1
|
Shrestha SK, Lachke SA. Lens Regeneration: The Application of iSyTE and In Silico Approaches to Evaluate Gene Expression in Lens Organoids. Methods Mol Biol 2025; 2848:37-58. [PMID: 39240515 DOI: 10.1007/978-1-0716-4087-6_3] [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
Several protocols have been established for the generation of lens organoids from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and other cells with regenerative potential in humans or various animal models. It is important to examine how well the regenerated lens organoids reflect lens biology, in terms of its development, homeostasis, and aging. Toward this goal, the iSyTE database (integrated Systems Tool for Eye gene discovery; https://research.bioinformatics.udel.edu/iSyTE/ ), a bioinformatics resource tool that contains meta-analyzed gene expression data in wild-type lens across different embryonic, postnatal, and adult stages, can serve as a resource for comparative analysis. This article outlines the approaches toward effective use of iSyTE to gain insights into normal gene expression in the mouse lens, enriched expression in the lens, and differential gene expression in select mouse gene-perturbation cataract/lens defects models, which in turn can be used to evaluate expression of key lens-relevant genes in lens organoids by transcriptomics (e.g., RNA-sequencing (RNA-seq), microarrays, etc.) or other downstream methods (e.g., RT-qPCR, etc.).
Collapse
Affiliation(s)
- Sanjaya K Shrestha
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Salil A Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.
- Center for Bioinformatics & Computational Biology, University of Delaware, Newark, DE, USA.
| |
Collapse
|
2
|
Subramanian V, Juhr D, Johnson LS, Yem JB, Giansanti P, Grumbach IM. Changes in the Proteome of the Circle of Willis during Aging Reveal Signatures of Vascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:4887877. [PMID: 38962180 PMCID: PMC11221951 DOI: 10.1155/2024/4887877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/22/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
Approximately 70% of all strokes occur in patients over 65 years old, and stroke increases the risk of developing dementia. The circle of Willis (CoW), the ring of arteries at the base of the brain, links the intracerebral arteries to one another to maintain adequate cerebral perfusion. The CoW proteome is affected in cerebrovascular and neurodegenerative diseases, but changes related to aging have not been described. Here, we report on a quantitative proteomics analysis comparing the CoW from five young (2-3-month-old) and five aged male (18-20-month-old) mice using gene ontology (GO) enrichment, ingenuity pathway analysis (IPA), and iPathwayGuide tools. This revealed 242 proteins that were significantly dysregulated with aging, among which 189 were upregulated and 53 downregulated. GO enrichment-based analysis identified blood coagulation as the top biological function that changed with age and integrin binding and extracellular matrix constituents as the top molecular functions. Consistent with these findings, iPathwayGuide-based impact analysis revealed associations between aging and the complement and coagulation, platelet activation, ECM-receptor interaction, and metabolic process pathways. Furthermore, IPA analysis revealed the enrichment of 97 canonical pathways that contribute to inflammatory responses, as well as 59 inflammation-associated upstream regulators including 39 transcription factors and 20 cytokines. Thus, aging-associated changes in the CoW proteome in male mice demonstrate increases in metabolic, thrombotic, and inflammatory processes.
Collapse
Affiliation(s)
- Vikram Subramanian
- Abboud Cardiovascular Research CenterDepartment of Internal MedicineCarver College of MedicineUniversity of Iowa, Iowa City, USA
| | - Denise Juhr
- Abboud Cardiovascular Research CenterDepartment of Internal MedicineCarver College of MedicineUniversity of Iowa, Iowa City, USA
| | - Lydia S. Johnson
- Abboud Cardiovascular Research CenterDepartment of Internal MedicineCarver College of MedicineUniversity of Iowa, Iowa City, USA
| | - Justin B. Yem
- Abboud Cardiovascular Research CenterDepartment of Internal MedicineCarver College of MedicineUniversity of Iowa, Iowa City, USA
| | - Piero Giansanti
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS@MRI)Technical University of Munich, Munich, Germany
| | - Isabella M. Grumbach
- Abboud Cardiovascular Research CenterDepartment of Internal MedicineCarver College of MedicineUniversity of Iowa, Iowa City, USA
- Free Radical and Radiation Biology ProgramDepartment of Radiation OncologyCarver College of MedicineUniversity of Iowa, Iowa City, USA
- Iowa City VA Healthcare System, Iowa City, IA, USA
| |
Collapse
|
3
|
Choquet H, Duot M, Herrera VA, Shrestha SK, Meyers TJ, Hoffmann TJ, Sangani PK, Lachke SA. Multi-tissue transcriptome-wide association study identifies novel candidate susceptibility genes for cataract. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1362350. [PMID: 38984127 PMCID: PMC11182099 DOI: 10.3389/fopht.2024.1362350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/01/2024] [Indexed: 07/11/2024]
Abstract
Introduction Cataract is the leading cause of blindness among the elderly worldwide. Twin and family studies support an important role for genetic factors in cataract susceptibility with heritability estimates up to 58%. To date, 55 loci for cataract have been identified by genome-wide association studies (GWAS), however, much work remains to identify the causal genes. Here, we conducted a transcriptome-wide association study (TWAS) of cataract to prioritize causal genes and identify novel ones, and examine the impact of their expression. Methods We performed tissue-specific and multi-tissue TWAS analyses to assess associations between imputed gene expression from 54 tissues (including 49 from the Genotype Tissue Expression (GTEx) Project v8) with cataract using FUSION software. Meta-analyzed GWAS summary statistics from 59,944 cataract cases and 478,571 controls, all of European ancestry and from two cohorts (GERA and UK Biobank) were used. We then examined the expression of the novel genes in the lens tissue using the iSyTE database. Results Across tissue-specific and multi-tissue analyses, we identified 99 genes for which genetically predicted gene expression was associated with cataract after correcting for multiple testing. Of these 99 genes, 20 (AC007773.1, ANKH, ASIP, ATP13A2, CAPZB, CEP95, COQ6, CREB1, CROCC, DDX5, EFEMP1, EIF2S2, ESRRB, GOSR2, HERC4, INSRR, NIPSNAP2, PICALM, SENP3, and SH3YL1) did not overlap with previously reported cataract-associated loci. Tissue-specific analysis identified 202 significant gene-tissue associations for cataract, of which 166 (82.2%), representing 9 unique genes, were attributed to the previously reported 11q13.3 locus. Tissue-enrichment analysis revealed that gastrointestinal tissues represented one of the highest proportions of the Bonferroni-significant gene-tissue associations (21.3%). Moreover, this gastrointestinal tissue type was the only anatomical category significantly enriched in our results, after correcting for the number of tissue donors and imputable genes for each reference panel. Finally, most of the novel cataract genes (e.g., Capzb) were robustly expressed in iSyTE lens data. Discussion Our results provide evidence of the utility of imputation-based TWAS approaches to characterize known GWAS risk loci and identify novel candidate genes that may increase our understanding of cataract etiology. Our findings also highlight the fact that expression of genes associated with cataract susceptibility is not necessarily restricted to lens tissue.
Collapse
Affiliation(s)
- Hélène Choquet
- Kaiser Permanente Northern California (KPNC), Division of Research, Oakland, CA, United States
| | - Matthieu Duot
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
- The National Centre for Scientific Research (CNRS), IGDR (Institut de Génétique et Développement de Rennes) - Joint Research Units (UMR), Univ Rennes, Rennes, France
| | - Victor A Herrera
- Kaiser Permanente Northern California (KPNC), Division of Research, Oakland, CA, United States
| | - Sanjaya K Shrestha
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Travis J Meyers
- Kaiser Permanente Northern California (KPNC), Division of Research, Oakland, CA, United States
| | - Thomas J Hoffmann
- Institute for Human Genetics, University of California San Francisco (UCSF), San Francisco, CA, United States
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, United States
| | - Poorab K Sangani
- Department of Ophthalmology, KPNC, South San Francisco, CA, United States
| | - Salil A Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, United States
| |
Collapse
|
4
|
Ye L, Yuan J, Zhu S, Ji S, Dai J. Swimming exercise reverses transcriptomic changes in aging mouse lens. BMC Med Genomics 2024; 17:67. [PMID: 38439070 PMCID: PMC10913554 DOI: 10.1186/s12920-024-01839-1] [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: 11/09/2023] [Accepted: 02/28/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND The benefits of physical activity for the overall well-being of elderly individuals are well-established, the precise mechanisms through which exercise improves pathological changes in the aging lens have yet to be fully understood. METHODS 3-month-old C57BL/6J mice comprised young sedentary (YS) group, while aging mice (18-month-old) were divided into aging sedentary (AS) group and aging exercising (AE) group. Mice in AE groups underwent sequential stages of swimming exercise. H&E staining was employed to observe alterations in lens morphology. RNA-seq analysis was utilized to examine transcriptomic changes. Furthermore, qPCR and immunohistochemistry were employed for validation of the results. RESULTS AE group showed alleviation of histopathological aging changes in AS group. By GSEA analysis of the transcriptomic changes, swimming exercise significantly downregulated approximately half of the pathways that underwent alterations upon aging, where notable improvements were 'calcium signaling pathway', 'neuroactive ligand receptor interaction' and 'cell adhesion molecules'. Furthermore, we revealed a total of 92 differentially expressed genes between the YS and AS groups, of which 10 genes were observed to be mitigated by swimming exercise. The result of qPCR was in consistent with the transcriptome data. We conducted immunohistochemical analysis on Ciart, which was of particular interest due to its dual association as a common aging gene and its significant responsiveness to exercise. The Protein-protein Interaction network of Ciart showed the involvement of the regulation of Rorb and Sptbn5 during the process. CONCLUSION The known benefits of exercise could extend to the aging lens and support further investigation into the specific roles of Ciart-related pathways in aging lens.
Collapse
Affiliation(s)
- Lin Ye
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayue Yuan
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijie Zhu
- School of Medicine, Tongji University, Shanghai, China
| | - Shunmei Ji
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinhui Dai
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
O'Neill LM, Wang Y, Duncan MK. Modeling Cataract Surgery in Mice. J Vis Exp 2023:10.3791/66050. [PMID: 38108456 PMCID: PMC10981495 DOI: 10.3791/66050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Cataract surgery (CS) is an effective treatment for cataracts, a major cause of visual disability worldwide. However, CS leads to ocular inflammation, and in the long term, it can result in posterior capsular opacification (PCO) and/or lens dislocation driven by the post-surgical overgrowth of lens epithelial cells (LECs) and their conversion to myofibroblasts and/or aberrant fiber cells. However, the molecular mechanisms by which CS results in inflammation and PCO are still obscure because most in vitro models do not recapitulate the wound healing response of LECs seen in vivo, while traditional animal models of cataract surgery, such as rabbits, do not allow the genetic manipulation of gene expression to test mechanisms. Recently, our laboratory and others have successfully used genetically modified mice to study the molecular mechanisms that drive the induction of proinflammatory signaling and LEC epithelial to mesenchymal transition, leading to new insight into PCO pathogenesis. Here, we report the established protocol for modeling cataract surgery in mice, which allows for robust transcriptional profiling of the response of LECs to lens fiber cell removal via RNAseq, the evaluation of protein expression by semi-quantitative immunofluorescence, and the use of modern mouse genetics tools to test the function of genes that are hypothesized to participate in the pathogenesis of acute sequelae like inflammation as well as the later conversion of LECs to myofibroblasts and/or aberrant lens fiber cells.
Collapse
Affiliation(s)
- Leah M O'Neill
- Department of Biological Sciences, University of Delaware
| | - Yan Wang
- Department of Biological Sciences, University of Delaware
| | | |
Collapse
|
6
|
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
|
7
|
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: 1.0] [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
|
8
|
Rodriguez J, Tan Q, Šikić H, Taber LA, Bassnett S. The effect of fibre cell remodelling on the power and optical quality of the lens. J R Soc Interface 2023; 20:20230316. [PMID: 37727073 PMCID: PMC10509584 DOI: 10.1098/rsif.2023.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023] Open
Abstract
Vertebrate eye lenses are uniquely adapted to form a refractive index gradient (GRIN) for improved acuity, and to grow slowly in size despite constant cell proliferation. The mechanisms behind these adaptations remain poorly understood. We hypothesize that cell compaction contributes to both. To test this notion, we examined the relationship between lens size and shape, refractive characteristics and the cross-sectional areas of constituent fibre cells in mice of different ages. We developed a block-face imaging method to visualize cellular cross sections and found that the cross-sectional areas of fibre cells rose and then decreased over time, with the most significant reduction occurring in denucleating cells in the adult lens cortex, followed by cells in the embryonic nucleus. These findings help reconcile differences between the predictions of lens growth models and empirical data. Biomechanical simulations suggested that compressive forces generated from continuous deposition of fibre cells could contribute to cellular compaction. However, optical measurements revealed that the GRIN did not mirror the pattern of cellular compaction, implying that compaction alone cannot account for GRIN formation and that additional mechanisms are likely to be involved.
Collapse
Affiliation(s)
- J. Rodriguez
- Department of Basic Sciences, University of Health Sciences and Pharmacy in St. Louis, 1 Pharmacy Place, St. Louis, MO 63110, USA
| | - Q. Tan
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8096, St. Louis, MO 63110, USA
| | - H. Šikić
- Department of Mathematics, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - L. A. Taber
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - S. Bassnett
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8096, St. Louis, MO 63110, USA
| |
Collapse
|
9
|
The Immediate Early Response of Lens Epithelial Cells to Lens Injury. Cells 2022; 11:cells11213456. [PMID: 36359852 PMCID: PMC9654717 DOI: 10.3390/cells11213456] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Cataracts are treated by lens fiber cell removal followed by intraocular lens (IOL) implantation into the lens capsule. While effective, this procedure leaves behind numerous lens epithelial cells (LECs) which undergo a wound healing response that frequently leads to posterior capsular opacification (PCO). In order to elucidate the acute response of LECs to lens fiber cell removal which models cataract surgery (post cataract surgery, PCS), RNA-seq was conducted on LECs derived from wild type mice at 0 and 6 h PCS. This analysis found that LECs upregulate the expression of numerous proinflammatory cytokines and profibrotic regulators by 6 h PCS suggesting rapid priming of pathways leading to inflammation and fibrosis PCS. LECs also highly upregulate the expression of numerous immediate early transcription factors (IETFs) by 6 h PCS and immunolocalization found elevated levels of these proteins by 3 h PCS, and this was preceded by the phosphorylation of ERK1/2 in injured LECs. Egr1 and FosB were among the highest expressed of these factors and qRT-PCR revealed that they also upregulate in explanted mouse lens epithelia suggesting potential roles in the LEC injury response. Analysis of lenses lacking either Egr1 or FosB revealed that both genes may regulate a portion of the acute LEC injury response, although neither gene was essential for expression of either proinflammatory or fibrotic markers at later times PCS suggesting that IETFs may work in concert to mediate the LEC injury response following cataract surgery.
Collapse
|
10
|
Biliverdin Reductase A Protects Lens Epithelial Cells against Oxidative Damage and Cellular Senescence in Age-Related Cataract. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5628946. [PMID: 35910837 PMCID: PMC9325611 DOI: 10.1155/2022/5628946] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023]
Abstract
Age-related cataract (ARC) is the common cause of blindness globally. Reactive oxygen species (ROS), one of the greatest contributors to aging process, leads to oxidative damage and senescence of lens epithelial cells (LECs), which are involved in the pathogenesis of ARC. Biliverdin reductase A (BVRA) has ROS-scavenging ability by converting biliverdin (BV) into bilirubin (BR). However, little is known about the protective effect of BVRA against ARC. In the present study, we measured the expression level of BVRA and BR generation in human samples. Then, the antioxidative property of BVRA was compared between the young and senescent LECs upon stress condition. In addition, we evaluated the effect of BVRA on attenuating H2O2-induced premature senescence in LECs. The results showed that the mRNA expression level of BVRA and BR concentration were decreased in both LECs and lens cortex of age-related nuclear cataract. Using the RNA interference technique, we found that BVRA defends LECs against oxidative stress via (i) restoring mitochondrial dysfunction in a BR-dependent manner, (ii) inducing heme oxygenase-1 (HO-1) expression directly, and (iii) promoting phosphorylation of ERK1/2 and nuclear delivery of nuclear factor erythroid 2-related factor 2 (Nrf2). Intriguingly, the antioxidative effect of BVRA was diminished along with the reduced BR concentration and repressed nuclear translocation of BVRA and Nrf2 in senescent LECs, which would be resulted from the decreased BVRA activity and impaired nucleocytoplasmic trafficking. Eventually, we confirmed that BVRA accelerates the G1 phase transition and prevents against H2O2-induced premature senescence in LECs. In summary, BVRA protects LECs against oxidative stress and cellular senescence in ARC by converting BV into BR, inducing HO-1 expression, and activating the ERK/Nrf2 pathway. This trial is registered with ChiCTR2000036059.
Collapse
|
11
|
Wei Z, Gordon P, Hao C, Huangfu J, Fan E, Zhang X, Yan H, Fan X. Aged Lens Epithelial Cells Suppress Proliferation and Epithelial–Mesenchymal Transition-Relevance for Posterior Capsule Opacification. Cells 2022; 11:cells11132001. [PMID: 35805085 PMCID: PMC9265589 DOI: 10.3390/cells11132001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 02/01/2023] Open
Abstract
Posterior capsule opacification (PCO) is a frequent complication after cataract surgery, and advanced PCO requires YAG laser (Nd: YAG) capsulotomy, which often gives rise to more complications. Lens epithelial cell (LEC) proliferation and transformation (i.e., epithelial–mesenchymal transition (EMT)) are two critical elements in PCO initiation and progression pathogenesis. While PCO marginally impacts aged cataract surgery patients, PCO incidences are exceptionally high in infants and children undergoing cataract surgery. The gene expression of lens epithelial cell aging and its role in the discrepancy of PCO prevalence between young and older people have not been fully studied. Here, we conducted a comprehensive differentially expressed gene (DEG) analysis of a cell aging model by comparing the early and late passage FHL124 lens epithelial cells (LECs). In vitro, TGFβ2, cell treatment, and in vivo mouse cataract surgical models were used to validate our findings. We found that aged LECs decelerated rates of cell proliferation accompanied by dysregulation of cellular immune response and cell stress response. Surprisingly, we found that LECs systematically downregulated epithelial–mesenchymal transition (EMT)-promoting genes. The protein expression of several EMT hallmark genes, e.g., fibronectin, αSMA, and cadherin 11, were gradually decreased during LECs aging. We then confirmed these findings in vitro and found that aged LECs markedly alleviated TGFβ2-mediated EMT. Importantly, we explicitly confirmed the in vitro findings from the in vivo mouse cataract surgery studies. We propose that both the high proliferation rate and EMT-enriched young LECs phenotypic characteristics contribute to unusually high PCO incidence in infants and children.
Collapse
Affiliation(s)
- Zongbo Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
| | - Pasley Gordon
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Caili Hao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
| | - Jingru Huangfu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
| | - Emily Fan
- Lakeside High School at Columbia County, Evans, GA 30809, USA;
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45221, USA;
| | - Hong Yan
- Xi’an Fourth Hospital, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Xingjun Fan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., CB Building, Room CB1119, Augusta, GA 30912, USA; (Z.W.); (C.H.); (J.H.)
- Correspondence:
| |
Collapse
|
12
|
Lachke SA. RNA-binding proteins and post-transcriptional regulation in lens biology and cataract: Mediating spatiotemporal expression of key factors that control the cell cycle, transcription, cytoskeleton and transparency. Exp Eye Res 2022; 214:108889. [PMID: 34906599 PMCID: PMC8792301 DOI: 10.1016/j.exer.2021.108889] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/29/2021] [Accepted: 12/05/2021] [Indexed: 01/03/2023]
Abstract
Development of the ocular lens - a transparent tissue capable of sustaining frequent shape changes for optimal focusing power - pushes the boundaries of what cells can achieve using the molecular toolkit encoded by their genomes. The mammalian lens contains broadly two types of cells, the anteriorly located monolayer of epithelial cells which, at the equatorial region of the lens, initiate differentiation into fiber cells that contribute to the bulk of the tissue. This differentiation program involves massive upregulation of select fiber cell-expressed RNAs and their subsequent translation into high amounts of proteins, such as crystallins. But intriguingly, fiber cells achieve this while also simultaneously undergoing significant morphological changes such as elongation - involving about 1000-fold length-wise increase - and migration, which requires modulation of cytoskeletal and cell adhesion factors. Adding further to the challenges, these molecular and cellular events have to be coordinated as fiber cells progress toward loss of their nuclei and organelles, which irreversibly compromises their potential for harnessing genetically hardwired information. A long-standing question is how processes downstream of signaling and transcription, which may also participate in feedback regulation, contribute toward orchestrating these cellular differentiation events in the lens. It is now becoming clear from findings over the past decade that post-transcriptional gene expression regulatory mechanisms are critical in controlling cellular proteomes and coordinating key processes in lens development and fiber cell differentiation. Indeed, RNA-binding proteins (RBPs) such as Caprin2, Celf1, Rbm24 and Tdrd7 have now been described in mediating post-transcriptional control over key factors (e.g. Actn2, Cdkn1a (p21Cip1), Cdkn1b (p27Kip1), various crystallins, Dnase2b, Hspb1, Pax6, Prox1, Sox2) that are variously involved in cell cycle, transcription, cytoskeleton maintenance and differentiation in the lens. Furthermore, deficiencies of these RBPs have been shown to result in various eye and lens defects and/or cataract. Because fiber cell differentiation in the lens occurs throughout life, the underlying regulatory mechanisms operational in development are expected to also be recruited for the maintenance of transparency in aged lenses. Indeed, in support of this, TDRD7 and CAPRIN2 loci have been linked to age-related cataract in humans. Here, I will review the role of key RBPs in the lens and their importance in understanding the pathology of lens defects. I will discuss advances in RBP-based gene expression control, in general, and the important challenges that need to be addressed in the lens to define the mechanisms that determine the epithelial and fiber cell proteome. Finally, I will also discuss in detail several key future directions including the application of bioinformatics approaches such as iSyTE to study RBP-based post-transcriptional gene expression control in the aging lens and in the context of age-related cataract.
Collapse
Affiliation(s)
- Salil A Lachke
- Department of Biological Sciences, University of Delaware, 105 The Green, Delaware Avenue, 236 Wolf Hall, Newark, DE, USA; Center for Bioinformatics & Computational Biology, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
13
|
Rowan S, Jiang S, Francisco SG, Pomatto LCD, Ma Z, Jiao X, Campos MM, Aryal S, Patel SD, Mahaling B, Riazuddin SA, Duh EJ, Lachke SA, Hejtmancik JF, de Cabo R, FitzGerald PG, Taylor A. Aged Nrf2-Null Mice Develop All Major Types of Age-Related Cataracts. Invest Ophthalmol Vis Sci 2021; 62:10. [PMID: 34882206 PMCID: PMC8665303 DOI: 10.1167/iovs.62.15.10] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Age-related cataracts affect the majority of older adults and are a leading cause of blindness worldwide. Treatments that delay cataract onset or severity have the potential to delay cataract surgery, but require relevant animal models that recapitulate the major types of cataracts for their development. Unfortunately, few such models are available. Here, we report the lens phenotypes of aged mice lacking the critical antioxidant transcription factor Nfe2l2 (designated as Nrf2 −/−). Methods Three independent cohorts of Nrf2 −/− and wild-type C57BL/6J mice were evaluated for cataracts using combinations of slit lamp imaging, photography of freshly dissected lenses, and histology. Mice were fed high glycemic diets, low glycemic diets, regular chow ad libitum, or regular chow with 30% caloric restriction. Results Nrf2 −/− mice developed significant opacities between 11 and 15 months and developed advanced cortical, posterior subcapsular, anterior subcapsular, and nuclear cataracts. Cataracts occurred similarly in male mice fed high or low glycemic diets, and were also observed in 21-month male and female Nrf2 −/− mice fed ad libitum or 30% caloric restriction. Histological observation of 18-month cataractous lenses revealed significant disruption to fiber cell architecture and the retention of nuclei throughout the cortical region of the lens. However, fiber cell denucleation and initiation of lens differentiation was normal at birth, with the first abnormalities observed at 3 months. Conclusions Nrf2 −/− mice offer a tool to understand how defective antioxidant signaling causes multiple forms of cataract and may be useful for screening drugs to prevent or delay cataractogenesis in susceptible adults.
Collapse
Affiliation(s)
- Sheldon Rowan
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, United States.,Department of Ophthalmology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts, United States.,Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts, United States
| | - Shuhong Jiang
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, United States
| | - Sarah G Francisco
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, United States
| | - Laura C D Pomatto
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, Maryland, United States
| | - Zhiwei Ma
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Xiaodong Jiao
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Maria M Campos
- NEI Histology Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Sandeep Aryal
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Shaili D Patel
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Binapani Mahaling
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - S Amer Riazuddin
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Elia J Duh
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Salil A Lachke
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States.,Center for Bioinformatics and Computational Biology, University of Delaware, Newark, Delaware, United States
| | - J Fielding Hejtmancik
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, Maryland, United States
| | - Paul G FitzGerald
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, California, United States
| | - Allen Taylor
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, United States.,Department of Ophthalmology, Tufts University School of Medicine, Tufts University, Boston, Massachusetts, United States.,Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts, United States
| |
Collapse
|
14
|
Shihan MH, Novo SG, Wang Y, Sheppard D, Atakilit A, Arnold TD, Rossi NM, Faranda AP, Duncan MK. αVβ8 integrin targeting to prevent posterior capsular opacification. JCI Insight 2021; 6:145715. [PMID: 34554928 PMCID: PMC8663568 DOI: 10.1172/jci.insight.145715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 09/22/2021] [Indexed: 12/18/2022] Open
Abstract
Fibrotic posterior capsular opacification (PCO), a major complication of cataract surgery, is driven by transforming growth factor–β (TGF-β). Previously, αV integrins were found to be critical for the onset of TGF-β–mediated PCO in vivo; however, the functional heterodimer was unknown. Here, β8 integrin–conditional knockout (β8ITG-cKO) lens epithelial cells (LCs) attenuated their fibrotic responses, while both β5 and β6 integrin–null LCs underwent fibrotic changes similar to WT at 5 days post cataract surgery (PCS). RNA-Seq revealed that β8ITG-cKO LCs attenuated their upregulation of integrins and their ligands, as well as known targets of TGF-β–induced signaling, at 24 hours PCS. Treatment of β8ITG-cKO eyes with active TGF-β1 at the time of surgery rescued the fibrotic response. Treatment of WT mice with an anti-αVβ8 integrin function blocking antibody at the time of surgery ameliorated both canonical TGF-β signaling and LC fibrotic response PCS, and treatment at 5 days PCS, after surgically induced fibrotic responses were established, largely reversed this fibrotic response. These data suggest that αVβ8 integrin is a major regulator of TGF-β activation by LCs PCS and that therapeutics targeting αVβ8 integrin could be effective for fibrotic PCO prevention and treatment.
Collapse
Affiliation(s)
- Mahbubul H Shihan
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Samuel G Novo
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Yan Wang
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | | | | | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Nicole M Rossi
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Adam P Faranda
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| |
Collapse
|