1
|
Böhm M, Parekh M, Deshpande N, Cheung Q, Shatz N, Kumar V, Jurkunas UV. Mitochondria-Targeted Antioxidant (MitoQ) and Nontargeted Antioxidant (Idebenone) Mitigate Mitochondrial Dysfunction in Corneal Endothelial Cells. Cornea 2025; 44:492-503. [PMID: 39819808 DOI: 10.1097/ico.0000000000003801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/07/2024] [Indexed: 01/19/2025]
Abstract
PURPOSE To investigate the effectiveness of mitochondrial-targeted antioxidant mitoquinone (MitoQ) and nontargeted antioxidant idebenone (Idb) in alleviating mitochondrial dysfunction in corneal endothelial cells (CEnCs). METHODS In vitro experiments were conducted using immortalized normal human corneal endothelial cells (HCEnC-21T; SVN1-67F) and Fuchs endothelial corneal dystrophy (FECD) cells (SVF5-54F; SVF3-76M). Cells were pretreated with MitoQ or Idb and then exposed to menadione (MN) with simultaneous antioxidant treatment. Mitochondrial parameters were evaluated through adenosine triphosphate viability assays, JC-1 staining for mitochondrial membrane potential, and Tom-20 antibody staining for fragmentation, with analysis performed using ImageJ software. HCEnC-21T cells were additionally exposed to ultraviolet-A (25 J/cm 2 ) to assess drug effects under physiological stress. Mitochondrial fragmentation in FECD specimens was analyzed pre- and post-treatment with the drugs. Statistical analysis was conducted using 1-/2-way analysis of variance with post-hoc Tukey test. RESULTS MitoQ and Idb enhanced cell viability and mitochondrial membrane potential in both normal and FECD cells under MN-induced stress. Idb reduced MN-induced mitochondrial fragmentation by 32% more than MitoQ in HCEnC-21T cells and by 13% more in SVF5-54F cells. Under ultraviolet-A stress, Idb and MitoQ improved mitochondrial function by 31% and 25%, respectively, with MitoQ increasing mitochondrial function by 42% in FECD specimens. CONCLUSIONS Differential responses in mitochondrial dysfunction across cell lines highlight disease heterogeneity. MitoQ and Idb protected CEnCs from oxidative stress and improved mitochondrial bioenergetics, suggesting that mitochondrial-targeted antioxidants could be considered for mitochondrial dysfunction in CEnCs.
Collapse
Affiliation(s)
- Myriam Böhm
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA
- Department of Ophthalmology, Harvard Medical School, Boston, MA; and
- Department of Ophthalmology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Mohit Parekh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA
- Department of Ophthalmology, Harvard Medical School, Boston, MA; and
| | - Neha Deshpande
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA
- Department of Ophthalmology, Harvard Medical School, Boston, MA; and
| | - Queenie Cheung
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA
- Department of Ophthalmology, Harvard Medical School, Boston, MA; and
| | - Nathan Shatz
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA
- Department of Ophthalmology, Harvard Medical School, Boston, MA; and
| | - Varun Kumar
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA
- Department of Ophthalmology, Harvard Medical School, Boston, MA; and
| | - Ula V Jurkunas
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA
- Department of Ophthalmology, Harvard Medical School, Boston, MA; and
| |
Collapse
|
2
|
Sun F, Xi LWQ, Luu W, Enkhbat M, Neo D, Mehta JS, Peh GSL, Yim EKF. Preclinical Models for Studying Fuchs Endothelial Corneal Dystrophy. Cells 2025; 14:505. [PMID: 40214459 PMCID: PMC11988011 DOI: 10.3390/cells14070505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Fuchs Endothelial Corneal Dystrophy (FECD) is a corneal endothelial disease that causes microenvironment alterations and endothelial cell loss, which leads to vision impairment. It has a high global prevalence, especially in elderly populations. FECD is also one of the leading indications of corneal transplantation globally. Currently, there is no clearly defined canonical pathway for this disease, and it has been proposed that the combinatorial effects of genetic mutations and exogenous factors cause FECD. Clinical studies and observations have provided valuable knowledge and understanding of FECD, while preclinical studies are essential for gaining insights into disease progression and mechanisms for the development and testing of regenerative medicine therapies. In this review, we first introduce the proposed genetic and molecular pathologies of FECD. Notably, we discuss the impact of abnormal extracellular matrix deposition (guttata), endothelial-to-mesenchymal transition, cell senescence, and oxidative stress on the pathology and etiology of FECD. We review and summarize the in vitro cell models, ex vivo tissues, and in vivo animal models used to study FECD. The benefits and challenges of each model are also discussed.
Collapse
Affiliation(s)
- Fancheng Sun
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Lexie W. Q. Xi
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Wesley Luu
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Myagmartsend Enkhbat
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Dawn Neo
- Singapore Eye Research Institute, Singapore 169856, Singapore; (D.N.); (J.S.M.)
| | - Jodhbir S. Mehta
- Singapore Eye Research Institute, Singapore 169856, Singapore; (D.N.); (J.S.M.)
- Ophthalmology Academic Clinical Program, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Gary S. L. Peh
- Singapore Eye Research Institute, Singapore 169856, Singapore; (D.N.); (J.S.M.)
| | - Evelyn K. F. Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
- Centre for Biotechnology and Bioengineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON NL2 3G1, Canada
| |
Collapse
|
3
|
Ashraf S, Deshpande N, Cheung Q, Asabere JB, Wong RJ, Gauthier AG, Parekh M, Adhikari Y, Melangath G, Jurkunas UV. Modulation of ATM enhances DNA repair in G2/M phase of cell cycle and averts senescence in Fuchs endothelial corneal dystrophy. Commun Biol 2024; 7:1482. [PMID: 39523410 PMCID: PMC11551145 DOI: 10.1038/s42003-024-07179-1] [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: 05/02/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Fuchs Endothelial Corneal Dystrophy (FECD) is an aging disorder characterized by expedited loss of corneal endothelial cells (CEnCs) and heightened DNA damage compared to normal CEnCs. We previously established that ultraviolet-A (UVA) light causes DNA damage and leads to FECD phenotype in a non-genetic mouse model. Here, we demonstrate that acute treatment with chemical stressor, menadione, or physiological stressors, UVA, and catechol estrogen (4-OHE2), results in an early and increased activation of ATM-mediated DNA damage response in FECD compared to normal CEnCs. Acute stress with UVA and 4OHE2 causes (i) greater cell-cycle arrest and DNA repair in G2/M phase, and (ii) greater cytoprotective senescence in NQO1-/- compared to NQO1+/+ cells, which was reversed upon ATM inhibition. Chronic stress with UVA and 4OHE2 results in ATM-driven cell-cycle arrest in G0/G1 phase, reduced DNA repair, and cytotoxic senescence, due to sustained damage. Likewise, UVA-induced cell-cycle reentry, gamma-H2AX foci, and senescence-associated heterochromatin were reduced in Atm-null mice. Remarkably, inhibiting ATM activation with KU-55933 restored DNA repair in G2/M phase and attenuated senescence in chronic cellular model of FECD lacking NQO1. This study provides insights into understanding the pivotal role of ATM in regulating cell-cycle, DNA repair, and senescence, in oxidative-stress disorders like FECD.
Collapse
Affiliation(s)
- Shazia Ashraf
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Neha Deshpande
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Queenie Cheung
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Jeffrey Boakye Asabere
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Raymond Jeff Wong
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Alex G Gauthier
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Mohit Parekh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yadav Adhikari
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Geetha Melangath
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Ula V Jurkunas
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
4
|
Bannon ST, Shatz N, Wong R, Parekh M, Jurkunas UV. MitoQ relieves mitochondrial dysfunction in UVA and cigarette smoke-induced Fuchs endothelial corneal dystrophy. Exp Eye Res 2024; 247:110056. [PMID: 39179169 PMCID: PMC11426332 DOI: 10.1016/j.exer.2024.110056] [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: 04/11/2024] [Revised: 07/24/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Fuchs endothelial corneal dystrophy (FECD), a degenerative corneal condition, is characterized by the droplet-like accumulation of the extracellular matrix, known as guttae and progressive loss of corneal endothelial cells ultimately leading to visual distortion and glare. FECD can be influenced by environmental stressors and genetic conditions. However, the role of mitochondrial dysfunction for advancing FECD pathogenesis is not yet fully studied. Therefore, in the present study we sought to determine whether a combination of environmental stressors (ultraviolet-A (UVA) light and cigarette smoke condensate (CSC)) can induce mitochondrial dysfunction leading to FECD. We also investigated if MitoQ, a water-soluble antioxidant, can target mitochondrial dysfunction induced by UVA and CSC in human corneal endothelial cells mitigating FECD pathogenesis. We modeled the FECD by increasing exogenous oxidative stress with CSC (0.2%), UVA (25J/cm2) and a combination of UVA + CSC and performed a temporal analysis of their cellular and mitochondrial effects on HCEnC-21T immortalized cells in vitro before and after MitoQ (0.05 μM) treatment. Interestingly, we observed that a combination of UVA + CSC exposure increased mitochondrial ROS and fragmentation leading to a lower mitochondrial membrane potential and increased levels of cytochrome c release leading to apoptosis and cell death. MitoQ intervention successfully mitigated these effects and restored cell viability. The UVA + CSC model could be used to study stress induced mitochondrial dysfunction. Additionally, MitoQ can serve as a viable antioxidant in attenuating mitochondrial dysfunction, underscoring its potential as a molecular-focused treatment approach to combat FECD pathogenesis.
Collapse
Affiliation(s)
- Sean T Bannon
- Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Kinesiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Nathan Shatz
- Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Raymond Wong
- Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Mohit Parekh
- Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Ula V Jurkunas
- Schepens Eye Research Institute, Mass Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Anney P, Charpentier P, Proulx S. Influence of Intraocular Pressure on the Expression and Activity of Sodium-Potassium Pumps in the Corneal Endothelium. Int J Mol Sci 2024; 25:10227. [PMID: 39337712 PMCID: PMC11432950 DOI: 10.3390/ijms251810227] [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: 08/08/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
The corneal endothelium is responsible for pumping fluid out of the stroma in order to maintain corneal transparency, which depends in part on the expression and activity of sodium-potassium pumps. In this study, we evaluated how physiologic pressure and flow influence transcription, protein expression, and activity of Na+/K+-ATPase. Native and engineered corneal endothelia were cultured in a bioreactor in the presence of pressure and flow (hydrodynamic culture condition) or in a Petri dish (static culture condition). Transcription of ATP1A1 was assessed using qPCR, the expression of the α1 subunit of Na+/K+-ATPase was measured using Western blots and ELISA assays, and Na+/K+-ATPase activity was evaluated using an ATPase assay in the presence of ouabain. Results show that physiologic pressure and flow increase the transcription and the protein expression of Na+/K+-ATPase α1 in engineered corneal endothelia, while they remain stable in native corneal endothelia. Interestingly, the activity of Na+/K+-ATPase was increased in the presence of physiologic pressure and flow in both native and engineered corneal endothelia. These findings highlight the role of the in vivo environment on the functionality of the corneal endothelium.
Collapse
Affiliation(s)
- Princia Anney
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec City, QC G1J 1Z4, Canada
- Département d'Ophtalmologie et ORL-Chirurgie Cervico-Faciale, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Pascale Charpentier
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec City, QC G1J 1Z4, Canada
- Département d'Ophtalmologie et ORL-Chirurgie Cervico-Faciale, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Stéphanie Proulx
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec City, QC G1J 1Z4, Canada
- Département d'Ophtalmologie et ORL-Chirurgie Cervico-Faciale, Université Laval, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
6
|
Parekh M, Miall A, Chou A, Buhl L, Deshpande N, Price MO, Price FW, Jurkunas UV. Enhanced Migration of Fuchs Corneal Endothelial Cells by Rho Kinase Inhibition: A Novel Ex Vivo Descemet's Stripping Only Model. Cells 2024; 13:1218. [PMID: 39056800 PMCID: PMC11274477 DOI: 10.3390/cells13141218] [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: 06/04/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Descemet's Stripping Only (DSO) is a surgical technique that utilizes the peripheral corneal endothelial cell (CEnC) migration for wound closure. Ripasudil, a Rho-associated protein kinase (ROCK) inhibitor, has shown potential in DSO treatment; however, its mechanism in promoting CEnC migration remains unclear. We observed that ripasudil-treated immortalized normal and Fuchs endothelial corneal dystrophy (FECD) cells exhibited significantly enhanced migration and wound healing, particularly effective in FECD cells. Ripasudil upregulated mRNA expression of Snail Family Transcriptional Repressor (SNAI1/2) and Vimentin (VIM) while decreasing Cadherin (CDH1), indicating endothelial-to-mesenchymal transition (EMT) activation. Ripasudil activated Rac1, driving the actin-related protein complex (ARPC2) to the leading edge, facilitating enhanced migration. Ex vivo studies on cadaveric and FECD Descemet's membrane (DM) showed increased migration and proliferation of CEnCs after ripasudil treatment. An ex vivo DSO model demonstrated enhanced migration from the DM to the stroma with ripasudil. Coating small incision lenticule extraction (SMILE) tissues with an FNC coating mix and treating the cells in conjunction with ripasudil further improved migration and resulted in a monolayer formation, as detected by the ZO-1 junctional marker, thereby leading to the reduction in EMT. In conclusion, ripasudil effectively enhanced cellular migration, particularly in a novel ex vivo DSO model, when the stromal microenvironment was modulated. This suggests ripasudil as a promising adjuvant for DSO treatment, highlighting its potential clinical significance.
Collapse
Affiliation(s)
- Mohit Parekh
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; (M.P.)
| | - Annie Miall
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; (M.P.)
| | - Ashley Chou
- Faculty of Arts and Sciences, Harvard College, Boston, MA 02138, USA
| | - Lara Buhl
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Neha Deshpande
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; (M.P.)
| | | | - Francis W. Price
- Cornea Research Foundation of America, Indianapolis, IN 46260, USA
| | - Ula V. Jurkunas
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; (M.P.)
- Cornea and Refractive Surgery Service, Massachusetts Eye and Ear Infirmary, 243 Charles Street, Boston, MA 02114, USA
| |
Collapse
|
7
|
Monfort-Lanzas P, Rusu EC, Parrakova L, Karg CA, Kernbichler DE, Rieder D, Lackner P, Hackl H, Gostner JM. ExonSurfer: a web-tool to design primers at exon-exon junctions. BMC Genomics 2024; 25:594. [PMID: 38867172 PMCID: PMC11170769 DOI: 10.1186/s12864-024-10456-2] [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] [Received: 01/23/2024] [Accepted: 05/24/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Reverse transcription quantitative PCR (RT-qPCR) with intercalating dyes is one of the main techniques to assess gene expression levels used in basic and applied research as well as in diagnostics. However, primer design for RT-qPCR can be complex due to the high demands on primer quality. Primers are best placed on exon junctions, should avoid polymorphic regions, be specific to the target transcripts and also prevent genomic amplification accurately, among others. Current software tools manage to meet all the necessary criteria only insufficiently. Here, we present ExonSurfer, a novel, user-friendly web-tool for qPCR primer design. RESULTS ExonSurfer combines the different steps of the primer design process, encompassing target selection, specificity and self-complementarity assessment, and the avoidance of issues arising from polymorphisms. Amplification of potentially contaminating genomic DNA is avoided by designing primers on exon-exon junctions, moreover, a genomic alignment is performed to filter the primers accordingly and inform the user of any predicted interaction. In order to test the whole performance of the application, we designed primer pairs for 26 targets and checked both primer efficiency, amplicon melting temperature and length and confirmed the targeted amplicon by Sanger sequencing. Most of the tested primers accurately and selectively amplified the corresponding targets. CONCLUSION ExonSurfer offers a comprehensive end-to-end primer design, guaranteeing transcript-specific amplification. The user interface is intuitive, providing essential specificity and amplicon details. The tool can also be used by command line and the source code is available. Overall, we expect ExonSurfer to facilitate RT-qPCR set-up for researchers in many fields.
Collapse
Affiliation(s)
- Pablo Monfort-Lanzas
- Institute of Medical Biochemistry, Biochemical Immunotoxicology Group, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Elena Cristina Rusu
- Institute of Medical Biochemistry, Biochemical Immunotoxicology Group, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
- SeqPlexing SL, Valencia, Spain
- Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - Lucia Parrakova
- Institute of Medical Biochemistry, Biochemical Immunotoxicology Group, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Cornelia A Karg
- Institute of Medical Biochemistry, Biochemical Immunotoxicology Group, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Dorina-Elina Kernbichler
- Institute of Medical Biochemistry, Biochemical Immunotoxicology Group, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
- Institute of Medical Biochemistry, Core Facility Metabolomics II, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Dietmar Rieder
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Peter Lackner
- Department of Biosciences and Medical Biology, University of Salzburg, 5020, Salzburg, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Johanna M Gostner
- Institute of Medical Biochemistry, Biochemical Immunotoxicology Group, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria.
- Institute of Medical Biochemistry, Core Facility Metabolomics II, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria.
| |
Collapse
|
8
|
Yan J, Mehta S, Patel K, Dhupar N, Little N, Ong Tone S. Transcription factor 4 promotes increased corneal endothelial cellular migration by altering microtubules in Fuchs endothelial corneal dystrophy. Sci Rep 2024; 14:10276. [PMID: 38704483 PMCID: PMC11069521 DOI: 10.1038/s41598-024-61170-8] [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: 01/11/2024] [Accepted: 05/02/2024] [Indexed: 05/06/2024] Open
Abstract
Fuchs endothelial corneal dystrophy (FECD) is a complex corneal disease characterized by the progressive decline and morphological changes of corneal endothelial cells (CECs) that leads to corneal edema and vision loss. The most common mutation in FECD is an intronic CTG repeat expansion in transcription factor 4 (TCF4) that leads to its altered expression. Corneal endothelial wound healing occurs primarily through cell enlargement and migration, and FECD CECs have been shown to display increased migration speeds. In this study, we aim to determine whether TCF4 can promote cellular migration in FECD CECs. We generated stable CEC lines derived from FECD patients that overexpressed different TCF4 isoforms and investigated epithelial-to-mesenchymal (EMT) expression, morphological analysis and cellular migration speeds. We found that full length TCF4-B isoform overexpression promotes cellular migration in FECD CECs in an EMT-independent manner. RNA-sequencing identified several pathways including the negative regulation of microtubules, with TUBB4A (tubulin beta 4A class IVa) as the top upregulated gene. TUBB4A expression was increased in FECD ex vivo specimens, and there was altered expression of cytoskeleton proteins, tubulin and actin, compared to normal healthy donor ex vivo specimens. Additionally, there was increased acetylation and detyrosination of microtubules in FECD supporting that microtubule stability is altered in FECD and could promote cellular migration. Future studies could be aimed at investigating if targeting the cytoskeleton and microtubules would have therapeutic potential for FECD by promoting cellular migration and regeneration.
Collapse
Affiliation(s)
- Judy Yan
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
| | - Shanti Mehta
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Keya Patel
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Narisa Dhupar
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Ness Little
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Stephan Ong Tone
- Sunnybrook Health Sciences Center and Sunnybrook Research Institute, 2075 Bayview Avenue, M-wing, 1st Floor, Toronto, ON, M4N 3M5, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Canada.
| |
Collapse
|
9
|
Alemi H, Wang S, Blanco T, Kahale F, Singh RB, Ortiz G, Musayeva A, Yuksel E, Pang K, Deshpande N, Dohlman TH, Jurkunas UV, Yin J, Dana R. The Neuropeptide α-Melanocyte-Stimulating Hormone Prevents Persistent Corneal Edema following Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:150-164. [PMID: 37827217 PMCID: PMC10768537 DOI: 10.1016/j.ajpath.2023.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/03/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Corneal endothelial cells (CEnCs) regulate corneal hydration and maintain tissue transparency through their barrier and pump function. However, these cells exhibit limited regenerative capacity following injury. Currently, corneal transplantation is the only established therapy for restoring endothelial function, and there are no pharmacologic interventions available for restoring endothelial function. This study investigated the efficacy of the neuropeptide α-melanocyte-stimulating hormone (α-MSH) in promoting endothelial regeneration during the critical window between ocular injury and the onset of endothelial decompensation using an established murine model of injury using transcorneal freezing. Local administration of α-MSH following injury prevented corneal edema and opacity, reduced leukocyte infiltration, and limited CEnC apoptosis while promoting their proliferation. These results suggest that α-MSH has a proregenerative and cytoprotective function on CEnCs and shows promise as a therapy for the prevention and management of corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Hamid Alemi
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Shudan Wang
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Tomas Blanco
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Francesca Kahale
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Rohan B Singh
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Gustavo Ortiz
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Aytan Musayeva
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Erdem Yuksel
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Kunpeng Pang
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Neha Deshpande
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Thomas H Dohlman
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Ula V Jurkunas
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Jia Yin
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Reza Dana
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
10
|
Sasseville S, Karami S, Tchatchouang A, Charpentier P, Anney P, Gobert D, Proulx S. Biomaterials used for tissue engineering of barrier-forming cell monolayers in the eye. Front Bioeng Biotechnol 2023; 11:1269385. [PMID: 37840667 PMCID: PMC10569698 DOI: 10.3389/fbioe.2023.1269385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell monolayers that form a barrier between two structures play an important role for the maintenance of tissue functionality. In the anterior portion of the eye, the corneal endothelium forms a barrier that controls fluid exchange between the aqueous humor of the anterior chamber and the corneal stroma. This monolayer is central in the pathogenesis of Fuchs endothelial corneal dystrophy (FECD). FECD is a common corneal disease, in which corneal endothelial cells deposit extracellular matrix that increases the thickness of its basal membrane (Descemet's membrane), and forms excrescences (guttae). With time, there is a decrease in endothelial cell density that generates vision loss. Transplantation of a monolayer of healthy corneal endothelial cells on a Descemet membrane substitute could become an interesting alternative for the treatment of this pathology. In the back of the eye, the retinal pigment epithelium (RPE) forms the blood-retinal barrier, controlling fluid exchange between the choriocapillaris and the photoreceptors of the outer retina. In the retinal disease dry age-related macular degeneration (dry AMD), deposits (drusen) form between the RPE and its basal membrane (Bruch's membrane). These deposits hinder fluid exchange, resulting in progressive RPE cell death, which in turn generates photoreceptor cell death, and vision loss. Transplantation of a RPE monolayer on a Bruch's membrane/choroidal stromal substitute to replace the RPE before photoreceptor cell death could become a treatment alternative for this eye disease. This review will present the different biomaterials that are proposed for the engineering of a monolayer of corneal endothelium for the treatment of FECD, and a RPE monolayer for the treatment of dry AMD.
Collapse
Affiliation(s)
- Samantha Sasseville
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Samira Karami
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Ange Tchatchouang
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Pascale Charpentier
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Princia Anney
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Delphine Gobert
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre universitaire d’ophtalmologie (CUO), Hôpital du Saint-Sacrement, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Stéphanie Proulx
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
11
|
Wang Q, Zhang Z, Gao X. Effects of ophthalmic surface anesthetic alcaine on the proliferation and apoptosis of human corneal endothelial cells through HIF-1α regulation. Cell Tissue Bank 2023; 24:561-570. [PMID: 36572744 DOI: 10.1007/s10561-022-10057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 11/22/2022] [Indexed: 12/27/2022]
Abstract
The corneal endothelium is a monolayer, which mediates solute and water flux across the posterior corneal surface. Alcaine's main component proparacaine is paramount in human corneal endothelium (HCE) cell regulation. This study explored the mechanism of alcaine in regulating HCE cells. HCE cell morphology under gradient concentrations was observed by an optical microscope. Cell proliferation and viability were detected by MTT assay to determine the half inhibitory concentration (IC 50). Cell apoptosis rate, HIF-1α mRNA expression, and HIF-1α, p/t-JNK and Caspase-3 protein levels were detected by flow cytometry, RT-qPCR, and Western blot. After treatment with alcaine at 0.625-5 g/L concentration range for 24 h, HCE cells showed cytoplasmic vacuolation, cell shrinkage, separation from culture matrix, and eventual death. Alcaine treated-HCE cell proliferation was decreased in a dose-dependent manner. The IC 50 of alcaine was 1.26 g/L. After alcaine treatment, HCE cell apoptosis rate was promoted and HIF-1α levels in HCE cells were stimulated. Knockdown of HIF-1α partially annulled the effects of alcaine on inhibiting HCE cell proliferation and facilitating apoptosis. Alcaine might activate the JNK/caspase-3 pathway by increasing HIF-1α. The inhibition of the JNK/caspase-3 pathway partially abrogated the effects of alcaine on inhibiting HCE cell proliferation and promoting apoptosis. Alcaine might affect HCE cell proliferation and apoptosis by upregulating HIF-1α and activating the JNK/caspase-3 pathway.
Collapse
Affiliation(s)
- Quan Wang
- Department of Anesthesiology, Tianjin Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, No. 4 Gansu Road, Heping District, Tianjin, 300022, China
| | - Zhao Zhang
- Department of Anesthesiology, Tianjin Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, No. 4 Gansu Road, Heping District, Tianjin, 300022, China
| | - Xuesong Gao
- Department of Anesthesiology, Tianjin Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, No. 4 Gansu Road, Heping District, Tianjin, 300022, China.
| |
Collapse
|
12
|
Zidan AA, Zhu S, Elbasiony E, Najafi S, Lin Z, Singh RB, Naderi A, Yin J. Topical application of calcitonin gene-related peptide as a regenerative, antifibrotic, and immunomodulatory therapy for corneal injury. RESEARCH SQUARE 2023:rs.3.rs-3204385. [PMID: 37609298 PMCID: PMC10441448 DOI: 10.21203/rs.3.rs-3204385/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Calcitonin gene-related peptide (CGRP) is a multifunctional neuropeptide abundantly expressed by corneal nerves. Using a murine model of corneal mechanical injury, we found CGRP levels in the cornea to be significantly reduced after injury. Topical application of CGRP as an eye drop three times daily accelerates corneal epithelial wound closure, reduces corneal opacification, and prevents corneal edema after injury in vivo. We then used a series of in vitro and in vivo techniques to investigate the mechanisms underlying CGRP's functions. CGRP promotes corneal epithelial cell migration, proliferation, and the secretion of laminin. It reduces TGF-β1 signaling and prevents TGF-β1-mediated stromal fibroblast activation and tissue fibrosis. CGRP reduces corneal endothelial cell apoptosis and death, preserves cell density and morphology, and promotes their pump function, thus reducing edema. Lastly, CGRP reduces neutrophil infiltration, macrophage maturation, and the production of inflammatory cytokines in the cornea. Taken together, our results show that corneal nerve-derived CGRP plays a cyto-protective, pro-regenerative, anti-fibrotic, and anti-inflammatory role in corneal wound healing. Given that current treatment options for corneal injury and opacity are scarce, CGRP has significant therapeutic potential in this area of unmet medical needs. In addition, our results highlight the critical role of sensory nerves in ocular surface homeostasis and injury repair.
Collapse
Affiliation(s)
- Asmaa A. Zidan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| | - Shuyan Zhu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| | - Sheyda Najafi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| | - Zhirong Lin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| | - Rohan Bir Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| | - Amirreza Naderi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| | - Jia Yin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114
| |
Collapse
|
13
|
Chalmers BT, Merriman AF, Ruff AL. Considerations for the optimization of in vitro models of chloropicrin toxicity. Arch Toxicol 2023; 97:255-261. [PMID: 36329301 DOI: 10.1007/s00204-022-03400-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Chloropicrin (CP) is a common agricultural fumigant historically used as a chemical warfare agent and is a concern for potential use in warfare and terrorist applications. Our inability to effectively treat CP-induced injuries makes it essential to better understand CP toxicity. We set out to elucidate variables that must be understood to achieve optimal exposure conditions for in vitro investigations given that such models are important for the study of CP injury and potential therapeutics. To this end, we evaluated the effects of volatility, cell seeding density, and serum concentration of cell culture medium on CP toxicity in an immortalized human corneal epithelial cell line. We found that even with very dilute solutions, CP remained highly volatile, so much so that a 0.0019% CP solution resulted in 90% cell death at time 0, but was nearly nontoxic 45 min later. Not surprisingly, the CP-induced IL-8 response was shown to vary with cell viability in this experiment. After exposure with 0.00115% CP, cells that were 12% confluent experienced over 40% more cell death than cells exposed at 87% confluency. Exposure with the same CP dose in medium containing concentrations of fetal bovine serum (FBS) ranging from 0.1% to 15% exhibited a 17% difference in cell viability. Given that chemical toxicity can be significantly influenced by volatility, cell density, and serum content of cell culture medium, these phenomena should be explored during the development and optimization of toxicant exposure models.
Collapse
Affiliation(s)
- Bailey T Chalmers
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Gunpowder, MD, USA
| | - Abigail F Merriman
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Gunpowder, MD, USA
| | - Albert L Ruff
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Gunpowder, MD, USA.
| |
Collapse
|
14
|
Santerre K, Proulx S. Isolation efficiency of collagenase and EDTA for the culture of corneal endothelial cells. Mol Vis 2022; 28:331-339. [PMID: 36338664 PMCID: PMC9603909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 09/29/2022] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Tissue engineering of the corneal endothelium, as well as cell therapy, has been proposed as an alternative approach for the treatment of corneal endotheliopathies. These approaches require in vitro amplification of functional corneal endothelial cells (CECs). The goal of this study was to compare two common isolation methods, collagenase A and EDTA (EDTA), and determine whether they influence cell viability, morphology, and barrier function. METHODS Human eye bank research-grade corneas were used to isolate and cultivate CECs. All donors were more than 40 years old. Two Descemet membranes from the same donor were used separately to compare the collagenase A and EDTA cell isolation methods. The number of isolated cells, cell viability, morphology, and barrier functionality were compared. RESULTS A higher isolation efficiency of viable CECs and a higher circularity index (endothelial morphology) were obtained using collagenase A. Passage 3 cells presented similar barrier functionalities regardless of the isolation method. CONCLUSIONS This study showed that isolation of CECs using collagenase A yields higher isolation efficiency than EDTA, delaying the loss of endothelial morphology for early passage cells.
Collapse
Affiliation(s)
- Kim Santerre
- Centre de recherche du Centre hospitalier universitaire (CHU) de Québec – Université Laval, axe médecine régénératrice, Hôpital du Saint-Sacrement, Québec, QC, Canada,Département d’Ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Stéphanie Proulx
- Centre de recherche du Centre hospitalier universitaire (CHU) de Québec – Université Laval, axe médecine régénératrice, Hôpital du Saint-Sacrement, Québec, QC, Canada,Département d’Ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
15
|
Corneal Toxicity After Stinging by a Sea Anemone, Anthopleura uchidai: A Case Report With Confirmation by In Vitro Study. Cornea 2022; 41:1035-1037. [DOI: 10.1097/ico.0000000000002842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/18/2021] [Indexed: 11/26/2022]
|
16
|
Aparicio-Yuste R, Muenkel M, Clark AG, Gómez-Benito MJ, Bastounis EE. A Stiff Extracellular Matrix Favors the Mechanical Cell Competition that Leads to Extrusion of Bacterially-Infected Epithelial Cells. Front Cell Dev Biol 2022; 10:912318. [PMID: 35813215 PMCID: PMC9257086 DOI: 10.3389/fcell.2022.912318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Cell competition refers to the mechanism whereby less fit cells (“losers”) are sensed and eliminated by more fit neighboring cells (“winners”) and arises during many processes including intracellular bacterial infection. Extracellular matrix (ECM) stiffness can regulate important cellular functions, such as motility, by modulating the physical forces that cells transduce and could thus modulate the output of cellular competitions. Herein, we employ a computational model to investigate the previously overlooked role of ECM stiffness in modulating the forceful extrusion of infected “loser” cells by uninfected “winner” cells. We find that increasing ECM stiffness promotes the collective squeezing and subsequent extrusion of infected cells due to differential cell displacements and cellular force generation. Moreover, we discover that an increase in the ratio of uninfected to infected cell stiffness as well as a smaller infection focus size, independently promote squeezing of infected cells, and this phenomenon is more prominent on stiffer compared to softer matrices. Our experimental findings validate the computational predictions by demonstrating increased collective cell extrusion on stiff matrices and glass as opposed to softer matrices, which is associated with decreased bacterial spread in the basal cell monolayer in vitro. Collectively, our results suggest that ECM stiffness plays a major role in modulating the competition between infected and uninfected cells, with stiffer matrices promoting this battle through differential modulation of cell mechanics between the two cell populations.
Collapse
Affiliation(s)
- Raúl Aparicio-Yuste
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Instituto de Investigación en Ingeniería de Aragón (I3A), University of Zaragoza, Zaragoza, Spain
- Interfaculty Institute of Microbiology and Infection Medicine, Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany
| | - Marie Muenkel
- Interfaculty Institute of Microbiology and Infection Medicine, Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany
| | - Andrew G. Clark
- Institute of Cell Biology and Immunology/Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
- Center for Personalized Medicine, University of Tübingen, Tübingen, Germany
| | - María J. Gómez-Benito
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Instituto de Investigación en Ingeniería de Aragón (I3A), University of Zaragoza, Zaragoza, Spain
- *Correspondence: María J. Gómez-Benito, ; Effie E. Bastounis,
| | - Effie E. Bastounis
- Interfaculty Institute of Microbiology and Infection Medicine, Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany
- *Correspondence: María J. Gómez-Benito, ; Effie E. Bastounis,
| |
Collapse
|
17
|
Zakharova I, Saaya S, Shevchenko A, Stupnikova A, Zhiven' M, Laktionov P, Stepanova A, Romashchenko A, Yanshole L, Chernonosov A, Volkov A, Kizilova E, Zavjalov E, Chernyavsky A, Romanov A, Karpenko A, Zakian S. Mitomycin-Treated Endothelial and Smooth Muscle Cells Suitable for Safe Tissue Engineering Approaches. Front Bioeng Biotechnol 2022; 10:772981. [PMID: 35360387 PMCID: PMC8963790 DOI: 10.3389/fbioe.2022.772981] [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: 09/09/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
In our previous study, we showed that discarded cardiac tissue from the right atrial appendage and right ventricular myocardium is an available source of functional endothelial and smooth muscle cells for regenerative medicine and tissue engineering. In the study, we aimed to find out what benefits are given by vascular cells from cardiac explants used for seeding on vascular patches engrafted to repair vascular defects in vivo. Additionally, to make the application of these cells safer in regenerative medicine we tested an in vitro approach that arrested mitotic division to avoid the potential tumorigenic effect of dividing cells. A tissue-engineered construction in the form of a patch based on a polycaprolactone-gelatin scaffold and seeded with endothelial and smooth muscle cells was implanted into the abdominal aorta of immunodeficient SCID mice. Aortic patency was assessed using ultrasound, MRI, immunohistochemical and histological staining. Endothelial and smooth muscle cells were treated with mitomycin C at a therapeutic concentration of 10 μg/ml for 2 h with subsequent analysis of cell proliferation and function. The absence of the tumorigenic effect of mitomycin C-treated cells, as well as their angiogenic potential, was examined by injecting them into immunodeficient mice. Cell-containing patches engrafted in the abdominal aorta of immunodeficient mice form the vessel wall loaded with the appropriate cells and extracellular matrix, and do not interfere with normal patency. Endothelial and smooth muscle cells treated with mitomycin C show no tumorigenic effect in the SCID immunodeficient mouse model. During in vitro experiments, we have shown that treatment with mitomycin C does not lead to a decrease in cell viability. Despite the absence of proliferation, mitomycin C-treated vascular cells retain specific cell markers, produce specific extracellular matrix, and demonstrate the ability to stimulate angiogenesis in vivo. We pioneered an approach to arresting cell division with mitomycin C in endothelial and smooth muscle cells from cardiac explant, which prevents the risk of malignancy from dividing cells in vascular surgery. We believe that this approach to the fabrication of tissue-engineered constructs based on mitotically inactivated cells from waste postoperative material may be valuable to bring closer the development of safe cell products for regenerative medicine.
Collapse
Affiliation(s)
- Irina Zakharova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- *Correspondence: Irina Zakharova,
| | - Shoraan Saaya
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Alexander Shevchenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alena Stupnikova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Deparment of Natural Science, Novosibirsk State University, Novosibirsk, Russia
| | - Maria Zhiven'
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Pavel Laktionov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alena Stepanova
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Romashchenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Lyudmila Yanshole
- International Tomography Center,The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Chernonosov
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Volkov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Elena Kizilova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Deparment of Natural Science, Novosibirsk State University, Novosibirsk, Russia
| | - Evgenii Zavjalov
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Chernyavsky
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Alexander Romanov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Andrey Karpenko
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Suren Zakian
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
18
|
Lužnik Marzidovšek Z, Blanco T, Sun Z, Alemi H, Ortiz G, Nakagawa H, Chauhan SK, Taylor AW, Jurkunas UV, Yin J, Dana R. The Neuropeptide Alpha-Melanocyte-Stimulating Hormone Is Critical for Corneal Endothelial Cell Protection and Graft Survival after Transplantation. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:270-280. [PMID: 34774519 PMCID: PMC8908049 DOI: 10.1016/j.ajpath.2021.10.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 02/03/2023]
Abstract
Corneal transplantation is the most common form of tissue transplantation. The success of corneal transplantation mainly relies on the integrity of corneal endothelial cells (CEnCs), which maintain tissue transparency by pumping out excess water from the cornea. After transplantation, the rate of CEnC loss far exceeds that seen with normal aging, which can threaten sight. The underlying mechanisms are poorly understood. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide that is constitutively found in the aqueous humor with both cytoprotective and immunomodulatory effects. The curent study found high expression of melanocortin 1 receptor (MC1R), the receptor for α-MSH, on CEnCs. The effect of α-MSH/MC1R signaling on endothelial function and allograft survival in vitro and in vivo was investigated using MC1R signaling-deficient mice (Mc1re/e mice with a nonfunctional MC1R). Herein, the results indicate that in addition to its well-known immunomodulatory effect, α-MSH has cytoprotective effects on CEnCs after corneal transplantation, and the loss of MC1R signaling significantly decreases long-term graft survival in vivo. In conclusion, α-MSH/MC1R signaling is critical for CEnC function and graft survival after corneal transplantation.
Collapse
Affiliation(s)
- Zala Lužnik Marzidovšek
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts,Eye Hospital, University Medical Centre, Ljubljana, Slovenia
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Zhongmou Sun
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Hamid Alemi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Gustavo Ortiz
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Hayate Nakagawa
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Sunil K. Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Andrew W. Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts
| | - Ula V. Jurkunas
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Jia Yin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts,Address correspondence to Reza Dana, M.D., M.P.H., M.Sc., or Jia Yin, M.D., Ph.D., M.P.H., Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA 02114.
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts,Address correspondence to Reza Dana, M.D., M.P.H., M.Sc., or Jia Yin, M.D., Ph.D., M.P.H., Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA 02114.
| |
Collapse
|
19
|
Hussain NA, Figueiredo FC, Connon CJ. Use of biomaterials in corneal endothelial repair. Ther Adv Ophthalmol 2022; 13:25158414211058249. [PMID: 34988369 PMCID: PMC8721373 DOI: 10.1177/25158414211058249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 10/08/2021] [Indexed: 11/20/2022] Open
Abstract
Human corneal endothelium (HCE) is a single layer of hexagonal cells that lines the posterior surface of the cornea. It forms the barrier that separates the aqueous humor from the rest of the corneal layers (stroma and epithelium layer). This layer plays a fundamental role in maintaining the hydration and transparency of the cornea, which in turn ensures a clear vision. In vivo, human corneal endothelial cells (HCECs) are generally believed to be nonproliferating. In many cases, due to their nonproliferative nature, any damage to these cells can lead to further issues with Descemet’s membrane (DM), stroma and epithelium which may ultimately lead to hazy vision and blindness. Endothelial keratoplasties such as Descemet’s stripping automated endothelial keratoplasty (DSAEK) and Descemet’s membrane endothelial keratoplasty (DEK) are the standard surgeries routinely used to restore vision following endothelial failure. Basically, these two similar surgical techniques involve the replacement of the diseased endothelial layer in the center of the cornea by a healthy layer taken from a donor cornea. Globally, eye banks are facing an increased demand to provide corneas that have suitable features for transplantation. Consequently, it can be stated that there is a significant shortage of corneal grafting tissue; for every 70 corneas required, only 1 is available. Nowadays, eye banks face long waiting lists due to shortage of donors, seriously aggravated when compared with previous years, due to the global COVID-19 pandemic. Thus, there is an urgent need to find alternative and more sustainable sources for treating endothelial diseases, such as utilizing bioengineering to use of biomaterials as a remedy. The current review focuses on the use of biomaterials to repair the corneal endothelium. A range of biomaterials have been considered based on their promising results and outstanding features, including previous studies and their key findings in the context of each biomaterial.
Collapse
Affiliation(s)
- Noor Ahmed Hussain
- University of Jeddah, Jeddah, Saudi ArabiaBiosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Francisco C Figueiredo
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UKDepartment of Ophthalmology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Che J Connon
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
20
|
Rinkoski TA, Bahler CK, Pacheco JM, Khanna ML, Holmes DM, Roy Chowdhury U, Baratz KH, Patel SV, Maguire LJ, Wieben ED, Fautsch MP. Characterization of a dual media system for culturing primary normal and Fuchs endothelial corneal dystrophy (FECD) endothelial cells. PLoS One 2021; 16:e0258006. [PMID: 34587219 PMCID: PMC8480743 DOI: 10.1371/journal.pone.0258006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Primary cultures of human corneal endothelial cells (HCECs) are an important model system for studying the pathophysiology of corneal endothelium. The purpose of this study was to identify and validate an optimal primary culture model of normal and Fuchs endothelial corneal dystrophy (FECD) endothelial cells by comparing cell morphology and marker expression under different media conditions to in vivo donor tissues. Primary and immortalized HCECs, isolated from normal and FECD donors, were cultured in proliferation media (Joyce, M4, Bartakova) alone or sequentially with maturation media (F99, Stabilization 1, M5). CD56, CD73 and CD166 expressions were quantified in confluent and matured cell lines by flow cytometry. HCECs that were allowed to proliferate in Joyce's medium followed by maturation in low-mitogen containing media yielded cells with similar morphology to corneal endothelial tissues. Elevated expression of CD56 and CD166 and low expression of CD73 correlated with regular, hexagonal-like HCEC morphology. CD56:CD73 > 2.5 was most consistent with normal HCEC morphology and mimicked corneal endothelial tissue. Immortalization of normal HCECs by hTERT transduction showed morphology and CD56:CD73 ratios similar to parental cell lines. HCECs established from FECD donors showed reduced CD56:CD73 ratios compared to normal HCECs which coincided with reduced uniformity and regularity of cell monolayers. Overall, a dual media system with Joyce's medium for proliferation and a low-mitogen media for maturation, provided normal cultures with regular, hexagonal-like cell morphologies consistent with corneal endothelial cells in vivo. CD56:CD73 expression ratio >2.5 was predictive of in vivo-like cellular morphology.
Collapse
Affiliation(s)
- Tommy A. Rinkoski
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Cindy K. Bahler
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Johann M. Pacheco
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Maya L. Khanna
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - David M. Holmes
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Uttio Roy Chowdhury
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Keith H. Baratz
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Sanjay V. Patel
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Leo J. Maguire
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Eric D. Wieben
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
| | - Michael P. Fautsch
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
21
|
Choi M, Bonanno JA. Mitochondrial Targeting of the Ammonia-Sensitive Uncoupler SLC4A11 by the Chaperone-Mediated Carrier Pathway in Corneal Endothelium. Invest Ophthalmol Vis Sci 2021; 62:4. [PMID: 34499705 PMCID: PMC8434753 DOI: 10.1167/iovs.62.12.4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose SLC4A11, an electrogenic H+ transporter, is found in the plasma membrane and mitochondria of corneal endothelium. However, the underlying mechanism of SLC4A11 targeting to mitochondria is unknown. Methods The presence of mitochondrial targeting sequences was examined using in silico mitochondrial proteomic analyses. Thiol crosslinked peptide binding to SLC4A11 was screened by untargeted liquid chromatography/tandem mass spectrometry (LC-MS/MS) analysis. Direct protein interactions between SLC4A11 and chaperones were examined using coimmunoprecipitation analysis and proximity ligation assay. Knockdown or pharmacologic inhibition of chaperones in human corneal endothelial cells (HCECs) or mouse corneal endothelial cells (MCECs), ex vivo kidney, or HA-SLC4A11–transfected fibroblasts was performed to investigate the functional consequences of interfering with mitochondrial SLC4A11 trafficking. Results SLC4A11 does not contain canonical N-terminal mitochondrial targeting sequences. LC-MS/MS analysis showed that HSC70 and/or HSP90 are bound to HA-SLC4A11–transfected PS120 fibroblast whole-cell lysates or isolated mitochondria, suggesting trafficking through the chaperone-mediated carrier pathway. SLC4A11 and either HSP90 or HSC70 complexes are directly bound to the mitochondrial surface receptor, TOM70. Interference with this trafficking leads to dysfunctional mitochondrial glutamine catabolism and increased reactive oxygen species production. In addition, glutamine (Gln) use upregulated SLC4A11, HSP70, and HSP90 expression in whole-cell lysates or purified mitochondria of HCECs and HA-SLC4A11–transfected fibroblasts. Conclusions HSP90 and HSC70 are critical in mediating mitochondrial SLC4A11 translocation in corneal endothelial cells and kidney. Gln promotes SLC4A11 import to the mitochondria, and the continuous oxidative stress derived from Gln catabolism induced HSP70 and HSP90, protecting cells against oxidative stress.
Collapse
Affiliation(s)
- Moonjung Choi
- Vision Science Program, Indiana University, School of Optometry, Bloomington, Indiana, United States
| | - Joseph A Bonanno
- Vision Science Program, Indiana University, School of Optometry, Bloomington, Indiana, United States
| |
Collapse
|
22
|
Arras W, Vercammen H, Ní Dhubhghaill S, Koppen C, Van den Bogerd B. Proliferation Increasing Genetic Engineering in Human Corneal Endothelial Cells: A Literature Review. Front Med (Lausanne) 2021; 8:688223. [PMID: 34268324 PMCID: PMC8275833 DOI: 10.3389/fmed.2021.688223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
The corneal endothelium is the inner layer of the cornea. Despite comprising only a monolayer of cells, dysfunction of this layer renders millions of people visually impaired worldwide. Currently, corneal endothelial transplantation is the only viable means of restoring vision for these patients. However, because the supply of corneal endothelial grafts does not meet the demand, many patients remain on waiting lists, or are not treated at all. Possible alternative treatment strategies include intracameral injection of human corneal endothelial cells (HCEnCs), biomedical engineering of endothelial grafts and increasing the HCEnC density on grafts that would otherwise have been unsuitable for transplantation. Unfortunately, the limited proliferative capacity of HCEnCs proves to be a major bottleneck to make these alternatives beneficial. To tackle this constraint, proliferation enhancing genetic engineering is being investigated. This review presents the diverse array of genes that have been targeted by different genetic engineering strategies to increase the proliferative capacity of HCEnCs and their relevance for clinical and research applications. Together these proliferation-related genes form the basis to obtain a stable and safe supply of HCEnCs that can tackle the corneal endothelial donor shortage.
Collapse
Affiliation(s)
- Wout Arras
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Hendrik Vercammen
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sorcha Ní Dhubhghaill
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.,Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium.,Netherlands Institute for Innovative Ocular Surgery (NIIOS), Rotterdam, Netherlands
| | - Carina Koppen
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.,Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium
| | - Bert Van den Bogerd
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
23
|
Bastounis EE, Serrano-Alcalde F, Radhakrishnan P, Engström P, Gómez-Benito MJ, Oswald MS, Yeh YT, Smith JG, Welch MD, García-Aznar JM, Theriot JA. Mechanical competition triggered by innate immune signaling drives the collective extrusion of bacterially infected epithelial cells. Dev Cell 2021; 56:443-460.e11. [PMID: 33621492 PMCID: PMC7982222 DOI: 10.1016/j.devcel.2021.01.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/02/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
Intracellular pathogens alter their host cells' mechanics to promote dissemination through tissues. Conversely, host cells may respond to the presence of pathogens by altering their mechanics to limit infection. Here, we monitored epithelial cell monolayers infected with intracellular bacterial pathogens, Listeria monocytogenes or Rickettsia parkeri, over days. Under conditions in which these pathogens trigger innate immune signaling through NF-κB and use actin-based motility to spread non-lytically intercellularly, we found that infected cell domains formed three-dimensional mounds. These mounds resulted from uninfected cells moving toward the infection site, collectively squeezing the softer and less contractile infected cells upward and ejecting them from the monolayer. Bacteria in mounds were less able to spread laterally in the monolayer, limiting the growth of the infection focus, while extruded infected cells underwent cell death. Thus, the coordinated forceful action of uninfected cells actively eliminates large domains of infected cells, consistent with this collective cell response representing an innate immunity-driven process.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | | | - Prathima Radhakrishnan
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Patrik Engström
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - María J Gómez-Benito
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Mackenzi S Oswald
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jason G Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - José M García-Aznar
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
24
|
White TL, Deshpande N, Kumar V, Gauthier AG, Jurkunas UV. Cell cycle re-entry and arrest in G2/M phase induces senescence and fibrosis in Fuchs Endothelial Corneal Dystrophy. Free Radic Biol Med 2021; 164:34-43. [PMID: 33418109 PMCID: PMC7897316 DOI: 10.1016/j.freeradbiomed.2020.12.445] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022]
Abstract
Fuchs endothelial corneal dystrophy (FECD) is an age-related disease whereby progressive loss of corneal endothelial cells (CEnCs) leads to loss of vision. There is currently a lack of therapeutic interventions as the etiology of the disease is complex, with both genetic and environmental factors. In this study, we have provided further insights into the pathogenesis of the disease, showing a causal relationship between senescence and endothelial-mesenchymal transition (EMT) using in vitro and in vivo models. Ultraviolet A (UVA) light induced EMT and senescence in CEnCs. Senescent cells were arrested in G2/M phase of the cell cycle and responsible for the resulting profibrotic phenotype. Inhibiting ATR signaling and subsequently preventing G2/M arrest attenuated EMT. In vivo, UVA irradiation induced cell cycle re-entry in post mitotic CEnCs, resulting in senescence and fibrosis at 1- and 2-weeks post-UVA. Selectively eliminating senescent cells using the senolytic cocktail of dasatinib and quercetin attenuated UVA-induced fibrosis, highlighting the potential for a new therapeutic intervention for FECD.
Collapse
Affiliation(s)
- Tomas L White
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Neha Deshpande
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Varun Kumar
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Alex G Gauthier
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Ula V Jurkunas
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, 02114, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
25
|
Ong Tone S, Kocaba V, Böhm M, Wylegala A, White TL, Jurkunas UV. Fuchs endothelial corneal dystrophy: The vicious cycle of Fuchs pathogenesis. Prog Retin Eye Res 2021; 80:100863. [PMID: 32438095 PMCID: PMC7648733 DOI: 10.1016/j.preteyeres.2020.100863] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/05/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
Abstract
Fuchs endothelial corneal dystrophy (FECD) is the most common primary corneal endothelial dystrophy and the leading indication for corneal transplantation worldwide. FECD is characterized by the progressive decline of corneal endothelial cells (CECs) and the formation of extracellular matrix (ECM) excrescences in Descemet's membrane (DM), called guttae, that lead to corneal edema and loss of vision. FECD typically manifests in the fifth decades of life and has a greater incidence in women. FECD is a complex and heterogeneous genetic disease where interaction between genetic and environmental factors results in cellular apoptosis and aberrant ECM deposition. In this review, we will discuss a complex interplay of genetic, epigenetic, and exogenous factors in inciting oxidative stress, auto(mito)phagy, unfolded protein response, and mitochondrial dysfunction during CEC degeneration. Specifically, we explore the factors that influence cellular fate to undergo apoptosis, senescence, and endothelial-to-mesenchymal transition. These findings will highlight the importance of abnormal CEC-DM interactions in triggering the vicious cycle of FECD pathogenesis. We will also review clinical characteristics, diagnostic tools, and current medical and surgical management options for FECD patients. These new paradigms in FECD pathogenesis present an opportunity to develop novel therapeutics for the treatment of FECD.
Collapse
Affiliation(s)
- Stephan Ong Tone
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Viridiana Kocaba
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Myriam Böhm
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Adam Wylegala
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Tomas L White
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Ula V Jurkunas
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, MA, United States; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States; Department of Ophthalmology, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
26
|
Khalili M, Asadi M, Kahroba H, Soleyman MR, Andre H, Alizadeh E. Corneal endothelium tissue engineering: An evolution of signaling molecules, cells, and scaffolds toward 3D bioprinting and cell sheets. J Cell Physiol 2020; 236:3275-3303. [PMID: 33090510 DOI: 10.1002/jcp.30085] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/31/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
Cornea is an avascular and transparent tissue that focuses light on retina. Cornea is supported by the corneal-endothelial layer through regulation of hydration homeostasis. Restoring vision in patients afflicted with corneal endothelium dysfunction-mediated blindness most often requires corneal transplantation (CT), which faces considerable constrictions due to donor limitations. An emerging alternative to CT is corneal endothelium tissue engineering (CETE), which involves utilizing scaffold-based methods and scaffold-free strategies. The innovative scaffold-free method is cell sheet engineering, which typically generates cell layers surrounded by an intact extracellular matrix, exhibiting tunable release from the stimuli-responsive surface. In some studies, scaffold-based or scaffold-free technologies have been reported to achieve promising outcomes. However, yet some issues exist in translating CETE from bench to clinical practice. In this review, we compare different corneal endothelium regeneration methods and elaborate on the application of multiple cell types (stem cells, corneal endothelial cells, and endothelial precursors), signaling molecules (growth factors, cytokines, chemical compounds, and small RNAs), and natural and synthetic scaffolds for CETE. Furthermore, we discuss the importance of three-dimensional bioprinting strategies and simulation of Descemet's membrane by biomimetic topography. Finally, we dissected the recent advances, applications, and prospects of cell sheet engineering for CETE.
Collapse
Affiliation(s)
- Mostafa Khalili
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Asadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Houman Kahroba
- Biomedicine Institute, and Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Soleyman
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Helder Andre
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Effat Alizadeh
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Lovatt M, Kocaba V, Hui Neo DJ, Soh YQ, Mehta JS. Nrf2: A unifying transcription factor in the pathogenesis of Fuchs' endothelial corneal dystrophy. Redox Biol 2020; 37:101763. [PMID: 33099215 PMCID: PMC7578533 DOI: 10.1016/j.redox.2020.101763] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor, erythroid 2 like 2 (Nrf2), is an oxidative stress induced transcription factor that regulates cytoprotective gene expression. Thus, Nrf2 is essential for cellular redox homeostasis. Loss or dysregulation of Nrf2 expression has been implicated in the pathogenesis of degenerative diseases, including diseases of the cornea. One of the most common diseases of the cornea in which Nrf2 is implicated is Fuchs' endothelial cornea dystrophy (FECD). FECD is the leading indication for corneal transplantation; and is associated with a loss of corneal endothelial cell (CEC) function. In this review, we propose that Nrf2 is an essential regulator of CEC function. Furthermore, we demonstrate that deficiency of Nrf2 function is a hallmark of FECD. In addition, we advocate that pharmacological targeting of Nrf2 as a possible therapy for FECD.
Collapse
Affiliation(s)
- Matthew Lovatt
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore.
| | - Viridiana Kocaba
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore; Netherlands Institute for Innovative Ocular Surgery (NIIOS), Rotterdam, the Netherlands
| | - Dawn Jing Hui Neo
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore
| | - Yu Qiang Soh
- Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore
| | - Jodhbir S Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore; Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore.
| |
Collapse
|
28
|
Holub A, Mousa S, Abdolahi A, Godugu K, Tu XM, Brenna JT, Block RC. The effects of aspirin and N-3 fatty acids on telomerase activity in adults with diabetes mellitus. Nutr Metab Cardiovasc Dis 2020; 30:1795-1799. [PMID: 32723580 PMCID: PMC7494550 DOI: 10.1016/j.numecd.2020.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 01/02/2023]
Abstract
Type 2 Diabetes mellitus is associated with aging and shortened telomere length. Telomerase replaces lost telomeric repeats at the ends of chromosomes and is necessary for the replicative immortality of cells. Aspirin and the n3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are commonly used therapies in people with type 2 diabetes for reducing cardiovascular disease events, though their relation to telomerase activity is not well studied. We explored the effects of aspirin, EPA + DHA, and the combined effects of aspirin and EPA + DHA treatment on telomerase activity in 30 adults with diabetes mellitus. EPA and DHA ingestion alone increased telomerase activity then a decrease occurred with the addition of aspirin consumption. Crude (F-stat = 2.09, p = 0.13) and adjusted (F-stat = 2.20, p = 0.14) analyses of this decrease showed signs of a trend. These results suggest that aspirin has an adverse effect on aging in diabetics who have relatively high EPA and DHA ingestion.
Collapse
Affiliation(s)
- Ashley Holub
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Shaker Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Amir Abdolahi
- Monitoring Analytics and Therapeutic Care, Clinical Science Innovations, Philips Healthcare, Cambridge, MA, USA
| | - Kavitha Godugu
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Xin M Tu
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California, San Diego, CA, USA
| | - J Thomas Brenna
- Pediatrics and Chemistry at the Dell Medical School of the University of Texas at Austin, Austin, TX, USA
| | - Robert C Block
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Cardiology Division, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
29
|
Liu J, Wen Y, Luo W, Liu Y, Sha X. Human Amniotic Epithelial Cells Promote the Proliferation of Human Corneal Endothelial Cells by Regulating Telomerase Activity via the Wnt/β-catenin Pathway. Curr Eye Res 2020; 46:159-167. [PMID: 32631162 DOI: 10.1080/02713683.2020.1792508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Human amniotic epithelial cells (HAECs) have regenerative properties and low immunogenicity, which have enabled their use without immune rejection in regenerative medicine applications, such as wound repair, corneal surgery and burn repair. The aim of this study was to explore the potential role of HAECs in the proliferation of human corneal endothelial cells (HCEnCs) and the possible mechanism of regulation. METHODS HAECs and HCEnCs were isolated from donated tissue samples and were cultured; the collected HAEC culture medium (HAEC-Me) was added to the human corneal endothelium medium (CEM) to establish the HAEC-CM system. HCEnCs were cultured in CEM, 20%HAEC-Me, 20% HAEC-CM, 20% HAEC-CM supplemented with a GSK-3β inhibitor TWS119 or CEM supplemented with TWS119. Then, cell proliferation, apoptosis, cell cycle progression, telomerase activity, and Wnt/β-catenin pathway-related protein levels were assessed. RESULTS We found that the HCEnCs cultured in the 20% HAEC-CM had increased proliferative capacity, telomerase activity and β-catenin and Tcf4 expression levels, and they had a decrease in the rate of apoptosis and α-SMA expression when they were compared with the HCEnCs cultured in the 20% HAEC-Me. After GSK-3β was inhibited by TWS119, HCEnCs cultured in CEM or 20% HAEC-CM had an increased proliferative capacity, telomerase activity, β-catenin/Tcf4 expression and a decreased α-SMA expression, and they had a decreased apoptotic rate. CONCLUSIONS These data indicate that the human amniotic epithelial cells microenvironment can promote the proliferation of human corneal endothelial cells, which may be related to regulating telomerase activity and epithelial-to-mesenchymal transition (EMT) via the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jiayan Liu
- Ophthalmic Center, The Second Affiliated Hospital of Guangzhou Medical University , Guangzhou, China.,Department of Ophthalmology, The Sixth Affiliated Hospital of Guangzhou Medical University , Qingyuan, China
| | - Ye Wen
- Ophthalmic Center, The Second Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Wei Luo
- Ophthalmic Center, The Second Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Yingying Liu
- Ophthalmic Center, The Second Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Xiangyin Sha
- Ophthalmic Center, The Second Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| |
Collapse
|
30
|
Bioengineering of Human Corneal Endothelial Cells from Single- to Four-Dimensional Cultures. CURRENT OPHTHALMOLOGY REPORTS 2020. [DOI: 10.1007/s40135-020-00244-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Phenotypic and functional characterization of corneal endothelial cells during in vitro expansion. Sci Rep 2020; 10:7402. [PMID: 32366916 PMCID: PMC7198491 DOI: 10.1038/s41598-020-64311-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
The advent of cell culture-based methods for the establishment and expansion of human corneal endothelial cells (CEnC) has provided a source of transplantable corneal endothelium, with a significant potential to challenge the one donor-one recipient paradigm. However, concerns over cell identity remain, and a comprehensive characterization of the cultured CEnC across serial passages has not been performed. To this end, we compared two established CEnC culture methods by assessing the transcriptomic changes that occur during in vitro expansion. In confluent monolayers, low mitogenic culture conditions preserved corneal endothelial cell state identity better than culture in high mitogenic conditions. Expansion by continuous passaging induced replicative cell senescence. Transcriptomic analysis of the senescent phenotype identified a cell senescence signature distinct for CEnC. We identified activation of both classic and new cell signaling pathways that may be targeted to prevent senescence, a significant barrier to realizing the potential clinical utility of in vitro expansion.
Collapse
|
32
|
Ramachandran C, Gupta P, Hazra S, Mandal BB. In Vitro Culture of Human Corneal Endothelium on Non-Mulberry Silk Fibroin Films for Tissue Regeneration. Transl Vis Sci Technol 2020; 9:12. [PMID: 32818099 PMCID: PMC7396167 DOI: 10.1167/tvst.9.4.12] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose The purpose of this study was to determine if non-mulberry varieties of silk are suitable for the culture of corneal endothelium (CE). Methods Aqueous silk fibroin derived from Philosamia ricini (PR), Antheraea assamensis (AA), and Bombyx mori (BM) were cast as approximately 15 µm films with and without pores on which human CE cells were cultured. Tensile strength, elasticity, transmittance in visible range, and degradation properties of the films were characterised. Adhesion of CE to the silk films was quantified using MTT assay in addition to quantifying the number and area of focal adhesions using paxillin. Expression of CE markers was determined at the gene and protein levels using PCR and immunostaining, respectively. Barrier integrity of the cultured cells was measured as permeability to FITC dextran (10 kDa) in the presence or absence of thrombin. Results The films exhibited robust tensile strength, >95% transmittance and a refractive index comparable to the native cornea. BM degraded significantly faster when compared to PR and AA. A comparison between the three varieties of silk showed that significantly more cells were adhered to PR and AA than to BM. This was also reflected in the expression of stable focal adhesions on PR and AA, thus enabling the formation of intact monolayers of cells on these varieties unlike on BM. Treatment with thrombin significantly increased cellular permeability to dextran. Conclusions Our data shows that PR and AA varieties sufficiently support the growth and function of CE cells. This could be attributed to the presence of natural cell binding motifs (RGD) in these varieties. Translational Relevance Development of a suitable carrier for engineering the CE to address a major clinical requirement of healthy donor tissues for transplantation.
Collapse
Affiliation(s)
- Charanya Ramachandran
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Prerak Gupta
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Swatilekha Hazra
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India.,Manipal University, Manipal, India
| | - Biman B Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
33
|
Miyajima T, Melangath G, Zhu S, Deshpande N, Vasanth S, Mondal B, Kumar V, Chen Y, Price MO, Price FW, Rogan EG, Zahid M, Jurkunas UV. Loss of NQO1 generates genotoxic estrogen-DNA adducts in Fuchs Endothelial Corneal Dystrophy. Free Radic Biol Med 2020; 147:69-79. [PMID: 31857234 PMCID: PMC6939626 DOI: 10.1016/j.freeradbiomed.2019.12.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/14/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Fuchs Endothelial Corneal Dystrophy (FECD) is an age-related genetically complex disease characterized by increased oxidative DNA damage and progressive degeneration of corneal endothelial cells (HCEnCs). FECD has a greater incidence and advanced phenotype in women, suggesting a possible role of hormones in the sex-driven differences seen in the disease pathogenesis. In this study, catechol estrogen (4-OHE2), the byproduct of estrogen metabolism, induced genotoxic estrogen-DNA adducts formation, macromolecular DNA damage, and apoptotic cell death in HCEnCs; these findings were potentiated by menadione (MN)-mediated reactive oxygen species (ROS). Expression of NQO1, a key enzyme that neutralizes reactive estrogen metabolites, was downregulated in FECD, indicating HCEnC susceptibility to reactive estrogen metabolism in FECD. NQO1 deficiency in vitro exacerbated the estrogen-DNA adduct formation and loss of cell viability, which was rescued by the supplementation of N-acetylcysteine, a ROS scavenger. Notably, overexpression of NQO1 in HCEnCs treated with MN and 4-OHE2 quenched the ROS formation, thereby reducing the DNA damage and endothelial cell loss. This study signifies a pivotal role for NQO1 in mitigating the macromolecular oxidative DNA damage arising from the interplay between intracellular ROS and impaired endogenous estrogen metabolism in post-mitotic ocular tissue cells. A dysfunctional Nrf2-NQO1 axis in FECD renders HCEnCs susceptible to catechol estrogens and estrogen-DNA adducts formation. This novel study highlights the potential role of NQO1-mediated estrogen metabolite genotoxicity in explaining the higher incidence of FECD in females.
Collapse
Affiliation(s)
- Taiga Miyajima
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Dokkyo Medical University, Tochigi, 321-0293, Japan
| | - Geetha Melangath
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Shan Zhu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Neha Deshpande
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Shivakumar Vasanth
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Bodhisattwa Mondal
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Varun Kumar
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Yuming Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Marianne O Price
- Price Vision Group and Cornea Research Foundation of America, Indianapolis, IN, USA
| | - Francis W Price
- Price Vision Group and Cornea Research Foundation of America, Indianapolis, IN, USA
| | - Eleanor G Rogan
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Muhammad Zahid
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ula V Jurkunas
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Liu C, Miyajima T, Melangath G, Miyai T, Vasanth S, Deshpande N, Kumar V, Ong Tone S, Gupta R, Zhu S, Vojnovic D, Chen Y, Rogan EG, Mondal B, Zahid M, Jurkunas UV. Ultraviolet A light induces DNA damage and estrogen-DNA adducts in Fuchs endothelial corneal dystrophy causing females to be more affected. Proc Natl Acad Sci U S A 2020; 117:573-583. [PMID: 31852820 PMCID: PMC6955350 DOI: 10.1073/pnas.1912546116] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fuchs endothelial corneal dystrophy (FECD) is a leading cause of corneal endothelial (CE) degeneration resulting in impaired visual acuity. It is a genetically complex and age-related disorder, with higher incidence in females. In this study, we established a nongenetic FECD animal model based on the physiologic outcome of CE susceptibility to oxidative stress by demonstrating that corneal exposure to ultraviolet A (UVA) recapitulates the morphological and molecular changes of FECD. Targeted irradiation of mouse corneas with UVA induced reactive oxygen species (ROS) production in the aqueous humor, and caused greater CE cell loss, including loss of ZO-1 junctional contacts and corneal edema, in female than male mice, characteristic of late-onset FECD. UVA irradiation caused greater mitochondrial DNA (mtDNA) and nuclear DNA (nDNA) damage in female mice, indicative of the sex-driven differential response of the CE to UVA, thus accounting for more severe phenotype in females. The sex-dependent effect of UVA was driven by the activation of estrogen-metabolizing enzyme CYP1B1 and formation of reactive estrogen metabolites and estrogen-DNA adducts in female but not male mice. Supplementation of N-acetylcysteine (NAC), a scavenger of reactive oxygen species (ROS), diminished the morphological and molecular changes induced by UVA in vivo. This study investigates the molecular mechanisms of environmental factors in FECD pathogenesis and demonstrates a strong link between UVA-induced estrogen metabolism and increased susceptibility of females for FECD development.
Collapse
Affiliation(s)
- Cailing Liu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Taiga Miyajima
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Geetha Melangath
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Takashi Miyai
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Shivakumar Vasanth
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Neha Deshpande
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Varun Kumar
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Stephan Ong Tone
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Reena Gupta
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Shan Zhu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Dijana Vojnovic
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Yuming Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Eleanor G Rogan
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198-4388
| | - Bodhiswatta Mondal
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198-4388
| | - Muhammad Zahid
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198-4388
| | - Ula V Jurkunas
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114;
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
35
|
Pan P, Weisenberger DJ, Zheng S, Wolf M, Hwang DG, Rose-Nussbaumer JR, Jurkunas UV, Chan MF. Aberrant DNA methylation of miRNAs in Fuchs endothelial corneal dystrophy. Sci Rep 2019; 9:16385. [PMID: 31705138 PMCID: PMC6841734 DOI: 10.1038/s41598-019-52727-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
Homeostatic maintenance of corneal endothelial cells is essential for maintenance of corneal deturgescence and transparency. In Fuchs endothelial corneal dystrophy (FECD), an accelerated loss and dysfunction of endothelial cells leads to progressively severe visual impairment. An abnormal accumulation of extracellular matrix (ECM) is a distinctive hallmark of the disease, however the molecular pathogenic mechanisms underlying this phenomenon are not fully understood. Here, we investigate genome-wide and sequence-specific DNA methylation changes of miRNA genes in corneal endothelial samples from FECD patients. We discover that miRNA gene promoters are frequent targets of aberrant DNA methylation in FECD. More specifically, miR-199B is extensively hypermethylated and its mature transcript miR-199b-5p was previously found to be almost completely silenced in FECD. Furthermore, we find that miR-199b-5p directly and negatively regulates Snai1 and ZEB1, two zinc finger transcription factors that lead to increased ECM deposition in FECD. Taken together, these findings suggest a novel epigenetic regulatory mechanism of matrix protein production by corneal endothelial cells in which miR-199B hypermethylation leads to miR-199b-5p downregulation and thereby the increased expression of its target genes, including Snai1 and ZEB1. Our results support miR-199b-5p as a potential therapeutic target to prevent or slow down the progression of FECD disease.
Collapse
Affiliation(s)
- Peipei Pan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel J Weisenberger
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA
| | - Siyu Zheng
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Marie Wolf
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - David G Hwang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.,Francis I. Proctor Foundation, University of California, San Francisco, CA, USA
| | - Jennifer R Rose-Nussbaumer
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.,Francis I. Proctor Foundation, University of California, San Francisco, CA, USA
| | - Ula V Jurkunas
- Department of Ophthalmology, Harvard Medical School, and Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, USA
| | - Matilda F Chan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA. .,Francis I. Proctor Foundation, University of California, San Francisco, CA, USA.
| |
Collapse
|
36
|
Kocaba V, Katikireddy KR, Gipson I, Price MO, Price FW, Jurkunas UV. Association of the Gutta-Induced Microenvironment With Corneal Endothelial Cell Behavior and Demise in Fuchs Endothelial Corneal Dystrophy. JAMA Ophthalmol 2019; 136:886-892. [PMID: 29852040 DOI: 10.1001/jamaophthalmol.2018.2031] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance The number and size of guttae increase over time in Fuchs endothelial corneal dystrophy (FECD); however, the association between these physical parameters and disease pathogenesis is unclear. Objective To determine the role of guttae in corneal endothelial cell function. Design, Settings, and Participants In an in vitro model, cells from a human corneal endothelial cell line, HCENC-21T, were seeded on decellularized normal (n = 30) and FECD (n = 70) endothelial basement (Descemet) membranes (DMs). Normal human corneas were sent to our laboratory from 3 sources. The study took place at the Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, and was performed from September 2015 to July 2017. Normal DMs were obtained from 3 different tissue banks and FECD-DMs were obtained from patients undergoing endothelial keratoplasty in 2 departments. Main Outcomes and Measures Endothelial cell shape, growth, and migration were assessed by live-cell imaging, and gene expression analysis as a function of guttae diameter was assessed by laser capture microscopy. Results Mean (SD) age of normal-DMs donors was 65.6 (4.4) years (16 women [53%]), and mean (SD) age of FECD-DMs donors was 68.9 (10.6) years (43 women [61%]). Cells covered a greater area (mean [SD], 97.7% [8.5%]) with a greater mean (SD) number of cells (2083 [153] cells/mm2) on the normal DMs compared with the FECD DMs (72.8% [11%]; P = .02 and 1541 [221] cells/mm2 221/mm2; P = .01, respectively). Differences in endothelial cell growth over guttae were observed on FECD DMs depending on the guttae diameter. Guttae with a mean (SD) diameter of 10.5 (2.9) μm did not impede cell growth, whereas those with a diameter of 21.1 (4.9) μm were covered only by the cell cytoplasm. Guttae with the largest mean (SD) diameter, 31.8 (3.8) μm, were not covered by cells, which instead surrounded them in a rosette pattern. Moreover, cells adjacent to large guttae upregulated αSMA, N-cadherin, Snail1, and NOX4 genes compared with ones grown on normal DMs or small guttae. Furthermore, large guttae induced TUNEL-positive apoptosis in a rosette pattern, similar to ex vivo FECD specimens. Conclusions and Relevance These findings highlight the important role of guttae in endothelial cell growth, migration, and survival. These data suggest that cell therapy procedures in FECD might be guided by the diameter of the host guttae if subsequent clinical studies confirm these laboratory findings.
Collapse
Affiliation(s)
- Viridiana Kocaba
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston
| | - Kishore Reddy Katikireddy
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston
| | - Ilene Gipson
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston
| | | | | | - Ula V Jurkunas
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston
| |
Collapse
|
37
|
Islam MM, Sharifi R, Mamodaly S, Islam R, Nahra D, Abusamra DB, Hui PC, Adibnia Y, Goulamaly M, Paschalis EI, Cruzat A, Kong J, Nilsson PH, Argüeso P, Mollnes TE, Chodosh J, Dohlman CH, Gonzalez-Andrades M. Effects of gamma radiation sterilization on the structural and biological properties of decellularized corneal xenografts. Acta Biomater 2019; 96:330-344. [PMID: 31284096 PMCID: PMC7043233 DOI: 10.1016/j.actbio.2019.07.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/21/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022]
Abstract
To address the shortcomings associated with corneal transplants, substantial efforts have been focused on developing new modalities such as xenotransplantion. Xenogeneic corneas are anatomically and biomechanically similar to the human cornea, yet their applications require prior decellularization to remove the antigenic components to avoid rejection. In the context of bringing decellularized corneas into clinical use, sterilization is a crucial step that determines the success of the transplantation. Well-standardized sterilization methods, such as gamma irradiation (GI), have been applied to decellularized porcine corneas (DPC) to avoid graft-associated infections in human recipients. However, little is known about the effect of GI on decellularized corneal xenografts. Here, we evaluated the radiation effect on the ultrastructure, optical, mechanical and biological properties of DPC. Transmission electron microscopy revealed that gamma irradiated decellularized porcine cornea (G-DPC) preserved its structural integrity. Moreover, the radiation did not reduce the optical properties of the tissue. Neither DPC nor G-DPC led to further activation of complement system compared to native porcine cornea when exposed to plasma. Although, DPC were mechanically comparable to the native tissue, GI increased the mechanical strength, tissue hydrophobicity and resistance to enzymatic degradation. Despite these changes, human corneal epithelial, stromal, endothelial and hybrid neuroblastoma cells grew and differentiated on DPC and G-DPC. Thus, GI may achieve effective tissue sterilization without affecting critical properties that are essential for corneal transplant survival.
Collapse
Affiliation(s)
- Mohammad Mirazul Islam
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Roholah Sharifi
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shamina Mamodaly
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rakibul Islam
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway
| | - Daniel Nahra
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dina B Abusamra
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pui Chuen Hui
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yashar Adibnia
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Yeditepe University School of Medicine, Istanbul, Turkey
| | - Mehdi Goulamaly
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eleftherios I Paschalis
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Andrea Cruzat
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jing Kong
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Per H Nilsson
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway; Linnaeus Center for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Pablo Argüeso
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway; Research Laboratory, Nordland Hospital, Bodø, and Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Norway; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - James Chodosh
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Claes H Dohlman
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Miguel Gonzalez-Andrades
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Department of Ophthalmology, Reina Sofia University Hospital and University of Cordoba, Cordoba, Spain.
| |
Collapse
|
38
|
Miyai T, Vasanth S, Melangath G, Deshpande N, Kumar V, Benischke AS, Chen Y, Price MO, Price FW, Jurkunas UV. Activation of PINK1-Parkin-Mediated Mitophagy Degrades Mitochondrial Quality Control Proteins in Fuchs Endothelial Corneal Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2061-2076. [PMID: 31361992 DOI: 10.1016/j.ajpath.2019.06.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 05/25/2019] [Accepted: 06/11/2019] [Indexed: 11/19/2022]
Abstract
Corneal endothelium (CE) is a monolayer of mitochondria-rich cells, critical for maintaining corneal transparency compatible with clear vision. Fuchs endothelial corneal dystrophy (FECD) is a heterogeneous, genetically complex disorder, where oxidative stress plays a key role in the rosette formation during the degenerative loss of CE. Increased mitochondrial fragmentation along with excessive mitophagy activation has been detected in FECD; however, the mechanism of aberrant mitochondrial dynamics in CE cell loss is poorly understood. Here, the role of oxidative stress in mitophagy activation in FECD is investigated. Immunoblotting of FECD ex vivo specimens revealed an accumulation of PINK1 and phospho-Parkin (Ser65) along with loss of total Parkin and total Drp1. Similarly, modeling of rosette formation with menadione (MN), led to phospho-Parkin accumulation in fragmented mitochondria resulting in mitophagy-induced mitochondrial clearance, albeit possibly in a PINK1-independent manner. Loss of PINK1, phospho-Drp1, and total Drp1 was prominent after MN-induced oxidative stress, but not after mitochondrial depolarization by carbonyl cyanide m-chlorophenyl hydrazone. Moreover, MN-induced mitophagy led to degradation of Parkin along with sequestration of Drp1 and PINK1 that was rescued by mitophagy inhibition. This study shows that in FECD, intracellular oxidative stress induces Parkin-mediated mitochondrial fragmentation where endogenous Drp1 and PINK1 are sequestered and degraded by mitophagy during degenerative loss of post-mitotic cells of ocular tissue.
Collapse
Affiliation(s)
- Takashi Miyai
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Shivakumar Vasanth
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Geetha Melangath
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Neha Deshpande
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Varun Kumar
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Anne-Sophie Benischke
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Yuming Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | | | | | - Ula V Jurkunas
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
39
|
Ogando DG, Choi M, Shyam R, Li S, Bonanno JA. Ammonia sensitive SLC4A11 mitochondrial uncoupling reduces glutamine induced oxidative stress. Redox Biol 2019; 26:101260. [PMID: 31254733 PMCID: PMC6604051 DOI: 10.1016/j.redox.2019.101260] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/07/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
SLC4A11 is a NH3 sensitive membrane transporter with H+ channel-like properties that facilitates Glutamine catabolism in Human and Mouse corneal endothelium (CE). Loss of SLC4A11 activity induces oxidative stress and cell death, resulting in Congenital Hereditary Endothelial Dystrophy (CHED) with corneal edema and vision loss. However, the mechanism by which SLC4A11 prevents ROS production and protects CE is unknown. Here we demonstrate that SLC4A11 is localized to the inner mitochondrial membrane of CE and SLC4A11 transfected PS120 fibroblasts, where it acts as an NH3-sensitive mitochondrial uncoupler that enhances glutamine-dependent oxygen consumption, electron transport chain activity, and ATP levels by suppressing damaging Reactive Oxygen Species (ROS) production. In the presence of glutamine, Slc4a11-/- (KO) mouse CE generate significantly greater mitochondrial superoxide, a greater proportion of damaged depolarized mitochondria, and more apoptotic cells than WT. KO CE can be rescued by MitoQ, reducing NH3 production by GLS1 inhibition or dimethyl αKetoglutarate supplementation, or by BAM15 mitochondrial uncoupling. Slc4a11 KO mouse corneal edema can be partially reversed by αKetoglutarate eye drops. Moreover, we demonstrate that this role for SLC4A11 is not specific to CE cells, as SLC4A11 knockdown in glutamine-addicted colon carcinoma cells reduced glutamine catabolism, increased ROS production, and inhibited cell proliferation. Overall, our studies reveal a unique metabolic mechanism that reduces mitochondrial oxidative stress while promoting glutamine catabolism.
Collapse
Affiliation(s)
- Diego G Ogando
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Moonjung Choi
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Rajalekshmy Shyam
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Shimin Li
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Joseph A Bonanno
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States.
| |
Collapse
|
40
|
ZEB1 insufficiency causes corneal endothelial cell state transition and altered cellular processing. PLoS One 2019; 14:e0218279. [PMID: 31194824 PMCID: PMC6564028 DOI: 10.1371/journal.pone.0218279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
The zinc finger e-box binding homeobox 1 (ZEB1) transcription factor is a master regulator of the epithelial to mesenchymal transition (EMT), and of the reverse mesenchymal to epithelial transition (MET) processes. ZEB1 plays an integral role in mediating cell state transitions during cell lineage specification, wound healing and disease. EMT/MET are characterized by distinct changes in molecular and cellular phenotype that are generally context-independent. Posterior polymorphous corneal dystrophy (PPCD), associated with ZEB1 insufficiency, provides a new biological context in which to understand and evaluate the classic EMT/MET paradigm. PPCD is characterized by a cadherin-switch and transition to an epithelial-like transcriptomic and cellular phenotype, which we study in a cell-based model of PPCD generated using CRISPR-Cas9-mediated ZEB1 knockout in corneal endothelial cells (CEnCs). Transcriptomic and functional studies support the hypothesis that CEnC undergo a MET-like transition in PPCD, termed endothelial to epithelial transition (EnET), and lead to the conclusion that EnET may be considered a corollary to the classic EMT/MET paradigm.
Collapse
|
41
|
Hutcheon AEK, Zieske JD, Guo X. 3D in vitro model for human corneal endothelial cell maturation. Exp Eye Res 2019; 184:183-191. [PMID: 30980816 DOI: 10.1016/j.exer.2019.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/15/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022]
Abstract
Corneal endothelium is a cellular monolayer positioned on the Descemet's membrane at the anterior cornea, and it plays a critical role in maintaining corneal clarity. Our present study examines the feasibility of utilizing our 3-dimensional (3D) corneal stromal construct, which consists of human corneal fibroblasts (HCF) and their self-assembled matrix, to observe the development and maturation of human corneal endothelial cells (HCEndoCs) in a co-culture model. Three-dimensional HCF constructs were created by growing the HCFs on Transwell membranes in Eagles' minimum essential medium (EMEM) + 10% FBS + 0.5 mM Vitamin C (VitC) for about 4 weeks. HCEndoCs, either primary (pHCEndoC) or cell line (HCEndoCL), were either seeded in chamber slides, directly on the Transwell membranes, or on the 3D HCF constructs and cultivated for 5 days or 2 weeks. The HCEndoCs that were seeded directly on the Transwell membranes were exposed indirectly to HCF by culturing the HCF on the plate beneath the membrane. Cultures were examined for morphology and ultrastructure using light and transmission electron microscopy (TEM). In addition, indirect-immunofluorescence microscopy (IF) was used to examine tight junction formation (ZO-1), maturation (ALDH1A1), basement membrane formation (Laminin), cell proliferation (Ki67), cell death (caspase-3), and fibrotic response (CTGF). As expected, both pHCEndoCs and HCEndoCLs formed monolayers on the constructs; however, the morphology of the HCEndoCLs appeared to be similar to that seen in vivo, uniform and closely packed, whereas the pHCEndoCs remained elongated. The IF data showed that laminin localization was present in the HCEndoCs' cytoplasm as cell-cell contact increased, and when they were grown in the 3D co-culture, the beginnings of what appears to be a continuous DM-like structure was observed. In addition, in co-cultures, ALDH1A1-positive HCEndoCs were present, ZO-1 expression localized within the tight junctions, minimal numbers of HCEndoCs were Ki67-or Caspase-3-positive, and CTGF was positive in both the HCEndoCs cytoplasm and the matrix of the co-culture. Also, laminin localization was stimulated in HCEndoCs upon indirect stimuli secreted by HCF. The present data suggests our 3D co-culture model is useful for studying corneal endothelium maturation in vitro since the co-culture promotes new DM-like formation, HCEndoCs develop in vivo-like characteristics, and the fibrotic response is activated. Our current findings are applicable to understanding the implications of corneal endothelial injection therapy, such as if the abnormal DM has to be removed from the patient, the newly injected endothelial cells will seed onto the wound area and deposit a new DM-like membrane. However, caution should be observed and as much of the normal DM should be left intact since removal of the DM can cause a posterior stromal fibrotic response.
Collapse
Affiliation(s)
- Audrey E K Hutcheon
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| | - James D Zieske
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Xiaoqing Guo
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| |
Collapse
|
42
|
Alka K, Casey JR. Ophthalmic Nonsteroidal Anti-Inflammatory Drugs as a Therapy for Corneal Dystrophies Caused by SLC4A11 Mutation. Invest Ophthalmol Vis Sci 2019; 59:4258-4267. [PMID: 30140924 DOI: 10.1167/iovs.18-24301] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose SLC4A11 is a plasma membrane protein of corneal endothelial cells. Some mutations of the SLC4A11 gene result in SLC4A11 protein misfolding and failure to mature to the plasma membrane. This gives rise to some cases of Fuchs' endothelial corneal dystrophy (FECD) and congenital hereditary endothelial dystrophy (CHED). We screened ophthalmic nonsteroidal anti-inflammatory drugs (NSAIDs) for their ability to correct SLC4A11 folding defects. Methods Five ophthalmic NSAIDs were tested for their therapeutic potential in some genetic corneal dystrophy patients. HEK293 cells expressing CHED and FECD-causing SLC4A11 mutants were grown on 96-well dishes in the absence or presence of NSAIDs. Ability of NSAIDs to correct mutant SLC4A11 cell-surface trafficking was assessed with a bioluminescence resonance energy transfer (BRET) assay and by confocal microscopy. The ability of mutant SLC4A11-expressing cells to mediate water flux (SLC4A11 mediates water flux across the corneal endothelial cell basolateral membrane as part of the endothelial water pump) was measured upon treatment with ophthalmic NSAIDs. Results BRET-assays revealed significant rescue of SLC4A11 mutants to the cell surface by 4 of 5 NSAIDs tested. The NSAIDs, diclofenac and nepafenac, were effective in moving endoplasmic reticulum-retained missense mutant SLC4A11 to the cell surface, as measured by confocal immunofluorescence. Among intracellular-retained SLC4A11 mutants, 20 of 30 had significant restoration of cell surface abundance upon treatment with diclofenac. Diclofenac restored mutant SLC4A11 water flux activity to the level of wild-type SLC4A11 in some cases. Conclusions These results encourage testing diclofenac eye drops as a treatment for corneal dystrophy in patients whose disease is caused by some SLC4A11 missense mutations.
Collapse
Affiliation(s)
- Kumari Alka
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joseph R Casey
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
43
|
Soragni E, Petrosyan L, Rinkoski TA, Wieben ED, Baratz KH, Fautsch MP, Gottesfeld JM. Repeat-Associated Non-ATG (RAN) Translation in Fuchs' Endothelial Corneal Dystrophy. Invest Ophthalmol Vis Sci 2019; 59:1888-1896. [PMID: 29677349 PMCID: PMC5886103 DOI: 10.1167/iovs.17-23265] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose The strongest genetic association with Fuchs' endothelial corneal dystrophy (FECD) is the presence of an intronic (CTG·CAG)n trinucleotide repeat (TNR) expansion in the transcription factor 4 (TCF4) gene. Repeat-associated non-ATG (RAN) translation, an unconventional protein translation mechanism that does not require an initiating ATG, has been described in many TNR expansion diseases, including myotonic dystrophy type 1 (DM1). Given the similarities between DM1 and FECD, we wished to determine whether RAN translation occurs in FECD. Methods Antibodies against peptides in the C-terminus of putative RAN translation products from TCF4 were raised and validated by Western blotting and immunofluorescence (IF). CTG·CAG repeats of various lengths in the context of the TCF4 gene were cloned in frame with a 3× FLAG tag and transfected in human cells. IF with antipeptide and anti-FLAG antibodies, as well as cytotoxicity and cell proliferation assays, were performed in these transfected cells. Corneal endothelium derived from patients with FECD was probed with validated antibodies by IF. Results CTG·CAG repeats in the context of the TCF4 gene are transcribed and translated via non-ATG initiation in transfected cells and confer toxicity to an immortalized corneal endothelial cell line. An antipeptide antibody raised against the C-terminus of the TCF4 poly-cysteine frame recognized RAN translation products by IF in cells transfected with CTG·CAG repeats and in FECD corneal endothelium. Conclusions Expanded CTG·CAG repeats in the context of the third intron of TCF4 are transcribed and translated via non-ATG initiation, providing evidence for RAN translation in corneal endothelium of patients with FECD.
Collapse
Affiliation(s)
- Elisabetta Soragni
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Lina Petrosyan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Tommy A Rinkoski
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Eric D Wieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States
| | - Keith H Baratz
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Michael P Fautsch
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Joel M Gottesfeld
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| |
Collapse
|
44
|
Abstract
The corneal endothelium (CE) is vital for maintaining the water balance and clarity of the cornea. The CE is a cell layer that is particularly susceptible to aging because of its postmitotic arrest, high metabolic activity involving pumping of ions, and lifelong exposure to ultraviolet light. Despite gradual age-related cell loss, a sufficient number of CE cells are preserved during the lifespan of an individual. However, in conditions such as Fuchs endothelial corneal dystrophy (FECD), permanent loss of CE cells leads to corneal edema and loss of vision requiring corneal transplantation. FECD is a genetic and oxidative stress disorder manifested by abnormal cell-matrix interactions and expedited cellular aging culminating in cellular death. Because the endothelium has minimal replicative capacity in vivo and an inability to replace its genome, it is particularly prone to cumulative DNA damage acquired throughout life. In FECD, the underlying genetic defects make the CE genome even more vulnerable to this damage, to the point of causing mitochondrial dysfunction, mitochondrial membrane potential loss, and excessive mitophagy activation. Endogenous and exogenous intracellular stressors alter the synthetic footprint of CE cells, leading to endothelial-mesenchymal transition and secretion of aberrant extracellular matrix (in the form of guttae), resembling scar formation in other organs. In turn, the guttae or endothelial scars contribute to a vicious cycle of FECD pathogenesis and, by further inducing endothelial-mesenchymal transition and oxidant-antioxidant imbalance, perpetuate the molecular changes of the degenerating endothelium.
Collapse
|
45
|
Satitpitakul V, Sun Z, Suri K, Amouzegar A, Katikireddy KR, Jurkunas UV, Kheirkhah A, Dana R. Vasoactive Intestinal Peptide Promotes Corneal Allograft Survival. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2016-2024. [PMID: 30097165 DOI: 10.1016/j.ajpath.2018.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/11/2018] [Accepted: 05/22/2018] [Indexed: 12/17/2022]
Abstract
Corneal transplantation is the most prevalent form of tissue transplantation. The success of corneal transplantation mainly relies on the integrity of corneal endothelial cells (CEnCs), which maintain graft transparency. CEnC density decreases significantly after corneal transplantation even in the absence of graft rejection. To date, different strategies have been used to enhance CEnC survival. The neuropeptide vasoactive intestinal peptide (VIP) improves CEnC integrity during donor cornea tissue storage and protects CEnCs against oxidative stress-induced apoptosis. However, little is known about the effect of exogenous administration of VIP on corneal transplant outcomes. We found that VIP significantly accelerates endothelial wound closure and suppresses interferon-γ- and tumor necrosis factor-α-induced CEnC apoptosis in vitro in a dose-dependent manner. In addition, we found that intracameral administration of VIP to mice undergoing syngeneic corneal transplantation with endothelial injury increases CEnC density and decreases graft opacity scores. Finally, using a mouse model of allogeneic corneal transplantation, we found for the first time that treatment with VIP significantly suppresses posttransplantation CEnC loss and improves corneal allograft survival.
Collapse
Affiliation(s)
- Vannarut Satitpitakul
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Zhongmou Sun
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Kunal Suri
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Kishore R Katikireddy
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Ula V Jurkunas
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Ahmad Kheirkhah
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
46
|
Abstract
PURPOSE Alternative splice isoforms of TCF4, a gene implicated in Fuchs corneal dystrophy, have been identified in multiple human tissues outside of the eye. The aim of this study was to identify the transcriptional profile of TCF4 in the corneal endothelium. METHODS We extracted RNA from the donor corneal endothelium and performed rapid amplification of cDNA ends. We tested the expression pattern of 1 newly identified isoform (7b) in a panel of cDNA derived from multiple human tissues and included cDNA from corneal endothelial (CE) and retinal pigment epithelial cell lines. To further delineate differential expression of TCF4 splice variants that span CTG18.1, we analyzed expression of 6 alternative splice isoforms that are transcribed from either exon 2 or 3 in RNA extracted from the corneal endothelium of 3 normal donors and a CE cell line. RESULTS We identified 11 different isoforms in control CE tissue, including 1 isoform (7b) not reported previously. This isoform is enriched specifically in the corneal endothelium and placenta compared with other tissues in a panel of human cDNA. CONCLUSIONS We demonstrate the complex expression profile of TCF4 in the human corneal endothelium and reveal expression of alternative splice variants of TCF4.
Collapse
|
47
|
Hypoxia and the Prolyl Hydroxylase Inhibitor FG-4592 Protect Corneal Endothelial Cells From Mechanical and Perioperative Surgical Stress. Cornea 2018; 37:501-507. [PMID: 29504956 DOI: 10.1097/ico.0000000000001430] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To determine whether hypoxia preconditioning can protect corneal endothelial cells from mechanical stress and perioperative procedures mimicking Descemet stripping automated endothelial keratoplasty (DSAEK). METHODS Preconditioning was delivered by 2 hours of 0.5% oxygen incubation in a hypoxia chamber or by exposure to the prolyl hydroxylase inhibitor FG-4592, which prevents hypoxia-inducible factor-1 alpha degradation. Damage to whole corneas was produced by brief sonication. To mimic use with DSAEK, FG-4592-preconditioned and control donor corneas were dissected with a microkeratome, and the posterior donor button was pulled through a transplant insertion device (Busin glide). The area of endothelial damage was determined by trypan blue staining. RESULTS In all cases, hypoxia preconditioning or incubation with FG-4592 protected corneal endothelial cells from death by mechanical stress. Hypoxia-preconditioned human and rabbit corneas showed 19% and 29% less cell loss, respectively, relative to controls, which were both significant at P < 0.05. FG-4592 preconditioning reduced endothelial cell loss associated with preparation and insertion of DSAEK grafts by 23% relative to the control (P < 0.01). CONCLUSIONS These results support the hypothesis that preconditioning by hypoxia or exposure to FG-4592 improves corneal endothelial cell survival and may also provide protection during surgical trauma.
Collapse
|
48
|
Kinoshita S, Koizumi N, Ueno M, Okumura N, Imai K, Tanaka H, Yamamoto Y, Nakamura T, Inatomi T, Bush J, Toda M, Hagiya M, Yokota I, Teramukai S, Sotozono C, Hamuro J. Injection of Cultured Cells with a ROCK Inhibitor for Bullous Keratopathy. N Engl J Med 2018. [PMID: 29539291 DOI: 10.1056/nejmoa1712770] [Citation(s) in RCA: 331] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Corneal endothelial cell (CEC) disorders, such as Fuchs's endothelial corneal dystrophy, induce abnormal corneal hydration and result in corneal haziness and vision loss known as bullous keratopathy. We investigated whether injection of cultured human CECs supplemented with a rho-associated protein kinase (ROCK) inhibitor into the anterior chamber could increase CEC density. METHODS We performed an uncontrolled, single-group study involving 11 persons who had received a diagnosis of bullous keratopathy and had no detectable CECs. Human CECs were cultured from a donor cornea; a total of 1×106 passaged cells were supplemented with a ROCK inhibitor (final volume, 300 μl) and injected into the anterior chamber of the eye that was selected for treatment. After the procedure, patients were placed in a prone position for 3 hours. The primary outcome was restoration of corneal transparency, with a CEC density of more than 500 cells per square millimeter at the central cornea at 24 weeks after cell injection. Secondary outcomes were a corneal thickness of less than 630 μm and an improvement in best corrected visual acuity equivalent to two lines or more on a Landolt C eye chart at 24 weeks after cell injection. RESULTS At 24 weeks after cell injection, we recorded a CEC density of more than 500 cells per square millimeter (range, 947 to 2833) in 11 of the 11 treated eyes (100%; 95% confidence interval [CI], 72 to 100), of which 10 had a CEC density exceeding 1000 cells per square millimeter. A corneal thickness of less than 630 μm (range, 489 to 640) was attained in 10 of the 11 treated eyes (91%; 95% CI, 59 to 100), and an improvement in best corrected visual acuity of two lines or more was recorded in 9 of the 11 treated eyes (82%; 95% CI, 48 to 98). CONCLUSIONS Injection of human CECs supplemented with a ROCK inhibitor was followed by an increase in CEC density after 24 weeks in 11 persons with bullous keratopathy. (Funded by the Japan Agency for Medical Research and Development and others; UMIN number, UMIN000012534 .).
Collapse
Affiliation(s)
- Shigeru Kinoshita
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Noriko Koizumi
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Morio Ueno
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Naoki Okumura
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Kojiro Imai
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Hiroshi Tanaka
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Yuji Yamamoto
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Takahiro Nakamura
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Tsutomu Inatomi
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - John Bush
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Munetoyo Toda
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Michio Hagiya
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Isao Yokota
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Satoshi Teramukai
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Chie Sotozono
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| | - Junji Hamuro
- From the Departments of Frontier Medical Science and Technology for Ophthalmology (S.K., T.N., M.T., M.H.), Ophthalmology (M.U., K.I., H.T., Y.Y., T.I., J.B., C.S., J.H.), and Biostatistics (I.Y., S.T.), Kyoto Prefectural University of Medicine, and the Department of Biomedical Engineering, Doshisha University (N.K., N.O.) - both in Kyoto, Japan
| |
Collapse
|
49
|
Katikireddy KR, White TL, Miyajima T, Vasanth S, Raoof D, Chen Y, Price MO, Price FW, Jurkunas UV. NQO1 downregulation potentiates menadione-induced endothelial-mesenchymal transition during rosette formation in Fuchs endothelial corneal dystrophy. Free Radic Biol Med 2018; 116:19-30. [PMID: 29294389 PMCID: PMC5815941 DOI: 10.1016/j.freeradbiomed.2017.12.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/08/2017] [Accepted: 12/29/2017] [Indexed: 12/18/2022]
Abstract
Fuchs endothelial corneal dystrophy (FECD) is a genetic and oxidative stress disorder of post-mitotic human corneal endothelial cells (HCEnCs), which normally exhibit hexagonal shape and form a compact monolayer compatible with normal corneal functioning and clear vision. FECD is associated with increased DNA damage, which in turn leads to HCEnC loss, resulting in the formation rosettes and aberrant extracellular matrix (ECM) deposition in the form of pro-fibrotic guttae. Since the mechanism of ECM deposition in FECD is currently unknown, we aimed to investigate the role of endothelial-mesenchymal transition (EMT) in FECD using a previously established cellular in vitro model that recapitulates the characteristic rosette formation, by employing menadione (MN)-induced oxidative stress. We demonstrate that MN treatment alone, or a combination of MN and TGF-β1 induces reactive oxygen species (ROS), cell death, and EMT in HCEnCs during rosette formation, resulting in upregulation of EMT- and FECD-associated markers such as Snail1, N-cadherin, ZEB1, and transforming growth factor-beta-induced (TGFβI), respectively. Additionally, FECD ex vivo specimens displayed a loss of organized junctional staining of plasma membrane-bound N-cadherin, with corresponding increase in fibronectin and Snail1 compared to ex vivo controls. Addition of N-acetylcysteine (NAC) downregulated all EMT markers and abolished rosette formation. Loss of NQO1, a metabolizing enzyme of MN, led to greater increase in intracellular ROS levels as well as a significant upregulation of Snail1, fibronectin, and N-cadherin compared to normal cells, indicating that NQO1 regulates Snail1-mediated EMT. This study provides first line evidence that MN-induced oxidative stress leads to EMT in corneal endothelial cells, and the effect of which is further potentiated when redox cycling activity of MN is enhanced by the absence of NQO1. Given that NAC inhibits Snail-mediated EMT, this may be a potential therapeutic intervention for FECD.
Collapse
Affiliation(s)
- Kishore Reddy Katikireddy
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | - Tomas L White
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | - Taiga Miyajima
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | - Shivakumar Vasanth
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | - Duna Raoof
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; Harvard Eye Associates, Laguna Hills, CA 92660, USA
| | - Yuming Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | - Marianne O Price
- Cornea Research Foundation of America, Indianapolis, IN 46260, USA
| | - Francis W Price
- Cornea Research Foundation of America, Indianapolis, IN 46260, USA
| | - Ula V Jurkunas
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA.
| |
Collapse
|
50
|
Rönkkö S, Vellonen KS, Järvinen K, Toropainen E, Urtti A. Human corneal cell culture models for drug toxicity studies. Drug Deliv Transl Res 2017; 6:660-675. [PMID: 27613190 PMCID: PMC5097077 DOI: 10.1007/s13346-016-0330-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In vivo toxicity and absorption studies of topical ocular drugs are problematic, because these studies involve invasive tissue sampling and toxic effects in animal models. Therefore, different human corneal models ranging from simple monolayer cultures to three-dimensional models have been developed for toxicological prediction with in vitro models. Each system has its own set of advantages and disadvantages. Use of non-corneal cells, inadequate characterization of gene-expression profiles, and accumulation of genomic aberrations in human corneal models are typical drawbacks that decrease their reliability and predictive power. In the future, further improvements are needed for verifying comparable expression profiles and cellular properties of human corneal models with their in vivo counterparts. A rapidly expanding stem cell technology combined with tissue engineering may give future opportunities to develop new tools in drug toxicity studies. One approach may be the production of artificial miniature corneas. In addition, there is also a need to use large-scale profiling approaches such as genomics, transcriptomics, proteomics, and metabolomics for understanding of the ocular toxicity.
Collapse
Affiliation(s)
- Seppo Rönkkö
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.Box 1627, 70211, Kuopio, Finland
| | - Kati-Sisko Vellonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.Box 1627, 70211, Kuopio, Finland
| | - Kristiina Järvinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.Box 1627, 70211, Kuopio, Finland
| | - Elisa Toropainen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.Box 1627, 70211, Kuopio, Finland
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.Box 1627, 70211, Kuopio, Finland. .,Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.
| |
Collapse
|