinases as pivotal regulators of FGF signaling, as genetic ablation of Abl kinases restores lens induction even in the absence of FGF signaling. Intriguingly, both Abl kinase deficiency and increased FGF-Ras activity result in Peters anomaly independent of ERK signaling, which can be rescued by allelic deletion of Abl substrate, Crk. However, contrary to the prevailing belief that Abl kinases regulate Crk proteins by direct phosphorylation, mutations at Abl kinase phosphorylation sites on Crk and CrkL did not yield any observable effects. Instead, our findings reveal that Abl kinases phosphorylate Ptpn12, which in turn inhibits p130Cas phosphorylation and Crk recruitment, crucial for Rho GTPases activation and cytoskeletal dynamics. Consequently, Abl kinase deficiency reduces actomyosin contractility within the lens vesicle and genetically interacts with RhoA inhibition. Conversely, Rac1 deletion mitigates Peters anomaly in models with aberrant FGF, Abl kinase and RhoA signaling. Our results demonstrate that Abl kinases regulate FGF signaling to balance RhoA and Rac1 activity via the Ptpn12-p130Cas pathway, suggesting that targeting tension-mediated lens vesicle separation could be a therapeutic strategy for Peters anomaly.
Collapse
Affiliation(s)
- Hao Wu
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Yingyu Mao
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Qian Wang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Honglian Yu
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Michael Bouaziz
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Neoklis Makrides
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Anthony J. Koleske
- Departments of Molecular Biophysics and Biochemistry and Neuroscience, Yale University, New Haven, CT 06520, USA
| | - Glenn L. Radice
- Department of Medicine, The Warren Alpert Medical School of Brown University, Lifespan Cardiovascular Institute, Rhode Island Hospital, Providence, RI 02903, USA
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
4
|
Zhang JJ, Cao ZF, Zhou BT, Yang JH, Li Z, Lin S, Chen XL, Zhang NW, Ye Q, Ma X, Zhu YH. Abnormal function of EPHA2/p.R957P mutant in congenital cataract. Int J Ophthalmol 2024; 17:1007-1017. [PMID: 38895685 PMCID: PMC11144770 DOI: 10.18240/ijo.2024.06.04] [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: 12/07/2023] [Accepted: 03/18/2024] [Indexed: 06/21/2024] Open
Abstract
AIM To identify genetic defects in a Chinese family with congenital posterior polar cataracts and assess the pathogenicity. METHODS A four-generation Chinese family affected with autosomal dominant congenital cataract was recruited. Nineteen individuals took part in this study including 5 affected and 14 unaffected individuals. Sanger sequencing targeted hot-spot regions of 27 congenital cataract-causing genes for variant discovery. The pathogenicity of the variant was evaluated by the guidelines of American College of Medical Genetics and InterVar software. Confocal microscopy was applied to detect the subcellular localization of fluorescence-labeled ephrin type-A receptor 2 (EPHA2). Co-immunoprecipitation assay was implemented to estimate the interaction between EphA2 and other lens membrane proteins. The mRNA and protein expression were analyzed by reverse transcription-polymerase chain reaction (qRT-PCR) and Western blotting assay, respectively. The cell migration was analyzed by wound healing assay. Zebrafish model was generated by ectopic expression of human EPHA2/p.R957P mutant to demonstrate whether the mutant could cause lens opacity in vivo. RESULTS A novel missense and pathogenic variant c.2870G>C was identified in the sterile alpha motif (SAM) domain of EPHA2. Functional studies demonstrated the variant's impact: reduced EPHA2 protein expression, altered subcellular localization, and disrupted interactions with other lens membrane proteins. This mutant notably enhanced human lens epithelial cell migration, and induced a central cloudy region and roughness in zebrafish lenses with ectopic expression of human EPHA2/p.R957P mutant under differential interference contrast (DIC) optics. CONCLUSION Novel pathogenic c.2870G>C variant of EPHA2 in a Chinese congenital cataract family contributes to disease pathogenesis.
Collapse
Affiliation(s)
- Jing-Jin Zhang
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University; Fujian Institute of Ophthalmology; Fujian Provincial Clinical Medical Research Center of Eye Diseases and Optometry, Fuzhou 350005, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Zong-Fu Cao
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100000, China
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing 100081, China
| | - Bi-Ting Zhou
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University; Fujian Institute of Ophthalmology; Fujian Provincial Clinical Medical Research Center of Eye Diseases and Optometry, Fuzhou 350005, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Ju-Hua Yang
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University; Fujian Institute of Ophthalmology; Fujian Provincial Clinical Medical Research Center of Eye Diseases and Optometry, Fuzhou 350005, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian Province, China
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Zhong Li
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Shuang Lin
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Xiao-Le Chen
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Nan-Wen Zhang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Qin Ye
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University; Fujian Institute of Ophthalmology; Fujian Provincial Clinical Medical Research Center of Eye Diseases and Optometry, Fuzhou 350005, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Xu Ma
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100000, China
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing 100081, China
| | - Yi-Hua Zhu
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University; Fujian Institute of Ophthalmology; Fujian Provincial Clinical Medical Research Center of Eye Diseases and Optometry, Fuzhou 350005, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| |
Collapse
|
5
|
Camerino M, Chang W, Cvekl A. Analysis of long-range chromatin contacts, compartments and looping between mouse embryonic stem cells, lens epithelium and lens fibers. Epigenetics Chromatin 2024; 17:10. [PMID: 38643244 PMCID: PMC11031936 DOI: 10.1186/s13072-024-00533-x] [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: 11/15/2023] [Accepted: 03/08/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Nuclear organization of interphase chromosomes involves individual chromosome territories, "open" and "closed" chromatin compartments, topologically associated domains (TADs) and chromatin loops. The DNA- and RNA-binding transcription factor CTCF together with the cohesin complex serve as major organizers of chromatin architecture. Cellular differentiation is driven by temporally and spatially coordinated gene expression that requires chromatin changes of individual loci of various complexities. Lens differentiation represents an advantageous system to probe transcriptional mechanisms underlying tissue-specific gene expression including high transcriptional outputs of individual crystallin genes until the mature lens fiber cells degrade their nuclei. RESULTS Chromatin organization between mouse embryonic stem (ES) cells, newborn (P0.5) lens epithelium and fiber cells were analyzed using Hi-C. Localization of CTCF in both lens chromatins was determined by ChIP-seq and compared with ES cells. Quantitative analyses show major differences between number and size of TADs and chromatin loop size between these three cell types. In depth analyses show similarities between lens samples exemplified by overlaps between compartments A and B. Lens epithelium-specific CTCF peaks are found in mostly methylated genomic regions while lens fiber-specific and shared peaks occur mostly within unmethylated DNA regions. Major differences in TADs and loops are illustrated at the ~ 500 kb Pax6 locus, encoding the critical lens regulatory transcription factor and within a larger ~ 15 Mb WAGR locus, containing Pax6 and other loci linked to human congenital diseases. Lens and ES cell Hi-C data (TADs and loops) together with ATAC-seq, CTCF, H3K27ac, H3K27me3 and ENCODE cis-regulatory sites are shown in detail for the Pax6, Sox1 and Hif1a loci, multiple crystallin genes and other important loci required for lens morphogenesis. The majority of crystallin loci are marked by unexpectedly high CTCF-binding across their transcribed regions. CONCLUSIONS Our study has generated the first data on 3-dimensional (3D) nuclear organization in lens epithelium and lens fibers and directly compared these data with ES cells. These findings generate novel insights into lens-specific transcriptional gene control, open new research avenues to study transcriptional condensates in lens fiber cells, and enable studies of non-coding genetic variants linked to cataract and other lens and ocular abnormalities.
Collapse
Affiliation(s)
- Michael Camerino
- The Departments Genetics, Albert Einstein College of Medicine, NY10461, Bronx, USA
| | - William Chang
- Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, NY10461, Bronx, USA
| | - Ales Cvekl
- The Departments Genetics, Albert Einstein College of Medicine, NY10461, Bronx, USA.
- Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, NY10461, Bronx, USA.
| |
Collapse
|
6
|
Sugiyama Y, Reed DA, Herrmann D, Lovicu FJ, Robinson ML, Timpson P, Masai I. Fibroblast growth factor-induced lens fiber cell elongation is driven by the stepwise activity of Rho and Rac. Development 2024; 151:dev202123. [PMID: 38240393 PMCID: PMC10911273 DOI: 10.1242/dev.202123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024]
Abstract
The spheroidal shape of the eye lens is crucial for precise light focusing onto the retina. This shape is determined by concentrically aligned, convexly elongated lens fiber cells along the anterior and posterior axis of the lens. Upon differentiation at the lens equator, the fiber cells increase in height as their apical and basal tips migrate towards the anterior and posterior poles, respectively. The forces driving this elongation and migration remain unclear. We found that, in the mouse lens, membrane protrusions or lamellipodia are observed only in the maturing fibers undergoing cell curve conversion, indicating that lamellipodium formation is not the primary driver of earlier fiber migration. We demonstrated that elevated levels of fibroblast growth factor (FGF) suppressed the extension of Rac-dependent protrusions, suggesting changes in the activity of FGF controlling Rac activity, switching to lamellipodium-driven migration. Inhibitors of ROCK, myosin and actin reduced the height of both early and later fibers, indicating that elongation of these fibers relies on actomyosin contractility. Consistent with this, active RhoA was detected throughout these fibers. Given that FGF promotes fiber elongation, we propose that it does so through regulation of Rho activity.
Collapse
Affiliation(s)
- Yuki Sugiyama
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
| | - Daniel A. Reed
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David Herrmann
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Frank J. Lovicu
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael L. Robinson
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Paul Timpson
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
| |
Collapse
|
7
|
Wang JD, Zhang JS, Li XX, Wang KJ, Li M, Mao YY, Wan XH. Knockout of TGF-β receptor II by CRISPR/Cas9 delays mesenchymal transition of Lens epithelium and posterior capsule opacification. Int J Biol Macromol 2024; 259:129290. [PMID: 38199534 DOI: 10.1016/j.ijbiomac.2024.129290] [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: 09/07/2022] [Revised: 12/16/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024]
Abstract
Posterior capsule opacification (PCO) is the most common postoperative complication of cataract surgery. Transforming growth factor-β (TGF-β) is related to epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) that is proven to induce PCO formation in clinical and experimental studies. In this study, CRISPR sequences targeting exon of TGF-βRII were knocked out with lentiviral transfection in LECs. Rabbits' PCO model was established and recombinant adeno-associated virus (AAV) for transferring the gRNA of TGF βRII were intravitreally injected. SgRNA inhibited TGF-βRII expression and human LECs proliferation. In TGF-βRII knockout group, LECs motility and migration were suppressed, N-cadherin and vimentin expressions were significantly decreased, whereas E-cadherin was increased. The animal model showed that TGF-βRII knockout in vivo was effective in suppressing PCO. The current study suggested that the CRISPR/Cas9 endonuclease system could suppress TGF-βRII secretion, which participates in the EMT procedure of LECs in vitro and PCO in vivo. These findings might provide a new gene-editing approach and insight into a novel therapeutic strategy for PCO.
Collapse
Affiliation(s)
- Jin Da Wang
- Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing 100730, China
| | - Jing Shang Zhang
- Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing 100730, China
| | - Xiao Xia Li
- Department of Ophthalmology, Beijing Shijitan Hospital of Capital Medical University, Beijing 100038, China
| | - Kai Jie Wang
- Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing 100730, China
| | - Meng Li
- Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing 100730, China
| | - Ying Yan Mao
- Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| | - Xiu Hua Wan
- Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing 100730, China.
| |
Collapse
|
8
|
Sugiyama Y, Reed DA, Herrmann D, Lovicu FJ, Robinson ML, Timpson P, Masai I. Fibroblast Growth Factor-induced lens fiber cell elongation is driven by the stepwise activity of Rho and Rac. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569812. [PMID: 38106159 PMCID: PMC10723307 DOI: 10.1101/2023.12.03.569812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The spheroidal shape of the eye lens is critical for precise light focusing onto the retina. This shape is determined by concentrically aligned, convexly elongated lens fiber cells along the anterior and posterior axis of the lens. Upon differentiation at the lens equator, the fiber cells increase in height as their apical and basal tips migrate towards the anterior and posterior poles, respectively. The forces driving this elongation and migration remain unclear. We found that membrane protrusions or lamellipodia are observed only in the maturing fibers undergoing cell curve conversion, indicating lamellipodium is not the primary driver of earlier fiber migration. We demonstrated that elevated levels of fibroblast growth factor (FGF) suppressed the extension of Rac-dependent protrusions, suggesting changes in the activity of FGF controling Rac activity, switching to lamellipodium-driven migration. Inhibitors of ROCK, myosin, and actin reduced the height of both early and later fibers, indicating elongation of these fibers relies on actomyosin contractility. Consistently, active RhoA was detected throughout these fibers. Given that FGF promotes fiber elongation, we propose it to do so through regulation of Rho activity.
Collapse
Affiliation(s)
- Yuki Sugiyama
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
| | - Daniel A. Reed
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David Herrmann
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Frank J. Lovicu
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael L. Robinson
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH, USA
| | - Paul Timpson
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
| |
Collapse
|
9
|
Xiang J, Pompetti AJ, Faranda AP, Wang Y, Novo SG, Li DWC, Duncan MK. ATF4 May Be Essential for Adaption of the Ocular Lens to Its Avascular Environment. Cells 2023; 12:2636. [PMID: 37998373 PMCID: PMC10670291 DOI: 10.3390/cells12222636] [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: 03/23/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023] Open
Abstract
The late embryonic mouse lens requires the transcription factor ATF4 for its survival although the underlying mechanisms were unknown. Here, RNAseq analysis revealed that E16.5 Atf4 null mouse lenses downregulate the mRNA levels of lens epithelial markers as well as known markers of late lens fiber cell differentiation. However, a comparison of this list of differentially expressed genes (DEGs) with other known transcriptional regulators of lens development indicated that ATF4 expression is not directly controlled by the previously described lens gene regulatory network. Pathway analysis revealed that the Atf4 DEG list was enriched in numerous genes involved in nutrient transport, amino acid biosynthesis, and tRNA charging. These changes in gene expression likely result in the observed reductions in lens free amino acid and glutathione levels, which would result in the observed low levels of extractable lens protein, finally leading to perinatal lens disintegration. These data demonstrate that ATF4, via its function in the integrated stress response, is likely to play a crucial role in mediating the adaption of the lens to the avascularity needed to maintain lens transparency.
Collapse
Affiliation(s)
- Jiawen Xiang
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510230, China
| | - Anthony J. Pompetti
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Adam P. Faranda
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yan Wang
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Samuel G. Novo
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - David Wan-Cheng Li
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510230, China
| | - Melinda K. Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
10
|
Duot M, Viel R, Viet J, Le Goff-Gaillard C, Paillard L, Lachke SA, Gautier-Courteille C, Reboutier D. Eye Lens Organoids Made Simple: Characterization of a New Three-Dimensional Organoid Model for Lens Development and Pathology. Cells 2023; 12:2478. [PMID: 37887322 PMCID: PMC10605248 DOI: 10.3390/cells12202478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Cataract, the opacification of the lens, is the leading cause of blindness worldwide. Although effective, cataract surgery is costly and can lead to complications. Toward identifying alternate treatments, it is imperative to develop organoid models relevant for lens studies and drug screening. Here, we demonstrate that by culturing mouse lens epithelial cells under defined three-dimensional (3D) culture conditions, it is possible to generate organoids that display optical properties and recapitulate many aspects of lens organization and biology. These organoids can be rapidly produced in large amounts. High-throughput RNA sequencing (RNA-seq) on specific organoid regions isolated via laser capture microdissection (LCM) and immunofluorescence assays demonstrate that these lens organoids display a spatiotemporal expression of key lens genes, e.g., Jag1, Pax6, Prox1, Hsf4 and Cryab. Further, these lens organoids are amenable to the induction of opacities. Finally, the knockdown of a cataract-linked RNA-binding protein encoding gene, Celf1, induces opacities in these organoids, indicating their use in rapidly screening for genes that are functionally relevant to lens biology and cataract. In sum, this lens organoid model represents a compelling new tool to advance the understanding of lens biology and pathology and can find future use in the rapid screening of compounds aimed at preventing and/or treating cataracts.
Collapse
Affiliation(s)
- Matthieu Duot
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Roselyne Viel
- CNRS, Inserm UMS Biosit, H2P2 Core Facility, Université de Rennes, 35000 Rennes, France
| | - Justine Viet
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - Catherine Le Goff-Gaillard
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - Luc Paillard
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - Salil A. Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA
| | - Carole Gautier-Courteille
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - David Reboutier
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| |
Collapse
|
11
|
Giannone AA, Sellitto C, Rosati B, McKinnon D, White TW. Single-Cell RNA Sequencing Analysis of the Early Postnatal Mouse Lens Epithelium. Invest Ophthalmol Vis Sci 2023; 64:37. [PMID: 37870847 PMCID: PMC10599162 DOI: 10.1167/iovs.64.13.37] [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: 08/14/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
Purpose The lens epithelium maintains the overall health of the organ. We used single-cell RNA sequencing (scRNA-seq) technology to assess transcriptional heterogeneity between cells in the postnatal day 2 (P2) epithelium and identify distinct epithelial cell subtypes. Analysis of these data was used to better understand lens growth, differentiation, and homeostasis on P2. Methods scRNA-seq on P2 mouse lenses was performed using the 10x Genomics Chromium Single Cell 3' Kit (v3.1) and short-read Illumina sequencing. Sequence alignment and preprocessing of data were conducted using 10x Genomics Cell Ranger software. Seurat was employed for preprocessing, quality control, dimensionality reduction, and cell clustering, and Monocle was utilized for trajectory analysis to understand the developmental progression of the lens cells. CellChat and GO analyses were used to explore cell-cell communication networks and signaling interactions. Results Lens epithelial cells (LECs) were divided into seven subclusters, classified by specific gene markers. The expression of crystallin, cell-cycle, and metabolic genes was not uniform, indicating distinct functional roles of LECs. Trajectory analysis predicted a bifurcation of differentiating and cycling cells from an Igfbp5+ progenitor pool. We also identified heterogeneity in signaling molecules and pathways, suggesting that cycling and progenitor subclusters have prominent roles in coordinating crosstalk. Conclusions scRNA-seq corroborated many known markers of epithelial differentiation and proliferation while providing further insight into the pathways and genes directing these processes. Interestingly, we demonstrated that the developing epithelium can be divided into distinct subpopulations. These clusters reflect the transcriptionally diverse roles of the epithelium in proliferation, signaling, and maintenance.
Collapse
Affiliation(s)
- Adrienne A. Giannone
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Caterina Sellitto
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Barbara Rosati
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
- Veterans Affairs Medical Center, Northport, New York, United States
| | - David McKinnon
- Department of Neurobiology and Behavior, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Thomas W. White
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| |
Collapse
|
12
|
Duot M, Viel R, Viet J, Le Goff-Gaillard C, Paillard L, Lachke SA, Gautier-Courteille C, Reboutier D. Eye lens organoids going simple: characterization of a new 3-dimensional organoid model for lens development and pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548679. [PMID: 37503005 PMCID: PMC10370037 DOI: 10.1101/2023.07.12.548679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The ocular lens, along with the cornea, focuses light on the retina to generate sharp images. Opacification of the lens, or cataract, is the leading cause of blindness worldwide. Presently, the best approach for cataract treatment is to surgically remove the diseased lens and replace it with an artificial implant. Although effective, this is costly and can have post-surgical complications. Toward identifying alternate treatments, it is imperative to develop organoid models relevant for lens studies and anti-cataract drug screening. Here, we demonstrate that by culturing mouse lens epithelial cells under defined 3-dimensional (3D) culture conditions, it is possible to generate organoids that display optical properties and recapitulate many aspects of lens organization at the tissue, cellular and transcriptomic levels. These 3D cultured lens organoids can be rapidly produced in large amounts. High-throughput RNA-sequencing (RNA-seq) on specific organoid regions isolated by laser capture microdissection (LCM) and immunofluorescence assays demonstrate that these lens organoids display spatiotemporal expression of key lens genes, e.g. , Jag1 , Pax6 , Prox1 , Hsf4 and Cryab . Further, these lens organoids are amenable to induction of opacities. Finally, knockdown of a cataract-linked RNA-binding protein encoding gene, Celf1 , induces opacities in these organoids, indicating their use in rapidly screening for genes functionally relevant to lens biology and cataract. In sum, this lens organoid model represents a compelling new tool to advance the understanding of lens biology and pathology, and can find future use in the rapid screening of compounds aimed at preventing and/or treating cataract.
Collapse
|
13
|
Islam ST, Cheng C, Parreno J, Fowler VM. Nonmuscle Myosin IIA Regulates the Precise Alignment of Hexagonal Eye Lens Epithelial Cells During Fiber Cell Formation and Differentiation. Invest Ophthalmol Vis Sci 2023; 64:20. [PMID: 37070941 PMCID: PMC10123325 DOI: 10.1167/iovs.64.4.20] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/27/2023] [Indexed: 04/19/2023] Open
Abstract
Purpose Epithelial cells in the equatorial region of the ocular lens undergo a remarkable transition from randomly packed cells into precisely aligned and hexagon-shaped cells organized into meridional rows. We investigated the function of nonmuscle myosin IIA (encoded by Myh9) in regulating equatorial epithelial cell alignment to form meridional rows during secondary fiber cell morphogenesis. Methods We used genetic knock-in mice to study a common human Myh9 mutation, E1841K, in the rod domain. The E1841K mutation disrupts bipolar filament assembly. Lens shape, clarity, and stiffness were evaluated, and Western blots were used to determine the level of normal and mutant myosins. Cryosections and lens whole mounts were stained and imaged by confocal microscopy to investigate cell shape and organization. Results We observed no obvious changes in lens size, shape, and biomechanical properties (stiffness and resilience) between the control and nonmuscle myosin IIA-E1841K mutant mice at 2 months of age. Surprisingly, we found misalignment and disorder of fiber cells in heterozygous and homozygous mutant lenses. Further analysis revealed misshapen equatorial epithelial cells that cause disorientation of the meridional rows before fiber cell differentiation in homozygous mutant lenses. Conclusions Our data indicate that nonmuscle myosin IIA bipolar filament assembly is required for the precise alignment of the meridional rows at the lens equator and that the organization of lens fiber cells depends on the proper patterning of meridional row epithelial cells. These data also suggest that lens fiber cell organization and a hexagonal shape are not required for normal lens size, shape transparency, or biomechanical properties.
Collapse
Affiliation(s)
- Sadia T. Islam
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Catherine Cheng
- School of Optometry and Vision Science Program, Indiana University, Bloomington, Indiana, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Justin Parreno
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Velia M. Fowler
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| |
Collapse
|
14
|
Li Z, Quan Y, Wang G, Ma B, Gu S, Jiang JX. The second extracellular domain of connexin 50 is important for in cell adhesion, lens differentiation, and adhesion molecule expression. J Biol Chem 2023; 299:102965. [PMID: 36736424 PMCID: PMC10011516 DOI: 10.1016/j.jbc.2023.102965] [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: 05/30/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Connexin (Cx)-forming channels play essential roles in maintaining lens homeostasis and transparency. We showed here channel-independent roles of Cx50 in cell-cell adhesion and confirmed the second extracellular (E2) domain as a critical domain for cell adhesion function. We found that cell adhesion decreased in cells expressing chimeric Cx50 in which the E2 domain was swapped with the E2 domain of either Cx43 or Cx46. In contrast, adhesion increased in cells expressing chimeric Cx43 and Cx46 with the Cx50 (E2) domain. This function is Cx channel-independent and Cx50 E2 domain-dependent cell adhesion acting in both homotypic and heterotypic manners. In addition, we generated eight site mutations of unique residues between Cx50 and the other two lens Cxs and found that mutation of any one of the residues abolished the adhesive function. Moreover, expression of adhesive-impaired mutants decreased adhesion-related proteins, N-cadherin and β-catenin. Expression of the adhesion-impaired Cx50W188P mutant in embryonic chick lens caused enlarged extracellular spaces, distorted fiber organization, delayed nuclear condensation, and cortical cataracts. In summary, the results from both in vitro and in vivo studies demonstrate the importance of the adhesive function of Cx50 in the lens.
Collapse
Affiliation(s)
- Zhen Li
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, USA; 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, Texas, USA
| | - Guangyan Wang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Bo Ma
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, USA.
| |
Collapse
|
15
|
Martin JB, Herman K, Houssin NS, Rich W, Reilly MA, Plageman TF. Arvcf Dependent Adherens Junction Stability is Required to Prevent Age-Related Cortical Cataracts. Front Cell Dev Biol 2022; 10:840129. [PMID: 35874813 PMCID: PMC9297370 DOI: 10.3389/fcell.2022.840129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
The etiology of age-related cortical cataracts is not well understood but is speculated to be related to alterations in cell adhesion and/or the changing mechanical stresses occurring in the lens with time. The role of cell adhesion in maintaining lens transparency with age is difficult to assess because of the developmental and physiological roles that well-characterized adhesion proteins have in the lens. This report demonstrates that Arvcf, a member of the p120-catenin subfamily of catenins that bind to the juxtamembrane domain of cadherins, is an essential fiber cell protein that preserves lens transparency with age in mice. No major developmental defects are observed in the absence of Arvcf, however, cortical cataracts emerge in all animals examined older than 6-months of age. While opacities are not obvious in young animals, histological anomalies are observed in lenses at 4-weeks that include fiber cell separations, regions of hexagonal lattice disorganization, and absence of immunolabeled membranes. Compression analysis of whole lenses also revealed that Arvcf is required for their normal biomechanical properties. Immunofluorescent labeling of control and Arvcf-deficient lens fiber cells revealed a reduction in membrane localization of N-cadherin, β-catenin, and αN-catenin. Furthermore, super-resolution imaging demonstrated that the reduction in protein membrane localization is correlated with smaller cadherin nanoclusters. Additional characterization of lens fiber cell morphology with electron microscopy and high resolution fluorescent imaging also showed that the cellular protrusions of fiber cells are abnormally elongated with a reduction and disorganization of cadherin complex protein localization. Together, these data demonstrate that Arvcf is required to maintain transparency with age by mediating the stability of the N-cadherin protein complex in adherens junctions.
Collapse
Affiliation(s)
- Jessica B. Martin
- College of Optometry, The Ohio State University, Columbus, OH, United States
| | - Kenneth Herman
- College of Optometry, The Ohio State University, Columbus, OH, United States
| | - Nathalie S. Houssin
- College of Optometry, The Ohio State University, Columbus, OH, United States
| | - Wade Rich
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Matthew A. Reilly
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
- Department of Ophthalmology and Visual Science, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Timothy F. Plageman
- College of Optometry, The Ohio State University, Columbus, OH, United States
- *Correspondence: Timothy F. Plageman Jr.,
| |
Collapse
|
16
|
Kuwata C, Maejima T, Hakamata S, Yahagi S, Matsuoka T, Tsuchiya Y. Disruption of Fetal Eye Development Caused by Insulin-induced Maternal Hypoglycemia in Rats. Reprod Toxicol 2022; 112:68-76. [PMID: 35738499 DOI: 10.1016/j.reprotox.2022.06.008] [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: 05/13/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 12/01/2022]
Abstract
We previously revealed that insulin-induced severe and long-lasting maternal hypoglycemia in rats caused anophthalmia and microphthalmia in fetuses; however, it remained unclear whether hypoglycemia-induced eye anomalies were developmental retardation or disruption, and when and how they developed. Hence, we induced hypoglycemia in pregnant Sprague-Dawley rats by injecting insulin from Days 6 to 11 of pregnancy and performed periodical histopathological examination of fetal eyes from embryonic days (E)10 to 20. On E10, optic vesicle had developed normally both in the control and insulin-treated group; however, on E11, optic cup (OC) had developed in the control group but not in the insulin-treated group. On E12, neural retina (NR), retinal pigmented epithelium (RPE), lens, and presumptive cornea had been observed in the control group. In contrast, lens pit and OC with remaining space between RPE and NR had developed in the insulin-treated group. From E13 to E15, developmental disruption characterized by defects, hypoplasia, and degeneration in the retina, lens, and cornea was observed in the insulin-treated group, resulting in anophthalmia or microphthalmia on E20. Moreover, the expression of MITF and chx10, which are essential for early eye development by expressing in the presumptive retina and lens and regulating each other's expression level, was ectopic and suppressed on E11. In conclusion, insulin-induced maternal hypoglycemia caused developmental disruption, but not simple developmental retardation of fetal eyes, and its trigger might be a failure of presumptive retina and lens to interact on E11.
Collapse
Affiliation(s)
- Chiharu Kuwata
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo, Japan.
| | - Takanori Maejima
- Translational Science, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, Japan
| | - Shinobu Hakamata
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo, Japan
| | - Satoko Yahagi
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo, Japan
| | - Toshiki Matsuoka
- Translational Science, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, Japan
| | - Yoshimi Tsuchiya
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo, Japan
| |
Collapse
|
17
|
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
|
18
|
Abstract
Developing organs are shaped, in part, by physical interaction with their environment in the embryo. In recent years, technical advances in live-cell imaging and material science have greatly expanded our understanding of the mechanical forces driving organ formation. Here, we provide a broad overview of the types of forces generated during embryonic development and then focus on a subset of organs underlying our senses: the eyes, inner ears, nose and skin. The epithelia in these organs emerge from a common origin: the ectoderm germ layer; yet, they arrive at unique and complex forms over developmental time. We discuss exciting recent animal studies that show a crucial role for mechanical forces in, for example, the thickening of sensory placodes, the coiling of the cochlea and the lengthening of hair. Finally, we discuss how microfabricated organoid systems can now provide unprecedented insights into the physical principles of human development.
Collapse
Affiliation(s)
- Anh Phuong Le
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Kim
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Karl R. Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Vigneux G, Pirkkanen J, Laframboise T, Prescott H, Tharmalingam S, Thome C. Radiation-Induced Alterations in Proliferation, Migration, and Adhesion in Lens Epithelial Cells and Implications for Cataract Development. Bioengineering (Basel) 2022; 9:29. [PMID: 35049738 PMCID: PMC8772889 DOI: 10.3390/bioengineering9010029] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
The lens of the eye is one of the most radiosensitive tissues. Although the exact mechanism of radiation-induced cataract development remains unknown, altered proliferation, migration, and adhesion have been proposed as factors. Lens epithelial cells were exposed to X-rays (0.1-2 Gy) and radiation effects were examined after 12 h and 7 day. Proliferation was quantified using an MTT assay, migration was measured using a Boyden chamber and wound-healing assay, and adhesion was assessed on three extracellular matrices. Transcriptional changes were also examined using RT-qPCR for a panel of genes related to these processes. In general, a nonlinear radiation response was observed, with the greatest effects occurring at a dose of 0.25 Gy. At this dose, a reduction in proliferation occurred 12 h post irradiation (82.06 ± 2.66%), followed by an increase at 7 day (116.16 ± 3.64%). Cell migration was increased at 0.25 Gy, with rates 121.66 ± 6.49% and 232.78 ± 22.22% greater than controls at 12 h and 7 day respectively. Cell adhesion was consistently reduced above doses of 0.25 Gy. Transcriptional alterations were identified at these same doses in multiple genes related to proliferation, migration, and adhesion. Overall, this research began to elucidate the functional changes that occur in lens cells following radiation exposure, thereby providing a better mechanistic understanding of radiation-induced cataract development.
Collapse
Affiliation(s)
- Graysen Vigneux
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (G.V.); (S.T.)
| | - Jake Pirkkanen
- Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (J.P.); (T.L.); (H.P.)
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
| | - Taylor Laframboise
- Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (J.P.); (T.L.); (H.P.)
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
| | - Hallie Prescott
- Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (J.P.); (T.L.); (H.P.)
| | - Sujeenthar Tharmalingam
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (G.V.); (S.T.)
- Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (J.P.); (T.L.); (H.P.)
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
- Nuclear Innovation Institute, 620 Tomlinson Drive, Port Elgin, ON N0H 2C0, Canada
| | - Christopher Thome
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (G.V.); (S.T.)
- Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada; (J.P.); (T.L.); (H.P.)
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
- Nuclear Innovation Institute, 620 Tomlinson Drive, Port Elgin, ON N0H 2C0, Canada
| |
Collapse
|
20
|
Karnam S, Maddala R, Stiber JA, Rao PV. Drebrin, an actin-binding protein, is required for lens morphogenesis and growth. Dev Dyn 2021; 250:1600-1617. [PMID: 33896079 PMCID: PMC8542647 DOI: 10.1002/dvdy.353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Lens morphogenesis, architecture, and clarity are known to be critically dependent on actin cytoskeleton organization and cell adhesive interactions. There is limited knowledge, however regarding the identity and role of key proteins regulating actin cytoskeletal organization in the lens. This study investigated the role of drebrin, a developmentally regulated actin-binding protein, in mouse lens development by generating and characterizing a conditional knockout (cKO) mouse model using the Cre-LoxP recombination approach. RESULTS Drebrin E, a splice variant of DBN1 is a predominant isoform expressed in the mouse lens and exhibits a maturation-dependent downregulation. Drebrin co-distributes with actin in both epithelium and fibers. Conditional deficiency (both haploinsufficiency and complete absence) of drebrin results in disrupted lens morphogenesis leading to cataract and microphthalmia. The drebrin cKO lens reveals a dramatic decrease in epithelial height and width, E-cadherin, and proliferation, and increased apoptotic cell death and expression of α-smooth muscle actin, together with severely impaired fiber cell organization, polarity, and cell-cell adhesion. CONCLUSIONS This study demonstrates the requirement of drebrin in lens development and growth, with drebrin deficiency leading to impaired lens morphogenesis and microphthalmia.
Collapse
Affiliation(s)
- Shruthi Karnam
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
| | - Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
| | - Jonathan A Stiber
- Department of Medicine, Duke University School of Medicine, Durham, NC. USA
| | - Ponugoti V Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC. USA
| |
Collapse
|
21
|
Harding P, Toms M, Schiff E, Owen N, Bell S, Lloyd IC, Moosajee M. EPHA2 Segregates with Microphthalmia and Congenital Cataracts in Two Unrelated Families. Int J Mol Sci 2021; 22:2190. [PMID: 33671840 PMCID: PMC7926380 DOI: 10.3390/ijms22042190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
EPHA2 is a transmembrane tyrosine kinase receptor that, when disrupted, causes congenital and age-related cataracts. Cat-Map reports 22 pathogenic EPHA2 variants associated with congenital cataracts, variable microcornea, and lenticonus, but no previous association with microphthalmia (small, underdeveloped eye, ≥2 standard deviations below normal axial length). Microphthalmia arises from ocular maldevelopment with >90 monogenic causes, and can include a complex ocular phenotype. In this paper, we report two pathogenic EPHA2 variants in unrelated families presenting with bilateral microphthalmia and congenital cataracts. Whole genome sequencing through the 100,000 Genomes Project and cataract-related targeted gene panel testing identified autosomal dominant heterozygous mutations segregating with the disease: (i) missense c.1751C>T, p.(Pro584Leu) and (ii) splice site c.2826-9G>A. To functionally validate pathogenicity, morpholino knockdown of epha2a/epha2b in zebrafish resulted in significantly reduced eye size ± cataract formation. Misexpression of N-cadherin and retained fibre cell nuclei were observed in the developing lens of the epha2b knockdown morphant fish by 3 days post-fertilisation, which indicated a putative mechanism for microphthalmia pathogenesis through disruption of cadherin-mediated adherens junctions, preventing lens maturation and the critical signals stimulating eye growth. This study demonstrates a novel association of EPHA2 with microphthalmia, suggesting further analysis of pathogenic variants in unsolved microphthalmia cohorts may increase molecular diagnostic rates.
Collapse
Affiliation(s)
- Philippa Harding
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK; (P.H.); (M.T.); (N.O.)
| | - Maria Toms
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK; (P.H.); (M.T.); (N.O.)
- The Francis Crick Institute, London NW1 1AT, UK
| | - Elena Schiff
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK; (E.S.); (S.B.)
| | - Nicholas Owen
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK; (P.H.); (M.T.); (N.O.)
| | - Suzannah Bell
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK; (E.S.); (S.B.)
| | - Ian Christopher Lloyd
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK;
- Manchester Academic Health Sciences Centre, University of Manchester, Manchester, M13 9PT, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Mariya Moosajee
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK; (P.H.); (M.T.); (N.O.)
- The Francis Crick Institute, London NW1 1AT, UK
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK; (E.S.); (S.B.)
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| |
Collapse
|
22
|
Long-term myofibroblast persistence in the capsular bag contributes to the late spontaneous in-the-bag intraocular lens dislocation. Sci Rep 2020; 10:20532. [PMID: 33239706 PMCID: PMC7689492 DOI: 10.1038/s41598-020-77207-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Late spontaneous in-the-bag intraocular lens (IOL) dislocation is a complication presenting 6 months or later after cataract surgery. We aimed to characterize the cells in the lens capsules (LCs) of 18 patients with spontaneous late in-the-bag IOL dislocation. Patients' average age was 82.6 ± 1.5 years (range 72-98), and most of them had pseudoexfoliation syndrome (PEX). Cells from the LCs were positive for myofibroblast (αSMA), proliferation (Ki-67, PCNA), early lens development/lens progenitor (SOX2, PAX6), chemokine receptor (CXCR4), and transmembrane (N-cadherin) markers, while negative for epithelial (E-cadherin) marker. Moreover, the cells produced abundant fibronectin, type I and type V collagen in the nearby extracellular matrix (ECM). During ex vivo cultivation of dislocated IOL-LCs in toto, the cells proliferated and likely migrated onto the IOL's anterior side. EdU proliferation assay confirmed the proliferation potential of the myofibroblasts (MFBs) in dislocated IOL-LCs. Primary cultured lens epithelial cells/MFBs isolated from the LC of dislocated IOLs could induce collagen matrix contraction and continuously proliferated, migrated, and induced ECM remodeling. Taken together, this indicates that long-lived MFBs of dislocated IOLs might contribute to the pathogenic mechanisms in late in-the-bag IOL dislocation.
Collapse
|
23
|
Piedade WP, Titialii-Torres K, Morris AC, Famulski JK. Proteasome-Mediated Regulation of Cdhr1a by Siah1 Modulates Photoreceptor Development and Survival in Zebrafish. Front Cell Dev Biol 2020; 8:594290. [PMID: 33330480 PMCID: PMC7719784 DOI: 10.3389/fcell.2020.594290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/22/2020] [Indexed: 01/05/2023] Open
Abstract
Congenital retinal dystrophies are a major cause of unpreventable and incurable blindness worldwide. Mutations in CDHR1, a retina specific cadherin, are associated with cone-rod dystrophy. The ubiquitin proteasome system (UPS) is responsible for mediating orderly and precise targeting of protein degradation to maintain biological homeostasis and coordinate proper development, including retinal development. Recently, our lab uncovered that the seven in absentia (Siah) family of E3 ubiquitin ligases play a role in optic fissure fusion and identified Cdhr1a as a potential target of Siah. Using two-color whole mount in situ hybridization and immunohistochemistry, we detected siah1 and cdhr1a co-expression as well as protein localization in the retinal outer nuclear layer (ONL), and more precisely in the connecting cilium of rods and cones between 3-5 days post fertilization (dpf). We confirmed that Siah1 targets Cdhr1a for proteasomal degradation by co-transfection and co-immunoprecipitation in cell culture. To analyze the functional importance of this interaction, we created two transgenic zebrafish lines that express siah1 or an inactive siah1 (siah1ΔRING) under the control of the heat shock promoter to modulate Siah activity during photoreceptor development. Overexpression of siah1, but not siah1ΔRING, resulted in a decrease in the number of rods and cones at 72 h post fertilization (hpf). The number of retinal ganglion cells, amacrine and bipolar cells was not affected by Siah1 overexpression, and there was no significant reduction of proliferating cells in the Siah1 overexpressing retina. We did, however, detect increased cell death, confirmed by an increase in the number of TUNEL + cells in the ONL, which was proteasome-dependent, as proteasome inhibition rescued the cell death phenotype. Furthermore, reduction in rods and cones resulting from increased Siah1 expression was rescued by injection of cdhr1a mRNA, and to an even greater extent by injection of a Siah1-insensitive cdhr1a variant mRNA. Lastly, CRISPR induced loss of Cdhr1a function phenocopied Siah1 overexpression resulting in a significant reduction of rods and cones. Taken together, our work provides the first evidence that Cdhr1a plays a role during early photoreceptor development and that Cdhr1a is regulated by Siah1 via the UPS.
Collapse
Affiliation(s)
| | | | | | - Jakub K. Famulski
- Department of Biology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
24
|
Wang R, Lu X, Yu R. Lycopene Inhibits Epithelial-Mesenchymal Transition and Promotes Apoptosis in Oral Cancer via PI3K/AKT/m-TOR Signal Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2461-2471. [PMID: 32606612 PMCID: PMC7321693 DOI: 10.2147/dddt.s251614] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
Background Oral cancer (OC) is one of the most common cancers around the world. Despite the progress in treatment, the prognosis of OC remains poor, especially for patients with advanced diseases. It urges the development of novel therapeutic options against OC. Lycopene (LYC) is an antioxidant with chemoprotective properties against cancer. However, little is known about the mechanisms underlying the protective role of LYC in OC tumorigenesis. Methods In this study, we investigated the anti-cancer effect of LYC on the progression of OC in vitro and in vivo and explored the underlying mechanisms involved in this process. Results LYC inhibited OC cell proliferation, migration, invasion, apoptosis, and xenograft tumor growth in a dose-dependent manner. Furthermore, we found that LYC might inhibit epithelial-mesenchymal transition and induce apoptosis in OC cells by deactivating the PI3K/AKT/m-TOR signaling through increasing the levels of E-cadherin and Bax and downregulating N-cadherin, p-PI3K, p-AKT, p-m-TOR, and bcl-2. Conclusion We reported for the first time that LYC exhibited anti-cancer effects on OC development both in vitro and in vivo via regulating EMT process and apoptosis. These findings provide support for the potential clinical use of LYC in OC treatment.
Collapse
Affiliation(s)
- Ran Wang
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xinxing Lu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Riyue Yu
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
25
|
Shukal D, Bhadresha K, Shastri B, Mehta D, Vasavada A, Johar K. Dichloroacetate prevents TGFβ-induced epithelial-mesenchymal transition of retinal pigment epithelial cells. Exp Eye Res 2020; 197:108072. [PMID: 32473169 DOI: 10.1016/j.exer.2020.108072] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022]
Abstract
Proliferative retinopathies are associated with formation of fibrous epiretinal membranes. At present, there is no pharmacological intervention for the treatment of retinopathies. Cytokines such as TGFβ are elevated in the vitreous humor of the patients with proliferative vitro-retinopathy, diabetic retinopathy and age-related macular degeneration. TGFβ isoforms lead to epithelial-mesenchymal transition (EMT) or trans-differentiation of the retinal pigment epithelial (RPE) cells. PI3K/Akt and MAPK/Erk pathways play important roles in the EMT of RPE cells. Therefore, inhibition of EMT by pharmacological agents is an important therapeutic strategy in retinopathy. Dichloroacetate (DCA) is shown to prevent proliferation and EMT of cancer cell lines but its effects are not explored on the prevention of EMT of RPE cells. In the present study, we have investigated the role of DCA in preventing TGFβ2 induced EMT of RPE cell line, ARPE-19. A wound-healing assay was utilized to detect the anti-EMT effect of DCA. The expressions of EMT and cell adhesion markers were carried out by immunofluorescence, western blotting, and quantitative real-time PCR. The expression of MAPK/Erk and PI3K/Akt pathway members was carried out using western blotting. We found that TGFβ2 exposure leads to an increase in the wound healing response, expression of EMT markers (Fibronectin, Collagen I, N-cadherin, MMP9, S100A4, α-SMA, Snai1, Slug) and a decrease in the expression of cell adhesion/epithelial markers (ZO-1, Connexin 43, E-cadherin). These changes were accompanied by the activation of PI3K/Akt and MAPK/Erk pathways. Simultaneous exposure of DCA along with TGFβ2 significantly inhibited wound healing response, expression of EMT markers and cell adhesion/epithelial markers. Furthermore, DCA and TGFβ2 effectively attenuated the activation of MAPK/Erk/JNK and PI3K/Akt/GSK3β pathways. Our results demonstrate that DCA has a strong anti-EMT effect on the ARPE-19 cells and hence can be utilized as a therapeutic agent in the prevention of proliferative retinopathies.
Collapse
Affiliation(s)
- Dhaval Shukal
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat, India; Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - Kinjal Bhadresha
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat, India.
| | - Bhoomi Shastri
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat, India.
| | - Deval Mehta
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat, India.
| | - Abhay Vasavada
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat, India.
| | - Kaid Johar
- Department of Zoology, BMTC, Human Genetics, USSC, Gujarat University, Ahmedabad, Gujarat, India.
| |
Collapse
|
26
|
Maddala R, Mongan M, Xia Y, Rao PV. Calponin-3 deficiency augments contractile activity, plasticity, fibrogenic response and Yap/Taz transcriptional activation in lens epithelial cells and explants. Sci Rep 2020; 10:1295. [PMID: 31992794 PMCID: PMC6987178 DOI: 10.1038/s41598-020-58189-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 01/09/2020] [Indexed: 01/19/2023] Open
Abstract
The transparent ocular lens plays a crucial role in vision by focusing light on to the retina with loss of lens transparency leading to impairment of vision. While maintenance of epithelial phenotype is recognized to be essential for lens development and function, knowledge of the identity of different molecular mechanisms regulating lens epithelial characteristics remains incomplete. This study reports that CNN-3, the acidic isoform of calponin, an actin binding contractile protein, is expressed preferentially and abundantly relative to the basic and neutral isoforms of calponin in the ocular lens, and distributes predominantly to the epithelium in both mouse and human lenses. Expression and MEKK1-mediated threonine 288 phosphorylation of CNN-3 is induced by extracellular cues including TGF-β2 and lysophosphatidic acid. Importantly, siRNA-induced deficiency of CNN3 in lens epithelial cell cultures and explants results in actin stress fiber reorganization, stimulation of focal adhesion formation, Yap activation, increases in the levels of α-smooth muscle actin, connective tissue growth factor and fibronectin, and decreases in E-cadherin expression. These results reveal that CNN3 plays a crucial role in regulating lens epithelial contractile activity and provide supporting evidence that CNN-3 deficiency is associated with the induction of epithelial plasticity, fibrogenic activity and mechanosensitive Yap/Taz transcriptional activation.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maureen Mongan
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ying Xia
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA. .,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
27
|
Gagnoux-Palacios L, Hueber AO. [Cell death for maintaining epithelial homeostasis]. Med Sci (Paris) 2019; 35:830-833. [PMID: 31845871 DOI: 10.1051/medsci/2019164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Laurent Gagnoux-Palacios
- Université Côte d'Azur, Institut de biologie Valrose, CNRS, Inserm, Parc Valrose, Bâtiment des sciences naturelles, 28 avenue de Valrose, 06108 Nice Cedex 2, France
| | - Anne-Odile Hueber
- Université Côte d'Azur, Institut de biologie Valrose, CNRS, Inserm, Parc Valrose, Bâtiment des sciences naturelles, 28 avenue de Valrose, 06108 Nice Cedex 2, France
| |
Collapse
|
28
|
Reis LM, Houssin NS, Zamora C, Abdul-Rahman O, Kalish JM, Zackai EH, Plageman TF, Semina EV. Novel variants in CDH2 are associated with a new syndrome including Peters anomaly. Clin Genet 2019; 97:502-508. [PMID: 31650526 DOI: 10.1111/cge.13660] [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: 09/13/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 11/26/2022]
Abstract
Peters anomaly (PA) is a congenital corneal opacity associated with corneo-lenticular attachments. PA can be isolated or part of a syndrome with most cases remaining genetically unsolved. Exome sequencing of a trio with syndromic PA and 145 additional unexplained probands with developmental ocular conditions identified a de novo splicing and three novel missense heterozygous CDH2 variants affecting the extracellular cadherin domains in four individuals with PA. Syndromic anomalies were seen in three individuals and included left-sided cardiac lesions, dysmorphic facial features, and decreasing height percentiles; brain magnetic resonance imaging identified agenesis of the corpus callosum and hypoplasia of the inferior cerebellar vermis. CDH2 encodes for N-cadherin, a transmembrane protein that mediates cell-cell adhesion in multiple tissues. Immunostaining in mouse embryonic eyes confirmed N-cadherin is present in the lens stalk at the time of separation from the future cornea and in the developing lens and corneal endothelium at later stages, supporting a possible role in PA. Previous studies in animal models have noted the importance of Cdh2/cdh2 in the development of the eye, heart, brain, and skeletal structures, also consistent with the patient features presented here. Examination of CDH2 in additional patients with PA is indicated to confirm this association.
Collapse
Affiliation(s)
- Linda M Reis
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Carlos Zamora
- Department of Radiology, Division of Neuroradiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Omar Abdul-Rahman
- Genetic Medicine, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jennifer M Kalish
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elaine H Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | | | - Elena V Semina
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Ophthalmology and Visual Sciences, Department of Cell Biology, Neurobiology and Anatomy, Children's Research Institute, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
29
|
Joseph R, Bales K, Srivastava K, Srivastava O. Lens epithelial cells-induced pluripotent stem cells as a model to study epithelial-mesenchymal transition during posterior capsular opacification. Biochem Biophys Rep 2019; 20:100696. [PMID: 31681860 PMCID: PMC6818140 DOI: 10.1016/j.bbrep.2019.100696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 10/02/2019] [Accepted: 10/11/2019] [Indexed: 12/11/2022] Open
Abstract
The overall goal was to generate an epithelial-mesenchymal transition (EMT) model using lens epithelial cells-induced pluripotent stem cells to elucidate EMT-regulatory factors during posterior capsular opacification (PCO). For this purpose, the mouse lens epithelial cells-derived mesenchymal cells were reprogrammed to induced pluripotent stem cells (iPSC) and differentiated to lens epithelial cells to be used to determine regulatory factors during EMT. Lens epithelial cells from one-month-old C57BL/6 mice were transitioned to mesenchymal cells in culture, and were reprogrammed to iPSC by delivering reprogramming factors in a single polycistronic lentiviral vector (co-expressing four transcription factors, Oct 4, Sox2, Klf4, and Myc). iPSC were differentiated to epithelial cells by a three-step process using noggin, basic fibroblast growth factor (bFGF), bone morphogenetic protein 4 (BMP4) and Wnt-3. At various time points, the cells/clones were immunocytochemically analyzed for epithelial cell markers (Connexin-43 and E-cadherin), mesenchymal cell markers (Alpha-smooth muscle actin), stem cell markers (Sox1, Oct4, SSEA4 and Tra60) and lens-specific epithelial cell markers (αA- and βA3/A1-crystallins). By increasing the number of genetic transductions, the time needed for generating iPSC from lens mesenchymal cells was reduced, successfully reprogrammed epithelial/mesenchymal cells into iPSC, and retransformed iPSC into lens epithelial cells by the growth factors’ treatment. The epithelial cells could serve as a model system to elucidate regulatory factors involved during EMT to therapeutically stop it. By increasing the number of genetic transductions, reduced the time needed for generating iPSC from lens mesenchymal cells. We successfully reprogrammed iPSC, and also differentiated iPSC into lens epithelial cells by the growth factors. Our model could elucidate regulatory factors involved in epithelial mesenchymal transition to therapeutically stop it.
Collapse
Affiliation(s)
| | | | | | - Om Srivastava
- Corresponding author. Department of Optometry and Vision Science, University of Alabama at Birmingham, 1716 University Boulevard, HPB-437, Birmingham, AL, 35294-0010, USA.
| |
Collapse
|
30
|
Kumari S, Taginik G, Varadaraj S, Varadaraj K. Positively charged amino acid residues in the extracellular loops A and C of lens aquaporin 0 interact with the negative charges in the plasma membrane to facilitate cell-to-cell adhesion. Exp Eye Res 2019; 185:107682. [PMID: 31150637 DOI: 10.1016/j.exer.2019.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 04/17/2019] [Accepted: 05/27/2019] [Indexed: 01/12/2023]
Abstract
This investigation was undertaken to find out whether the positive charges in the Extracellular Loops A (ELA) and C (ELC) of Aquaporin 0 (AQP0) are involved in lens fiber cell-to-cell adhesion (CTCA), and the possible mechanism of CTCA. AQP0 ELA or ELC was substituted with the corresponding AQP1 loop via Polymerase Chain Reaction. Positively charged arginine (R) and histidine (H) of mouse AQP0 ELA and ELC were substituted individually with glutamine (Q) to create R33Q, H40Q, R113Q and H122Q by mutagenesis. cRNA expression, immunostaining, Förster Resonance Energy Transfer (FRET) studies and protein analyses showed localization of all mutants except AQP0-AQP1ELC chimera (AQP0 ELC substituted with AQP1 ELC) at the plasma membrane. Osmotic Swelling Assay revealed comparable water permeability (Pf) among AQP0-AQP1ELA, R33Q, R113Q, and WT. CTCA assay demonstrated a significant reduction in adhesion in all mutants compared to the WT (14-73%) suggesting the importance of the conserved positively charged residues of ELA and ELC for adhesion. Studies involving AQP0-transfected L-cells, and lipid vesicles indicated that CTCA was due to the electrostatic interaction between the positively charged amino acids of AQP0 extracellular loops and the negative charges of the plasma membrane. Schematic models are provided to illustrate the mechanism.
Collapse
Affiliation(s)
- Sindhu Kumari
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794-8661, USA
| | - Gozde Taginik
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794-8661, USA
| | - Sangeeth Varadaraj
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794-8661, USA
| | - Kulandaiappan Varadaraj
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794-8661, USA; SUNY Eye Institute, New York, NY, USA.
| |
Collapse
|
31
|
Abstract
This chapter provides an overview of the early developmental origins of six ocular tissues: the cornea, lens, ciliary body, iris, neural retina, and retina pigment epithelium. Many of these tissue types are concurrently specified and undergo a complex set of morphogenetic movements that facilitate their structural interconnection. Within the context of vertebrate eye organogenesis, we also discuss the genetic hierarchies of transcription factors and signaling pathways that regulate growth, patterning, cell type specification and differentiation.
Collapse
Affiliation(s)
- Joel B Miesfeld
- Department of Cell Biology & Human Anatomy, University of California Davis School of Medicine, Davis, CA, United States
| | - Nadean L Brown
- Department of Cell Biology & Human Anatomy, University of California Davis School of Medicine, Davis, CA, United States.
| |
Collapse
|
32
|
Ankyrin-G regulated epithelial phenotype is required for mouse lens morphogenesis and growth. Dev Biol 2018; 446:119-131. [PMID: 30562487 DOI: 10.1016/j.ydbio.2018.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 11/24/2022]
Abstract
Epithelial cell polarity, adhesion, proliferation, differentiation and survival are essential for morphogenesis of various organs and tissues including the ocular lens. The molecular mechanisms regulating the lens epithelial phenotype however, are not well understood. Here we investigated the role of scaffolding protein ankyrin-G (AnkG) in mouse lens development by conditional suppression of AnkG expression using the Cre-LoxP recombination approach. AnkG, which serves to link integral membrane proteins to the spectrin/actin cytoskeleton, was found to distribute predominantly to the lateral membranes of lens epithelium with several isoforms of the protein being detected in the mouse lens. Conditional deficiency of AnkG impaired mouse lens morphogenesis starting from embryonic stage E15.5, with neonatal (P1) AnkG cKO lenses exhibiting overt abnormalities in shape, size, epithelial cell height, sheet length and lateral membrane assembly together with defective fiber cell orientation relative to lenses from littermate AnkG floxed or Cre expressing mice. Severe disruptions in E-cadherin/β-catenin-based adherens junctions, and the membrane organization of spectrin-actin cytoskeleton, ZO-1, connexin-50 and Na+-K+-ATPase were noted in AnkG deficient lenses, along with detection in lens epithelium of α-smooth muscle actin, a marker of epithelial to mesenchymal transition. Moreover, lens epithelial cell proliferation and survival were severely compromised while differentiation appears to be normal in AnkG deficient mouse lenses. Collectively, these results indicate that AnkG regulates establishment of the epithelial phenotype via lateral membrane assembly, stabilization of E-cadherin-based cell-cell junctions, polarity and membrane organization of transport and adhesion proteins and the spectrin-actin skeleton, and provide evidence for an obligatory role for AnkG in lens morphogenesis and growth.
Collapse
|
33
|
Azimi M, Le TT, Brown NL. Presenilin gene function and Notch signaling feedback regulation in the developing mouse lens. Differentiation 2018; 102:40-52. [PMID: 30059908 DOI: 10.1016/j.diff.2018.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/06/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022]
Abstract
Presenilins (Psen1 and Psen2 in mice) are polytopic transmembrane proteins that act in the γ-secretase complex to make intra-membrane cleavages of their substrates, including the well-studied Notch receptors. Such processing releases the Notch intracellular domain, allowing it to physically relocate from the cell membrane to the nucleus where it acts in a transcriptional activating complex to regulate downstream genes in the signal-receiving cell. Previous studies of Notch pathway mutants for Jagged1, Notch2, and Rbpj demonstrated that canonical signaling is a necessary component of normal mouse lens development. However, the central role of Psens within the γ-secretase complex has never been explored in any developing eye tissue or cell type. By directly comparing Psen single and double mutant phenotypes during mouse lens development, we found a stronger requirement for Psen1, although both genes are needed for progenitor cell growth and to prevent apoptosis. We also uncovered a novel genetic interaction between Psen1 and Jagged1. By quantifying protein and mRNA levels of key Notch pathway genes in Psen1/2 or Jagged1 mutant lenses, we identified multiple points in the overall signaling cascade where feedback regulation can occur. Our data are consistent with the loss of particular genes indirectly influencing the transcription level of another. However, we conclude that regulating Notch2 protein levels is particularly important during normal signaling, supporting the importance of post-translational regulatory mechanisms in this tissue.
Collapse
Affiliation(s)
- Mina Azimi
- Department of Cell Biology & Human Anatomy; University of California, Davis One Shields Avenue, Davis, CA 95616, USA
| | - Tien T Le
- Division of Developmental Biology, Cincinnati Childrens Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Nadean L Brown
- Department of Cell Biology & Human Anatomy; University of California, Davis One Shields Avenue, Davis, CA 95616, USA; Division of Developmental Biology, Cincinnati Childrens Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
34
|
Chauhan BK, Medsinge A, Baumgartner MP, Scanga HL, Kamakari S, Gajdosova E, Camacho CJ, Nischal KK. Case series: Pyramidal cataracts, intact irides and nystagmus from three novel PAX6 mutations. Am J Ophthalmol Case Rep 2018; 10:172-179. [PMID: 29780932 PMCID: PMC5956696 DOI: 10.1016/j.ajoc.2018.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/07/2018] [Accepted: 02/26/2018] [Indexed: 11/29/2022] Open
Abstract
Purpose To investigate the association between novel PAX6 mutations to bilateral anterior pyramidal congenital cataracts (APyC), complete and intact irides, and nystagmus. Observations This is a retrospective observational case series in a multi-center setting with genetic testing. Three female patients were diagnosed with bilateral APyC, intact irides and nystagmus. Genetic testing identified the three patients had novel missense mutations in PAX6 – c.128C > T; p.Ser43Phe (S43F), c. 197T > A; p.Ile66Asn (I66N) and c.781C > G; p.Arg261Gly (R261G). Conclusions and importance This study demonstrates a novel phenotype of bilateral APyC, intact irides, and nystagmus in whom genetic testing for PAX6 identified novel missense mutations (S43F, I66N, R261G) in highly conserved DNA-binding domains. Implications of understanding why the iris is present in these cases is discussed.
Collapse
Affiliation(s)
- Bharesh K Chauhan
- UPMC Eye Center, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA.,Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Anagha Medsinge
- UPMC Eye Center, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Matthew P Baumgartner
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Hannah L Scanga
- UPMC Eye Center, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Smaragda Kamakari
- Ophthalmic Genetics Unit, OMMA, Ophthalmological Institute of Athens, Katehaki 74, 11525, Athens, Greece
| | - Eva Gajdosova
- Great Ormond Street Hospital for Children, London, WC1N 3JH, UK
| | - Carlos J Camacho
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ken K Nischal
- UPMC Eye Center, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA.,Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
35
|
Collins TN, Mao Y, Li H, Bouaziz M, Hong A, Feng GS, Wang F, Quilliam LA, Chen L, Park T, Curran T, Zhang X. Crk proteins transduce FGF signaling to promote lens fiber cell elongation. eLife 2018; 7:32586. [PMID: 29360039 PMCID: PMC5818251 DOI: 10.7554/elife.32586] [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: 10/07/2017] [Accepted: 01/23/2018] [Indexed: 12/17/2022] Open
Abstract
Specific cell shapes are fundamental to the organization and function of multicellular organisms. Fibroblast Growth Factor (FGF) signaling induces the elongation of lens fiber cells during vertebrate lens development. Nonetheless, exactly how this extracellular FGF signal is transmitted to the cytoskeletal network has previously not been determined. Here, we show that the Crk family of adaptor proteins, Crk and Crkl, are required for mouse lens morphogenesis but not differentiation. Genetic ablation and epistasis experiments demonstrated that Crk and Crkl play overlapping roles downstream of FGF signaling in order to regulate lens fiber cell elongation. Upon FGF stimulation, Crk proteins were found to interact with Frs2, Shp2 and Grb2. The loss of Crk proteins was partially compensated for by the activation of Ras and Rac signaling. These results reveal that Crk proteins are important partners of the Frs2/Shp2/Grb2 complex in mediating FGF signaling, specifically promoting cell shape changes. As an embryo develops, its cells divide multiple times to transform into the specialized cell types that form our tissues and organs. To carry out specific roles, cells need to be of a certain shape. For example, in mammals, the cells that make up the main portion of the eye lens, develop into a fiber-like shape to be perfectly aligned with each other. This enables them to transmit light to the retina at the rear end of the eye. To do so, the lens cells increase over 1000 times in length with the help of a group of proteins called the Fibroblast Growth Factor, or FGF for short. The FGF pathway includes a network of interacting proteins that transmit signals to molecules inside the lens cells to control how they specialize and grow. However, until now it was not clear how it does this. Here, Zhang et al. used mouse lens-cells grown in the laboratory to investigate how FGF signaling causes cells to change their structure. The experiments revealed two related proteins called Crk and Crkl that linked the FGF pathway with another signaling system. When these two proteins were removed from the lens cells, the lens cells were still able to specialize, but could no longer grow in length. This suggests that these two processes are independent of each other. Moreover, Crk and Crkl helped the cells to change shape by increasing the amount of another group of proteins called Ras, which are known to both help cells to specialize and to regulate their shape. Zhang et al. discovered that the amount of Ras proteins determined whether cells specialized or modified their shape by changing the organization of proteins in the cell. Millions of children are born with cataracts, a disease caused when lens cells fail to shape properly. A better knowledge of FGF signaling may help to understand how cataracts develop and inspire future treatments. Moreover, the pathways identified in this study could also apply to other organs and diseases in which FGF signaling is active.
Collapse
Affiliation(s)
- Tamica N Collins
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, United States
| | - Yingyu Mao
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, United States
| | - Hongge Li
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, United States
| | - Michael Bouaziz
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, United States
| | - Angela Hong
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, United States
| | - Gen-Sheng Feng
- Department of Pathology, University of California San Diego, La Jolla, United States
| | - Fen Wang
- Center for Cancer Biology and Nutrition, Houston, United States
| | - Lawrence A Quilliam
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, United States
| | - Lin Chen
- Department of Rehabilitation Medicine, Third Military Medical University, Chongqing, China
| | - Taeju Park
- The Children's Research Institute, Children's Mercy Kansas City, Kansas City, United States
| | - Tom Curran
- The Children's Research Institute, Children's Mercy Kansas City, Kansas City, United States
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, United States
| |
Collapse
|
36
|
Logan CM, Bowen CJ, Menko AS. Induction of Immune Surveillance of the Dysmorphogenic Lens. Sci Rep 2017; 7:16235. [PMID: 29176738 PMCID: PMC5701161 DOI: 10.1038/s41598-017-16456-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/12/2017] [Indexed: 01/10/2023] Open
Abstract
The lens has been considered to be an immune privileged site not susceptible to the immune processes normally associated with tissue injury and wound repair. However, as greater insight into the immune surveillance process is gained, we have reevaluated the concept of immune privilege. Our studies using an N-cadherin lens-specific conditional knockout mouse, N-cadΔlens, show that loss of this cell-cell junctional protein leads to lens degeneration, necrosis and fibrotic change, postnatally. The degeneration of this tissue induces an immune response resulting in immune cells populating the lens that contribute to the development of fibrosis. Additionally, we demonstrate that the lens is connected to the lymphatic system, with LYVE(+) labeling reaching the lens along the suspensory ligaments that connect the lens to the ciliary body, providing a potential mechanism for the immune circulation. Importantly, we observe that degeneration of the lens activates an immune response throughout the eye, including cornea, vitreous humor, and retina, suggesting a coordinated protective response in the visual system to defects of a component tissue. These studies demonstrate that lens degeneration induces an immune response that can contribute to the fibrosis that often accompanies lens dysgenesis, a consideration for understanding organ system response to injury.
Collapse
Affiliation(s)
- Caitlin M Logan
- Thomas Jefferson University, Department of Pathology, Anatomy and Cell Biology, Philadelphia, Pennsylvania, 19107, United States
| | - Caitlin J Bowen
- Thomas Jefferson University, Department of Pathology, Anatomy and Cell Biology, Philadelphia, Pennsylvania, 19107, United States
| | - A Sue Menko
- Thomas Jefferson University, Department of Pathology, Anatomy and Cell Biology, Philadelphia, Pennsylvania, 19107, United States.
| |
Collapse
|
37
|
Mochizuki T, Luo YJ, Tsai HF, Hagiwara A, Masai I. Cell division and cadherin-mediated adhesion regulate lens epithelial cell movement in zebrafish. Development 2017; 144:708-719. [PMID: 28196805 DOI: 10.1242/dev.138909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/04/2017] [Indexed: 11/20/2022]
Abstract
In vertebrates, lens epithelial cells cover the anterior half of the lens fiber core. During development, lens epithelial cells proliferate, move posteriorly and differentiate into lens fiber cells after passing through the equator. To elucidate the mechanisms underlying lens epithelial cell movement, we conducted time-lapse imaging of zebrafish lens epithelium. Lens epithelial cells do not intermingle but maintain their relative positions during development. Cell division induces epithelial rearrangement, which subsequently promotes cell movement towards the equator. These data suggest that cell division is the major driving force for cell movement. In zebrafish, E-cadherin is expressed in lens epithelium, whereas N-cadherin is required for lens fiber growth. E-cadherin reduced lens epithelial cell movement, whereas N-cadherin enhanced it. Laser ablation experiments revealed that lens epithelium is governed by pulling tension, which is modulated by these cadherins. Thus, cell division and cadherin-mediated adhesion regulate lens epithelial cell movement via modulation of epithelial tension.
Collapse
Affiliation(s)
- Toshiaki Mochizuki
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Yi-Jyun Luo
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Hsieh-Fu Tsai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Akane Hagiwara
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| |
Collapse
|
38
|
Cvekl A, Zhang X. Signaling and Gene Regulatory Networks in Mammalian Lens Development. Trends Genet 2017; 33:677-702. [PMID: 28867048 DOI: 10.1016/j.tig.2017.08.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 11/16/2022]
Abstract
Ocular lens development represents an advantageous system in which to study regulatory mechanisms governing cell fate decisions, extracellular signaling, cell and tissue organization, and the underlying gene regulatory networks. Spatiotemporally regulated domains of BMP, FGF, and other signaling molecules in late gastrula-early neurula stage embryos generate the border region between the neural plate and non-neural ectoderm from which multiple cell types, including lens progenitor cells, emerge and undergo initial tissue formation. Extracellular signaling and DNA-binding transcription factors govern lens and optic cup morphogenesis. Pax6, c-Maf, Hsf4, Prox1, Sox1, and a few additional factors regulate the expression of the lens structural proteins, the crystallins. Extensive crosstalk between a diverse array of signaling pathways controls the complexity and order of lens morphogenetic processes and lens transparency.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
39
|
Huang L, Jiang J, Guo Q, Yang H. E‑cadherin involvement in human lens epithelial cell transdifferentiation may be associated with N‑cadherin. Mol Med Rep 2017; 16:5031-5035. [PMID: 28765930 DOI: 10.3892/mmr.2017.7132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 05/04/2017] [Indexed: 11/06/2022] Open
Abstract
E-cadherin, β-catenin and N‑cadherin serve key roles in the epithelial‑to‑mesenchymal transition (EMT) that leads to human lens epithelial cell (LEC) transdifferentiation and subsequent cataract formation. The present study aimed to investigate the role of E‑cadherin in LEC transdifferentiation. SRA01/04 human LECs were transfected with E‑cadherin short interfering (si)RNA (E‑cadherin siRNA group), negative control siRNA (NC group) or the transfection regent Lipofectamine 2000 (blank group). Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was used to detect mRNA expression levels of E‑cadherin, N‑cadherin and β‑catenin, and western blot analysis was performed to measure the protein expression levels in the three groups. SRA01/04 cells transfected with E‑cadherin siRNA exhibited a significant decrease in the mRNA expression level of E‑cadherin (P<0.05) and N‑cadherin (P<0.05), whereas no significant changes were identified for β‑catenin expression (P>0.05). Consistent with the results of RT‑qPCR, western blotting demonstrated that the protein expression levels of E‑cadherin and N‑cadherin were notably decreased in E‑cadherin siRNA‑transfected cells, whereas the protein expression level of β‑catenin remained unchanged. Results from the present study indicated that E‑cadherin may be involved in human LEC transdifferentiation by affecting N‑cadherin expression.
Collapse
Affiliation(s)
- Lei Huang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Nangang, Harbin 150001, P.R. China
| | - Jie Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Nangang, Harbin 150001, P.R. China
| | - Qiang Guo
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Nangang, Harbin 150001, P.R. China
| | - Hongbin Yang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Nangang, Harbin 150001, P.R. China
| |
Collapse
|
40
|
Maddala R, Rao PV. Switching of α-Catenin From Epithelial to Neuronal Type During Lens Epithelial Cell Differentiation. Invest Ophthalmol Vis Sci 2017; 58:3445-3455. [PMID: 28692740 PMCID: PMC5505122 DOI: 10.1167/iovs.17-21539] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Ocular lens fiber cell elongation, differentiation, and compaction are associated with extensive reorganization of cell adhesive interactions and cytoskeleton; however, our knowledge of proteins critical to these events is still evolving. This study characterizes the distribution pattern of neuronal-specific α-catenin (αN-catenin) and its interaction with the N-cadherin–associated adherens junctions (AJs) and their stability in the mouse lens fibers. Methods Expression and distribution of αN-catenin in developing mouse and adult human lenses was determined by RT-PCR, immunoblot, and immunofluorescence analyses. Characterization of αN-catenin and N-cadherin interacting proteins and colocalization analyses were performed using immunoprecipitation, mass spectrometry, and confocal imaging. Effects of periaxin deficiency on the stability of lens fiber cell AJs were evaluated using perixin-null mice. Results αN-catenin exhibits discrete distribution to lens fibers in both mouse and human lenses, undergoing a robust up-regulation during fiber cell differentiation and maturation. Epithelial-specific α-catenin (αE-catenin), in contrast, distributes primarily to the lens epithelium. αN-catenin and N-cadherin reciprocally coimmunoprecipitate and colocalize along with β-catenin, actin, spectrin, vinculin, Armadillo repeat protein deleted in velo-cardio-facial syndrome homolog, periaxin, and ankyrin-B in lens fibers. Fiber cells from periaxin-null mouse lenses revealed disrupted N-cadherin/αN-catenin–based AJs. Conclusions These results suggest that the discrete shift in α-catenin expression from αE-catenin to αN-catenin subtype that occurs during lens epithelial cell differentiation may play a key role in fiber cell cytoarchitecture by regulating the assembly and stability of N-cadherin–based AJs. This study also provides evidence for the importance of the fiber cell–specific cytoskeletal interacting periaxin, in the stability of N-cadherin/αN-catenin–based AJs in lens fibers.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States 2Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
41
|
Logan CM, Rajakaruna S, Bowen C, Radice GL, Robinson ML, Menko AS. N-cadherin regulates signaling mechanisms required for lens fiber cell elongation and lens morphogenesis. Dev Biol 2017; 428:118-134. [PMID: 28552735 DOI: 10.1016/j.ydbio.2017.05.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 12/14/2022]
Abstract
Tissue development and regeneration involve high-ordered morphogenetic processes that are governed by elements of the cytoskeleton in conjunction with cell adhesion molecules. Such processes are particularly important in the lens whose structure dictates its function. Studies of our lens-specific N-cadherin conditional knockout mouse (N-cadcKO) revealed an essential role for N-cadherin in the migration of the apical tips of differentiating lens fiber cells along the apical surfaces of the epithelium, a region termed the Epithelial Fiber Interface (EFI), that is necessary for normal fiber cell elongation and the morphogenesis. Studies of the N-cadcKO lens suggest that N-cadherin function in fiber cell morphogenesis is linked to the activation of Rac1 and myosin II, both signaling pathways central to the regulation of cell motility including determining the directionality of cellular movement. The absence of N-cadherin did not disrupt lateral contacts between fiber cells during development, and the maintenance of Aquaporin-0 and increased expression of EphA2 at cell-cell interfaces suggests that these molecules may function in this role. E-cadherin was maintained in newly differentiating fiber cells without interfering with expression of lens-specific differentiation proteins but was not able to replace N-cadherin function in these cells. The dependence of migration of the fiber cell apical domains along the EFI for lens morphogenesis on N-cadherin provides new insight into the process of tissue development.
Collapse
Affiliation(s)
- Caitlin M Logan
- Thomas Jefferson University, Pathology Anatomy and Cell Biology, Philadelphia, PA, United States
| | - Suren Rajakaruna
- Thomas Jefferson University, Pathology Anatomy and Cell Biology, Philadelphia, PA, United States
| | - Caitlin Bowen
- Thomas Jefferson University, Pathology Anatomy and Cell Biology, Philadelphia, PA, United States
| | - Glenn L Radice
- Thomas Jefferson University, Pathology Anatomy and Cell Biology, Philadelphia, PA, United States
| | - Michael L Robinson
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, United States
| | - A Sue Menko
- Thomas Jefferson University, Pathology Anatomy and Cell Biology, Philadelphia, PA, United States.
| |
Collapse
|
42
|
Congenital Cataract in Gpr161vl/vl Mice Is Modified by Proximal Chromosome 15. PLoS One 2017; 12:e0170724. [PMID: 28135291 PMCID: PMC5279759 DOI: 10.1371/journal.pone.0170724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 01/10/2017] [Indexed: 11/24/2022] Open
Abstract
The morphology and severity of human congenital cataract varies even among individuals with the same mutation, suggesting that genetic background modifies phenotypic penetrance. The spontaneous mouse mutant, vacuolated lens (vl), arose on the C3H/HeSnJ background. The mutation disrupts secondary lens fiber development by E16.5, leading to full penetrance of congenital cataract. The vl locus was mapped to a frameshift deletion in the orphan G protein-coupled receptor, Gpr161, which is expressed in differentiating lens fiber cells. When Gpr161vl/vl C3H mice are crossed to MOLF/EiJ mice an unexpected rescue of cataract is observed, suggesting that MOLF modifiers affect cataract penetrance. Subsequent QTL analysis mapped three modifiers (Modvl3-5: Modifier of vl) and in this study we characterized Modvl4 (Chr15; LOD = 4.4). A Modvl4MOLF congenic was generated and is sufficient to rescue congenital cataract and the lens fiber defect at E16.5. Additional phenotypic analysis on three subcongenic lines narrowed down the interval from 55 to 15Mb. In total only 18 protein-coding genes and 2 micro-RNAs are in this region. Fifteen of the 20 genes show detectable expression in the E16.5 eye. Subsequent expression studies in Gpr161vl/vl and subcongenic E16.5 eyes, bioinformatics analysis of C3H/MOLF polymorphisms, and the biological relevancy of the genes in the interval identified three genes (Cdh6, Ank and Trio) that likely contribute to the rescue of the lens phenotype. These studies demonstrate that modification of the Gpr161vl/vl cataract phenotype is likely due to genetic variants in at least one of three closely linked candidate genes on proximal Chr15.
Collapse
|
43
|
Fujimura N. WNT/β-Catenin Signaling in Vertebrate Eye Development. Front Cell Dev Biol 2016; 4:138. [PMID: 27965955 PMCID: PMC5127792 DOI: 10.3389/fcell.2016.00138] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/09/2016] [Indexed: 01/04/2023] Open
Abstract
The vertebrate eye is a highly specialized sensory organ, which is derived from the anterior neural plate, head surface ectoderm, and neural crest-derived mesenchyme. The single central eye field, generated from the anterior neural plate, divides to give rise to the optic vesicle, which evaginates toward the head surface ectoderm. Subsequently, the surface ectoderm, in conjunction with the optic vesicle invaginates to form the lens vesicle and double-layered optic cup, respectively. This complex process is controlled by transcription factors and several intracellular and extracellular signaling pathways including WNT/β-catenin signaling. This signaling pathway plays an essential role in multiple developmental processes and has a profound effect on cell proliferation and cell fate determination. During eye development, the activity of WNT/β-catenin signaling is tightly controlled. Faulty regulation of WNT/β-catenin signaling results in multiple ocular malformations due to defects in the process of cell fate determination and differentiation. This mini-review summarizes recent findings on the role of WNT/β-catenin signaling in eye development. Whilst this mini-review focuses on loss-of-function and gain-of-function mutants of WNT/β-catenin signaling components, it also highlights some important aspects of β-catenin-independent WNT signaling in the eye development at later stages.
Collapse
Affiliation(s)
- Naoko Fujimura
- Laboratory of Eye Biology, BIOCEV Division, Institute of Molecular Genetics Prague, Czechia
| |
Collapse
|
44
|
Nguyen T, Mège RM. N-Cadherin and Fibroblast Growth Factor Receptors crosstalk in the control of developmental and cancer cell migrations. Eur J Cell Biol 2016; 95:415-426. [PMID: 27320194 DOI: 10.1016/j.ejcb.2016.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/13/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022] Open
Abstract
Cell migrations are diverse. They constitutemajor morphogenetic driving forces during embryogenesis, but they contribute also to the loss of tissue homeostasis and cancer growth. Capabilities of cells to migrate as single cells or as collectives are controlled by internal and external signalling, leading to the reorganisation of their cytoskeleton as well as by the rebalancing of cell-matrix and cell-cell adhesions. Among the genes altered in numerous cancers, cadherins and growth factor receptors are of particular interest for cell migration regulation. In particular, cadherins such as N-cadherin and a class of growth factor receptors, namely FGFRs cooperate to regulate embryonic and cancer cell behaviours. In this review, we discuss on reciprocal crosstalk between N-cadherin and FGFRs during cell migration. Finally, we aim at clarifying the synergy between N-cadherin and FGFR signalling that ensure cellular reorganization during cell movements, mainly during cancer cell migration and metastasis but also during developmental processes.
Collapse
Affiliation(s)
- Thao Nguyen
- Institut Jacques Monod, CNRS, Université Paris Diderot, Paris, France
| | - René Marc Mège
- Institut Jacques Monod, CNRS, Université Paris Diderot, Paris, France.
| |
Collapse
|
45
|
He S, Limi S, McGreal RS, Xie Q, Brennan LA, Kantorow WL, Kokavec J, Majumdar R, Hou H, Edelmann W, Liu W, Ashery-Padan R, Zavadil J, Kantorow M, Skoultchi AI, Stopka T, Cvekl A. Chromatin remodeling enzyme Snf2h regulates embryonic lens differentiation and denucleation. Development 2016; 143:1937-47. [PMID: 27246713 PMCID: PMC4920164 DOI: 10.1242/dev.135285] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/21/2016] [Indexed: 12/30/2022]
Abstract
Ocular lens morphogenesis is a model for investigating mechanisms of cellular differentiation, spatial and temporal gene expression control, and chromatin regulation. Brg1 (Smarca4) and Snf2h (Smarca5) are catalytic subunits of distinct ATP-dependent chromatin remodeling complexes implicated in transcriptional regulation. Previous studies have shown that Brg1 regulates both lens fiber cell differentiation and organized degradation of their nuclei (denucleation). Here, we employed a conditional Snf2h(flox) mouse model to probe the cellular and molecular mechanisms of lens formation. Depletion of Snf2h induces premature and expanded differentiation of lens precursor cells forming the lens vesicle, implicating Snf2h as a key regulator of lens vesicle polarity through spatial control of Prox1, Jag1, p27(Kip1) (Cdkn1b) and p57(Kip2) (Cdkn1c) gene expression. The abnormal Snf2h(-/-) fiber cells also retain their nuclei. RNA profiling of Snf2h(-/) (-) and Brg1(-/-) eyes revealed differences in multiple transcripts, including prominent downregulation of those encoding Hsf4 and DNase IIβ, which are implicated in the denucleation process. In summary, our data suggest that Snf2h is essential for the establishment of lens vesicle polarity, partitioning of prospective lens epithelial and fiber cell compartments, lens fiber cell differentiation, and lens fiber cell nuclear degradation.
Collapse
Grants
- R01 EY012200 NEI NIH HHS
- R01 CA079057 NCI NIH HHS
- R01 DK096266 NIDDK NIH HHS
- R01 GM116143 NIGMS NIH HHS
- R01 EY013022 NEI NIH HHS
- R01 CA076329 NCI NIH HHS
- T32 GM007491 NIGMS NIH HHS
- R56 CA079057 NCI NIH HHS
- R01 EY014237 NEI NIH HHS
- 001 World Health Organization
- R01 EY022645 NEI NIH HHS
- Grant support: R01 EY012200 (AC), EY014237 (AC), EY014237-7S1 (AC), EY013022 (MK), CA079057 (AIS), EY022645 (WL), T32 GM007491 (SL), GACR: P305/12/1033 (TS, JK), UNCE: 204021 (TS, JK), and an unrestricted grant from Research to Prevent Blindness to the Department of Ophthalmology and Visual Sciences. TS is member of the BIOCEV ? Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (CZ.1.05/1.1.00/02.0109) supported by the European Regional Development Fund. The Israel Science Foundation 610/10, the Israel Ministry of Science 36494, the Ziegler Foundation and the Binational Science Foundation (2013016) to RAP.
Collapse
Affiliation(s)
- Shuying He
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Saima Limi
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rebecca S McGreal
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qing Xie
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lisa A Brennan
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Wanda Lee Kantorow
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Juraj Kokavec
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Romit Majumdar
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Harry Hou
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wei Liu
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Jiri Zavadil
- Department of Pathology and NYU Center for Health Informatics and Bioinformatics, New York University Langone Medical Center, New York, NY 10016, USA Mechanisms of Carcinogenesis Section, International Agency for Research on Cancer, Lyon Cedex 08 69372, France
| | - Marc Kantorow
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Arthur I Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tomas Stopka
- First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Ales Cvekl
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
46
|
Audette DS, Scheiblin DA, Duncan MK. The molecular mechanisms underlying lens fiber elongation. Exp Eye Res 2016; 156:41-49. [PMID: 27015931 DOI: 10.1016/j.exer.2016.03.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 12/28/2022]
Abstract
Lens fiber cells are highly elongated cells with complex membrane morphologies that are critical for the transparency of the ocular lens. Investigations into the molecular mechanisms underlying lens fiber cell elongation were first reported in the 1960s, however, our understanding of the process is still poor nearly 50 years later. This review summarizes what is currently hypothesized about the regulation of lens fiber cell elongation along with the available experimental evidence, and how this information relates to what is known about the regulation of cell shape/elongation in other cell types, particularly neurons.
Collapse
Affiliation(s)
- Dylan S Audette
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - David A Scheiblin
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
47
|
Wu JJ, Wu W, Tholozan FM, Saunter CD, Girkin JM, Quinlan RA. A dimensionless ordered pull-through model of the mammalian lens epithelium evidences scaling across species and explains the age-dependent changes in cell density in the human lens. J R Soc Interface 2016; 12:20150391. [PMID: 26236824 PMCID: PMC4528606 DOI: 10.1098/rsif.2015.0391] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We present a mathematical (ordered pull-through; OPT) model of the cell-density profile for the mammalian lens epithelium together with new experimental data. The model is based upon dimensionless parameters, an important criterion for inter-species comparisons where lens sizes can vary greatly (e.g. bovine (approx. 18 mm); mouse (approx. 2 mm)) and confirms that mammalian lenses scale with size. The validated model includes two parameters: β/α, which is the ratio of the proliferation rate in the peripheral and in the central region of the lens; and γGZ, a dimensionless pull-through parameter that accounts for the cell transition and exit from the epithelium into the lens body. Best-fit values were determined for mouse, rat, rabbit, bovine and human lens epithelia. The OPT model accounts for the peak in cell density at the periphery of the lens epithelium, a region where cell proliferation is concentrated and reaches a maximum coincident with the germinative zone. The β/α ratio correlates with the measured FGF-2 gradient, a morphogen critical to lens cell survival, proliferation and differentiation. As proliferation declines with age, the OPT model predicted age-dependent changes in cell-density profiles, which we observed in mouse and human lenses.
Collapse
Affiliation(s)
- Jun Jie Wu
- Biophysical Sciences Institute and School of Engineering and Computing Sciences, Durham University, Durham DH1 3LE, UK
- e-mail:
| | - Weiju Wu
- Biophysical Sciences Institute and School of Biological and Biomedical Sciences, Durham University, Durham DH1 3LE, UK
| | - Frederique M. Tholozan
- Biophysical Sciences Institute and School of Biological and Biomedical Sciences, Durham University, Durham DH1 3LE, UK
| | - Christopher D. Saunter
- Biophysical Sciences Institute and Department of Physics, Durham University, Durham DH1 3LE, UK
| | - John M. Girkin
- Biophysical Sciences Institute and Department of Physics, Durham University, Durham DH1 3LE, UK
| | - Roy A. Quinlan
- Biophysical Sciences Institute and School of Biological and Biomedical Sciences, Durham University, Durham DH1 3LE, UK
- e-mail:
| |
Collapse
|
48
|
Cheng C, Nowak RB, Fowler VM. The lens actin filament cytoskeleton: Diverse structures for complex functions. Exp Eye Res 2016; 156:58-71. [PMID: 26971460 DOI: 10.1016/j.exer.2016.03.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 03/01/2016] [Accepted: 03/07/2016] [Indexed: 01/05/2023]
Abstract
The eye lens is a transparent and avascular organ in the front of the eye that is responsible for focusing light onto the retina in order to transmit a clear image. A monolayer of epithelial cells covers the anterior hemisphere of the lens, and the bulk of the lens is made up of elongated and differentiated fiber cells. Lens fiber cells are very long and thin cells that are supported by sophisticated cytoskeletal networks, including actin filaments at cell junctions and the spectrin-actin network of the membrane skeleton. In this review, we highlight the proteins that regulate diverse actin filament networks in the lens and discuss how these actin cytoskeletal structures assemble and function in epithelial and fiber cells. We then discuss methods that have been used to study actin in the lens and unanswered questions that can be addressed with novel techniques.
Collapse
Affiliation(s)
- Catherine Cheng
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Roberta B Nowak
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Velia M Fowler
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
49
|
Bykhovskaya Y, Gromova A, Makarenkova HP, Rabinowitz YS. Abnormal regulation of extracellular matrix and adhesion molecules in corneas of patients with keratoconus. ACTA ACUST UNITED AC 2016; 5:63-70. [PMID: 28989906 DOI: 10.5005/jp-journals-10025-1123] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AIM To identify changes in the expression of genes coding for extracellular matrix (ECM) proteins in patients with non-inflammatory corneal disorder keratoconus (KC), patients with corneal scarring, and normal controls. MATERIALS AND METHODS Total RNA extracted from corneal tissue of 13 KC patients, 2 patients with corneal scaring and 4 normal controls was analyzed using Human Extracellular Matrix & Adhesion Molecules Profiler PCR Array. Statistically significant changes in gene expression were identified using the Data Analysis software. RESULTS Comparison of KC and control corneas with thresholds of 1.5 or greater fold change and a p-value of 0.05 or lower, revealed 21 differentially expressed genes, 16 genes were downregulated and 5 were upregulated. Among transcripts downregulated in KC patients we identified THBS1, ADAMTS1, SPP1, several collagens and integrins. We found TGFBI (BIGH3) gene was the most significantly upregulated transcript. CONCLUSION Development of keratoconus results in deregulation of gene expression of extracellular matrix and adhesion molecules. CLINICAL SIGNIFICANCE Downregulation of collagens and upregulation of TGFBI repeatedly identified in KC patients may be used as clinical markers of the disease.
Collapse
Affiliation(s)
- Yelena Bykhovskaya
- Regenerative Medicine Institute and Department of Surgery, Cedars-Sinai, USA.,Cornea Genetic Eye Institute, USA
| | - Anastasia Gromova
- Department of Cell and Molecular Biology, The Scripps Research Institute, USA
| | - Helen P Makarenkova
- Department of Cell and Molecular Biology, The Scripps Research Institute, USA
| | - Yaron S Rabinowitz
- Regenerative Medicine Institute and Department of Surgery, Cedars-Sinai, USA.,Cornea Genetic Eye Institute, USA.,The Jules Stein Eye Institute, University of California Los Angeles, USA
| |
Collapse
|
50
|
Zhang Y, Fan J, Ho JWK, Hu T, Kneeland SC, Fan X, Xi Q, Sellarole MA, de Vries WN, Lu W, Lachke SA, Lang RA, John SWM, Maas RL. Crim1 regulates integrin signaling in murine lens development. Development 2015; 143:356-66. [PMID: 26681494 PMCID: PMC4725338 DOI: 10.1242/dev.125591] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 12/07/2015] [Indexed: 12/19/2022]
Abstract
The developing lens is a powerful system for investigating the molecular basis of inductive tissue interactions and for studying cataract, the leading cause of blindness. The formation of tightly controlled cell-cell adhesions and cell-matrix junctions between lens epithelial (LE) cells, between lens fiber (LF) cells, and between these two cell populations enables the vertebrate lens to adopt a highly ordered structure and acquire optical transparency. Adhesion molecules are thought to maintain this ordered structure, but little is known about their identity or interactions. Cysteine-rich motor neuron 1 (Crim1), a type I transmembrane protein, is strongly expressed in the developing lens and its mutation causes ocular disease in both mice and humans. How Crim1 regulates lens morphogenesis is not understood. We identified a novel ENU-induced hypomorphic allele of Crim1, Crim1glcr11, which in the homozygous state causes cataract and microphthalmia. Using this and two other mutant alleles, Crim1null and Crim1cko, we show that the lens defects in Crim1 mouse mutants originate from defective LE cell polarity, proliferation and cell adhesion. Crim1 adhesive function is likely to be required for interactions both between LE cells and between LE and LF cells. We show that Crim1 acts in LE cells, where it colocalizes with and regulates the levels of active β1 integrin and of phosphorylated FAK and ERK. The RGD and transmembrane motifs of Crim1 are required for regulating FAK phosphorylation. These results identify an important function for Crim1 in the regulation of integrin- and FAK-mediated LE cell adhesion during lens development. Summary: Crim1, a type I transmembrane protein, acts in lens epithelial cells where it colocalizes with and regulates the levels of active β1 integrin to control cell adhesion during mouse lens morphogenesis.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jieqing Fan
- Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joshua W K Ho
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA Victor Chang Cardiac Research Institute, and The University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Tommy Hu
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephen C Kneeland
- Howard Hughes Medical Institute and The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Xueping Fan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | - Qiongchao Xi
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Sellarole
- Victor Chang Cardiac Research Institute, and The University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Wilhelmine N de Vries
- Victor Chang Cardiac Research Institute, and The University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | - Salil A Lachke
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Richard A Lang
- Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Simon W M John
- Victor Chang Cardiac Research Institute, and The University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Richard L Maas
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|