1
|
Nishant P, Gurnani B, Singh P, Sinha S, Kaur K, Kumar A, Sinha RK. Current concepts and recent trends in endothelial keratoplasty. World J Transplant 2025; 15:102507. [DOI: 10.5500/wjt.v15.i2.102507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial keratoplasty (EK) is defined as an umbrella term comprising methods for selective surgical replacement of corneal endothelium and adjacent corneal tissue, which retains healthy portions of a patient's cornea while replacing diseased innermost corneal layer(s) with healthy donor tissue, to achieve corneal dehydration and transparency before the onset of irreversible stromal edema and permanent loss of corneal clarity. Recently, the pathophysiology of corneal decompensation is increasingly being researched upon. Consequent improvement in pharmacotherapy is progressively leading to reduction in the indications of EK. In addition, EK techniques have progressed towards using thinner tissue, optimizing visual outcomes. Improvements have enabled better donor tissue formulation, usage, and attachment, and surgical modifications have enhanced the tissue utilization in difficult clinical scenarios lowering failure and rejection. However, challenges are encountered in various complex clinical scenarios including eyes with prior intraocular surgery, complex anterior chamber anatomy, glaucoma, ocular surface disease etc. These complexities demand tailored surgical strategies, including modifications in graft handling, instrumentation, and postoperative management to ensure success. Attention to these details and addressing patient-specific factors can help improve outcomes in these difficult cases. The choice of procedure depends on multiple factors, including the surgeon's experience, patient's ocular anatomy, and the specific clinical scenario. This review article encompasses the recent developments in this field presenting a comprehensive picture of our modern understanding of the indications, contraindications, surgical techniques, clinical situations, community aspects and future directions pertaining to EK.
Collapse
Affiliation(s)
- Prateek Nishant
- Department of Ophthalmology-Refractive Surgery, Uvea and Neuro-Ophthalmology, Akhand Jyoti Eye Hospital, Mastichak, Saran 841219, Bihar, India
| | - Bharat Gurnani
- Department of Cataract, Cornea, External Diseases, Trauma, Ocular Surface, Refractive Surgery and Contact Lens, Gomabai Netralaya and Research Center, Neemuch 458441, Madhya Pradesh, India
| | - Prabhakar Singh
- Department of Ophthalmology-Cornea and Ocular Surface, All India Institute of Medical Sciences, Kalyani 741245, West Bengal, India
| | - Sony Sinha
- Department of Ophthalmology-Vitreo-Retina, Neuro-Ophthalmology and Oculoplasty, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| | - Kirandeep Kaur
- Department of Cataracts, Pediatric Ophthalmology and Strabismus, Gomabai Netralaya and Research Center, Neemuch 458441, Madhya Pradesh, India
| | - Ashish Kumar
- Department of Cornea and Refractive Surgery, Laxmi Netralaya, Jamal Road, Patna 800001, Bihar, India
| | - Ranjeet Kumar Sinha
- Department of Community Medicine, Patna Medical College, Patna 800004, Bihar, India
| |
Collapse
|
2
|
Brunel LG, Cai B, Hull SM, Han U, Wungcharoen T, Fernandes-Cunha GM, Seo YA, Johansson PK, Heilshorn SC, Myung D. In situ UNIversal Orthogonal Network (UNION) bioink deposition for direct delivery of corneal stromal stem cells to corneal wounds. Bioact Mater 2025; 48:414-430. [PMID: 40083774 PMCID: PMC11903395 DOI: 10.1016/j.bioactmat.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/16/2025] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
The scarcity of human donor corneal graft tissue worldwide available for corneal transplantation necessitates the development of alternative therapeutic strategies for treating patients with corneal blindness. Corneal stromal stem cells (CSSCs) have the potential to address this global shortage by allowing a single donor cornea to treat multiple patients. To directly deliver CSSCs to corneal defects within an engineered biomatrix, we developed a UNIversal Orthogonal Network (UNION) collagen bioink that crosslinks in situ with a bioorthogonal, covalent chemistry. This cell-gel therapy is optically transparent, stable against contraction forces exerted by CSSCs, and permissive to the efficient growth of corneal epithelial cells. Furthermore, CSSCs remain viable within the UNION collagen gel precursor solution under standard storage and transportation conditions. This approach promoted corneal transparency and re-epithelialization in a rabbit anterior lamellar keratoplasty model, indicating that the UNION collagen bioink serves effectively as an in situ-forming, suture-free therapy for delivering CSSCs to corneal wounds.
Collapse
Affiliation(s)
- Lucia G. Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Betty Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Sarah M. Hull
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Uiyoung Han
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Thitima Wungcharoen
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | - Youngyoon Amy Seo
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Patrik K. Johansson
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - David Myung
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
3
|
Matsumae H, Kataoka K, Yagi-Yaguchi Y, Higa K, Suzuki T, Kasamatsu H, Shimazaki J, Yamaguchi T. Correlation between preoperative aqueous cytokine levels and mid-term corneal endothelial cell loss following penetrating keratoplasty. Eye (Lond) 2025; 39:1286-1291. [PMID: 39833574 PMCID: PMC12043822 DOI: 10.1038/s41433-025-03610-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 11/29/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE To investigate the association between preoperative aqueous humour (AqH) cytokines and mid-term endothelial cell density (ECD) following penetrating keratoplasty (PKP). METHODS This study analysed a total of 87 eyes of which 54 underwent PKP and 33 eyes underwent cataract surgery. AqH samples were collected at the beginning of surgery. The levels of cytokines (interleukins [ILs]-1α, -1β, -4, -6, -8, -10, -17A, monocyte chemotactic protein [MCP]-1, interferon [IFN]-α, IFN-γ, E-selectin, P-selectin, and soluble intercellular adhesion molecule-1 [sICAM-1]) in the AqH were measured using a multiplex beads immunoassay. The subjects who underwent PKP were classified into two groups: Group 1: ECD < 1200 cells/mm2 at 36 months and graft failure during follow-up (38 eyes); Group 2 ECD ≥ 1200 cells/mm2 at 36 months (16 eyes). RESULTS The ECD reduction per year was significantly correlated with the preoperative levels of protein (r = 0.48; P < 0.001), IL-6 (r = 0.29; P = 0.048), IL-8 (r = 0.42; P = 0.003), IL-12p70 (r = 0.46; P = .007), MCP-1 (r = 0.28; P = 0.049), IFN -γ (r = 0.43; P = 0.008), and s-ICAM-1 (r = 0.30; P = 0.03). The preoperative levels of IL-8, and IFN-γ in AqH were significantly higher in eyes with ECD < 1200 cells/mm2 compared to those with ECD ≥ 1200 cells/mm2 at 3 years (P ≤ 0.02). CONCLUSION Preoperative AqH cytokine levels are associated with mid-term ECD reduction after PKP. Pathological alterations of the AqH microenvironment may affect mid-term corneal endothelial cell survival.
Collapse
Affiliation(s)
- Hiroshi Matsumae
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kaon Kataoka
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Yukari Yagi-Yaguchi
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Kazunari Higa
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Takanori Suzuki
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Hirotsugu Kasamatsu
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Jun Shimazaki
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Takefumi Yamaguchi
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan.
| |
Collapse
|
4
|
Manoochehrabadi T, Solouki A, Majidi J, Khosravimelal S, Lotfi E, Lin K, Daryabari SH, Gholipourmalekabadi M. Silk biomaterials for corneal tissue engineering: From research approaches to therapeutic potentials; A review. Int J Biol Macromol 2025; 305:141039. [PMID: 39956223 DOI: 10.1016/j.ijbiomac.2025.141039] [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: 01/02/2025] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
The corneal complications can result in opacity and eventual blindness. Furthermore, a shortage of available donors constrains the existing therapeutic options. Therefore, one of the most promising strategies involves the application of biomaterials, particularly silk. Silk has garnered significant attention among these biomaterials due to its natural origin and diverse features derived from different sources. One of the most critical factors of silk is its transparency, which is crucial for the cornea, and there are no concerns about infection. This material also possesses several advantages, including cost-effectiveness in production, biocompatibility in vivo and in vitro, biodegradation, and desirable mechanical characteristics. Modifications in the topographical structure, porosity, and crystallinity of silk enhance its properties and optimize its suitability for wound dressing, efficient drug delivery systems, and various cornea-related treatments. In each layer, silk was examined as a single biomaterial or blended with the others, so, this review aims to explore silk as a potential material for corneal regenerative medicine from a novel viewpoint. By considering a range of studies, a classification system has been developed that categorizes the utilization of silk in the various layers of the cornea and sub-categorizes the different modifications and applications of silk.
Collapse
Affiliation(s)
- Tahereh Manoochehrabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Solouki
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jila Majidi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sadjad Khosravimelal
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Ehsan Lotfi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kaili Lin
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China.
| | | | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran; NanoBiotechnology & Regenerative Medicine Innovation Group, Noavarn Salamat ZHINO (PHC), Tehran, Iran.
| |
Collapse
|
5
|
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
|
6
|
Peshkar-Kulkarni S, Chung DD, Aldave AJ. Antioxidant MitoQ increases viability of human corneal endothelial cells with congenital hereditary endothelial dystrophy-associated SLC4A11 mutations. Ophthalmic Genet 2025; 46:166-173. [PMID: 39834031 PMCID: PMC12003074 DOI: 10.1080/13816810.2025.2450455] [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: 02/07/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE To assess the impact of MitoQ, a mitochondria-targeted antioxidant, on viability of human corneal endothelial cell (hCEnC) lines expressing SLC4A11 mutations associated with congenital hereditary endothelial dystrophy (CHED) and Fuchs endothelial corneal dystrophy type 4 (FECD4). METHODS SLC4A11 wildtype (SLC4A11WT) and mutant (SLC4A11MU) hCEnC lines were created to express either SLC4A11 variant 2 (V2) or variant 3 (V3) by stable transduction of SLC4A11-/- hCEnC-21T with lentiviruses containing either SLC4A11WT or one of the following mutations: V2 (V3) mutants c.374 G>A (c.326 G>A) (CHED), c.1813C>T (c.1765C>T) (CHED), c.2263C>T (c.2215C>T) (CHED), or c.2224 G>A (c.2176 G>A) (FECD4). A SLC4A11-/- empty hCEnC line was created by stable transduction of SLC4A11-/- hCEnC-21T with an empty lentiviral plasmid. Cell viability was measured by exposing MitoQ treated and untreated cells to oxidative stress agent tert-butyl hydroperoxide (tBH) followed by performing XTT assays and spectrophotometry. RESULTS SLC4A11-/- empty, SLC4A11 V2WT, and SLC4A11 V3WT hCEnC exposed to ≤0.01 μM MitoQ retained over 90% of the viability of untreated SLC4A11-/- empty hCEnC. When treated with MitoQ, SLC4A11-/- empty was able to demonstrate partial restoration of cell viability. All CHED-associated mutant hCEnC lines treated with 0.01 μM MitoQ demonstrated increased viability compared to untreated following exposure to tBH. The FECD4-associated mutant hCEnC line treated with 0.01 μM MitoQ showed no significant increase in cell viability compared to untreated following exposure to tBH. CONCLUSIONS Media supplementation with antioxidant MitoQ has beneficial effects on cell viability in hCEnC harboring CHED-associated SLC4A11 mutations following exposure to tBH-induced oxidative stress.
Collapse
Affiliation(s)
| | - Doug D Chung
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, USA
| | - Anthony J Aldave
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, USA
| |
Collapse
|
7
|
Li X, Liu A, Zhou Y, Qi H, Wang J, Chen M, Sun T, Wu J, Huang Y, Wang L. Proliferator-Activated Receptor Alpha Inhibits Abnormal Extracellular Matrix Accumulation and Maintains Energy Metabolism in Late-Onset Fuchs Endothelial Corneal Dystrophy. Invest Ophthalmol Vis Sci 2025; 66:36. [PMID: 40232711 PMCID: PMC12007668 DOI: 10.1167/iovs.66.4.36] [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: 11/18/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Purpose Fuchs endothelial corneal dystrophy (FECD) is the most common corneal endothelial dystrophy and guttae are crucial in causing progressive loss of corneal endothelium. This study aimed to find a way to inhibit the formation of guttae in FECD. Methods and Results Mitochondria fatty acid β-oxidation (FAO) and tricarboxylic acid (TCA) cycle processes were negatively enriched in the FECD group according to gene set enrichment analysis in GSE171830. In vivo UV-A-induced late-onset FECD mouse model were established. After irradiation, aged proliferator-activated receptor alpha (PPARα-/-) mice manifested greater corneal opacity, cornea edema, and varied corneal endothelial cell morphology compared with wild-type mice. The total metabolites in cornea of aged PPARα-/- mice and wild-type mice were detected by mass spectrometry. Metabolites of the FAO pathway were decreased in corneas of PPARα-/- mice, coincident with enzymes of FAO decreased in GSE171830. The score for FAO energy metabolism was negatively related to that of the TGF-β pathway according to gene set variation analysis. The express of alpha smooth muscle actin (αSMA) and Col1a were increased in aged PPARα-/- mice and small interfering PPARα B4G12 cell lines. After irradiation, activation or overexpression of PPARα demonstrated reduced corneal endothelial damage and reversal of Descemet membrane thickening, along with downregulation of fibrosis-related genes such as αSMA and collagen type I alpha 1 (Col1a). In vitro experiments revealed that fenofibrate could reverse fibrosis and damage of cell-to-cell connections induced by TGF-β. Additionally, fenofibrate was found to alleviate mitochondrial damage in B4G12 and increase oxygen consumption rates after TGF-β treatment. Conclusions Overall, we suggested that the overexpression or activation of PPARα can inhibit FAO energy dysfunction of corneal endothelium and the abnormal extracellular matrix formation in Descemet's membrane, which is the primary pathology of FECD. Thus, PPARα may be a potential target for attenuating the progression of FECD.
Collapse
Affiliation(s)
- Xiaoqi Li
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Chinese PLA Navy No. 971 Hospital, Qingdao, China
| | - Anqi Liu
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yannan Zhou
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Haolan Qi
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Junyi Wang
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Mingxiong Chen
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Tunan Sun
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Wu
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yifei Huang
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Liqiang Wang
- State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Raut AK, Mohapatra S, SiddiquI G, Rajak SK, Sonar R, Basu S, Joshi V, Singh V. The Human Cornea: Unraveling Its Structural, Chemical, and Biochemical Complexities. Chem Biodivers 2025; 22:e202402224. [PMID: 39559954 DOI: 10.1002/cbdv.202402224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/20/2024]
Abstract
The cornea, the transparent part of the anterior eye, is vital for light refraction and vision. This review examines the intricate chemical and biochemical interactions essential for maintaining corneal transparency and highlights significant advancements in corneal biology. The cornea comprises five layers: the epithelium, Bowman's layer, stroma, Descemet's membrane, and endothelium, each contributing uniquely to its structure and function. The epithelium, maintained by limbal stem cells, serves as a barrier and interacts with the tear film to maintain ocular surface health. The stroma, abundant in organized collagen fibrils and regulated by proteoglycans, is crucial for corneal clarity and biomechanical integrity, whereas the endothelium regulates corneal hydration and nutrition. Recent imaging advances have improved visualization of these molecular structures, enhancing our understanding of collagen organization and cross-linking. Proteoglycans such as decorin and lumican regulate collagen spacing and hydration, directly influencing corneal clarity. Biochemical processes within the cornea involve signaling molecules, growth factors, and cytokines, which are essential for wound healing, inflammation, and injury response. Despite progress, questions remain regarding corneal wound healing mechanisms, the impact of oxidative stress, and the roles of microRNAs. This review synthesizes recent discoveries to advance our understanding of corneal physiology and biochemical functions.
Collapse
Affiliation(s)
- Arun Kumar Raut
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| | - Sonali Mohapatra
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| | - Gufran SiddiquI
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| | - Suraj Kumar Rajak
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| | - Rohini Sonar
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| | - Sayan Basu
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| | - Vineet Joshi
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| | - Vivek Singh
- LV Prasad Eye Institute, Kallam Anji Reddy Campus, Centre for Ocular Regeneration, Brien Holden Eye Research Centre, Champalimaud Translational Centre for Eye Research, Hyderabad, Telangana, India
| |
Collapse
|
9
|
Wu Y, Benson MA, Sun SX. Fluid and solute transport by cells and a model of systemic circulation. PLoS Comput Biol 2025; 21:e1012935. [PMID: 40258085 PMCID: PMC12040233 DOI: 10.1371/journal.pcbi.1012935] [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: 05/16/2024] [Revised: 04/29/2025] [Accepted: 03/06/2025] [Indexed: 04/23/2025] Open
Abstract
Active fluid circulation and solute transport are essential functions of living organisms, enabling the efficient delivery of oxygen and nutrients to various physiological compartments. Since fluid circulation occurs in a network, the systemic flux and pressure are not simple outcomes of individual components. Rather, they are emergent properties of network elements and network topology. Moreover, consistent pressure and osmolarity gradients are maintained across compartments such as the kidney, interstitium, and blood vessels. The mechanisms by which these gradients and network properties are established and maintained are unanswered questions in systems physiology. Previous studies have shown that epithelial cells are fluid pumps and can actively generate pressure and osmolarity gradients. The polarization and activity of solute transporters in epithelial cells, which drive fluid flux, are influenced by pressure and osmolarity gradients. Therefore, there is an unexplored coupling between pressure and osmolarity in the circulatory network. In this work, we develop a mathematical framework that integrates the influence of pressure and osmolarity on solute transport. We use this model to explore both cellular fluid transport and systemic circulation. Using a simple network featuring the kidney-vascular interface, we show that our model naturally generates pressure and osmolarity gradients across the kidney, vessels and renal interstitium. While the current model uses this interface as an example, the findings can be generalized to other physiological compartments. This model demonstrates how systemic transport properties can depend on cellular properties and, conversely, how cell states are influenced by systemic properties. When epithelial and endothelial pumps are considered together, we predict how pressures at various points in the network depend on the overall osmolarity of the system. The model can be improved by including physiological geometries and expanding solute species, and highlights the interplay of fluid properties with cell function in living organisms.
Collapse
Affiliation(s)
- Yufei Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Morgan A. Benson
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sean X. Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
10
|
Wu KY, Kearn N, Truong D, Choulakian MY, Tran SD. Advances in Regenerative Medicine, Cell Therapy, and 3D Bioprinting for Corneal, Oculoplastic, and Orbital Surgery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40131704 DOI: 10.1007/5584_2025_855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Advances in regenerative medicine, cell therapy, and 3D bioprinting are reshaping the landscape of ocular surgery, offering innovative approaches to address complex conditions affecting the cornea, ocular adnexal structures, and the orbit. These technologies hold the potential to enhance treatment precision, improve functional outcomes, and address limitations in traditional surgical and therapeutic interventions.The cornea, as the eye's primary refractive and protective barrier, is particularly well-suited for regenerative approaches due to its avascular and immune-privileged nature. Cell-based therapies, including limbal stem cell transplantation as well as stromal keratocyte and corneal endothelial cell regeneration, are being investigated for their potential to restore corneal clarity and function in conditions such as limbal stem cell deficiency, keratoconus, and endothelial dysfunction. Simultaneously, 3D bioprinting technologies are enabling the development of biomimetic corneal constructs, potentially addressing the global shortage of donor tissues and facilitating personalized surgical solutions.In oculoplastic and orbital surgery, regenerative strategies and cell therapies are emerging as possible alternatives to conventional approaches for conditions such as eyelid defects, meibomian gland dysfunction, and Graves' orbitopathy. Stem cell-based therapies and bioengineered scaffolds are showing potential in restoring lacrimal glands' function as well as reconstructing complex ocular adnexal and orbital structures. Moreover, 3D-printed orbital implants and scaffolds offer innovative solutions for repairing traumatic, post-tumor resection, and congenital defects, with the potential for improved biocompatibility and precision.Molecular and gene-based therapies, including exosome delivery systems, nanoparticle-based interventions, and gene-editing techniques, are expanding the therapeutic arsenal for ophthalmic disorders. These approaches aim to enhance the efficacy of regenerative treatments by addressing underlying pathophysiological mechanisms of diseases. This chapter provides an overview of these advancements and the challenges of translating laboratory discoveries into effective therapies in clinical practice.
Collapse
Affiliation(s)
- Kevin Y Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Natalie Kearn
- Department of Medicine, School of Medicine, Queen's University, Kingston, ON, Canada
| | - Doanh Truong
- College of Arts & Science, Case Western Reserve University, Cleveland, OH, USA
| | - Mazen Y Choulakian
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
11
|
Abbasi M, Aghamollaei H, Vaez A, Amani AM, Kamyab H, Chelliapan S, Jamalpour S, Zambrano-Dávila R. Bringing ophthalmology into the scientific world: Novel nanoparticle-based strategies for ocular drug delivery. Ocul Surf 2025; 37:140-172. [PMID: 40147816 DOI: 10.1016/j.jtos.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
The distinctive benefits and drawbacks of various drug delivery strategies to supply corneal tissue improvement for sense organs have been the attention of studies worldwide in recent decades. Static and dynamic barriers of ocular tissue prevent foreign chemicals from entering and inhibit the active absorption of therapeutic medicines. The distribution of different medications to ocular tissue is one of the most appealing and demanding tasks for investigators in pharmacology, biomaterials, and ophthalmology, and it is critical for cornea wound healing due to the controlled release rate and increased drug bioavailability. It should be mentioned that the transport of various types of medications into the different sections of the eye, particularly the cornea, is exceedingly challenging because of its distinctive structure and various barriers throughout the eye. Nanoparticles are being studied to improve medicine delivery strategies for ocular disease. Repetitive corneal drug delivery using biodegradable nanocarriers allows a medicine to remain in different parts of the cornea for extended periods of time and thus improve administration route effectiveness. In this review, we discussed eye anatomy, ocular delivery barriers, as well as the emphasis on the biodegradable nanomaterials ranging from organic nanostructures, such as nanomicelles, polymers, liposomes, niosomes, nanowafers, nanoemulsions, nanosuspensions, nanocrystals, cubosomes, olaminosomes, hybridized NPs, dendrimers, bilosomes, solid lipid NPs, nanostructured lipid carriers, and nanofiber to organic nanomaterials like silver, gold, and mesoporous silica nanoparticles. In addition, we describe the nanotechnology-based ophthalmic medications that are presently on the market or in clinical studies. Finally, drawing on current trends and therapeutic approaches, we discuss the challenges that innovative optical drug delivery systems confront and propose future research routes. We hope that this review will serve as a source of motivation and inspiration for developing innovative ophthalmic formulations.
Collapse
Affiliation(s)
- Milad Abbasi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, India; The KU-KIST Graduate School of Energy and Environment, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Republic of Korea; Universidad UTE, Quito, 170527, Ecuador.
| | - Shreeshivadasan Chelliapan
- Department of Smart Engineering and Advanced Technology, Faculty of Artificial Intelligence, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Sajad Jamalpour
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Renato Zambrano-Dávila
- Universidad UTE, Centro de Investigación en Salud Públicay Epidemiología Clínica (CISPEC), Quito, 170527, Ecuador
| |
Collapse
|
12
|
Jammes M, Tabasi A, Bach T, Ritter T. Healing the cornea: Exploring the therapeutic solutions offered by MSCs and MSC-derived EVs. Prog Retin Eye Res 2025; 105:101325. [PMID: 39709150 DOI: 10.1016/j.preteyeres.2024.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Affecting a large proportion of the population worldwide, corneal disorders constitute a concerning health hazard associated to compromised eyesight or blindness for most severe cases. In the last decades, mesenchymal stem/stromal cells (MSCs) demonstrated promising abilities in improving symptoms associated to corneal diseases or alleviating these affections, especially through their anti-inflammatory, immunomodulatory and pro-regenerative properties. More recently, MSC therapeutic potential was shown to be mediated by the molecules they release, and particularly by their extracellular vesicles (EVs; MSC-EVs). Consequently, using MSC-EVs emerged as a pioneering strategy to mitigate the risks related to cell therapy while providing MSC therapeutic benefits. Despite the promises given by MSC- and MSC-EV-based approaches, many improvements are considered to optimize the therapeutic significance of these therapies. This review aspires to provide a comprehensive and detailed overview of current knowledge on corneal therapies involving MSCs and MSC-EVs, the strategies currently under evaluation, and the gaps remaining to be addressed for clinical implementation. From encapsulating MSCs or their EVs into biomaterials to enhance the ocular retention time to loading MSC-EVs with therapeutic drugs, a wide range of ground-breaking strategies are currently contemplated to lead to the safest and most effective treatments. Promising research initiatives also include diverse gene therapies and the targeting of specific cell types through the modification of the EV surface, paving the way for future therapeutic innovations. As one of the most important challenges, MSC-EV large-scale production strategies are extensively investigated and offer a wide array of possibilities to meet the needs of clinical applications.
Collapse
Affiliation(s)
- Manon Jammes
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Abbas Tabasi
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Trung Bach
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland; CURAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
13
|
Mulpuri L, Ouano DP, Riaz KM, Warner EJ, Stone DU, Cheung AY, Gomez A, Rangu N, Sabater AL, Tonk RS. Intracameral Enoxaparin for Descemet Membrane Endothelial Keratoplasty: A Pilot Safety Study. Cornea 2025; 44:342-349. [PMID: 39902781 DOI: 10.1097/ico.0000000000003662] [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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 02/06/2025]
Abstract
PURPOSE The purpose of this study was to investigate the safety and outcomes of Descemet membrane endothelial keratoplasty (DMEK) performed with intracameral enoxaparin. METHODS Two arms were used: a clinical multicenter retrospective cohort arm (CA) and an ex vivo basic science arm (BSA). In CA, DMEKs were performed by 6 experienced corneal surgeons at multiple sites. Intracameral enoxaparin (40 mg/500 mL) was added to the irrigation fluid for all cases. Primary outcomes were measured at 6 and 12 months. In BSA, mated graft pairs were randomized to control or enoxaparin exposure (0.8 mg/mL × 1 hour) and assessed for endothelial cell death count at 0-, 1-, and 24-hour intervals and cellular stress by ELISA Annexin V protein quantification. RESULTS In the cohort arm, the mean age of 159 eyes of 134 patients was 69.3 years with Fuchs dystrophy as the primary diagnosis. Mean BCVA improved from 0.42 ± 0.3 logMAR preoperatively to 0.13 ± 0.1 logMAR postoperatively at 6 months (P < 0.001) and to 0.1 ± 0.1 logMAR at 12 months (P < 0.001). At 6 months, 58.4% of patients achieved a final BCVA of 20/25 or better and 91% improved to 20/40 or better. Rebubble rate was 13% (n = 21), with 6 of these 21 eyes requiring more than 1 rebubble. One total graft detachment was noted with no reports of intraoperative or postoperative hemorrhage. PGF occurred in 0 of 159 eyes. In BSA, enoxaparin had no significant effect on endothelial cell death count or cellular apoptosis compared with control. CONCLUSIONS Enoxaparin can safely be used in DMEK surgery without apparent increased risk of intraoperative hemorrhage, graft detachment/failure, or endothelial cell toxicity.
Collapse
Affiliation(s)
- Lakshman Mulpuri
- Department of Ophthalmology, Cook County Health System, Chicago, IL
| | | | - Kamran M Riaz
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma, Oklahoma City, OK
| | - Evan J Warner
- Department of Ophthalmology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | | | - Angela Gomez
- Department of Ophthalmology, Cook County Health System, Chicago, IL
| | - Neal Rangu
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma, Oklahoma City, OK
| | | | - Rahul S Tonk
- Department of Ophthalmology, Cook County Health System, Chicago, IL
| |
Collapse
|
14
|
Sicks B, Hessling M, Stucke-Straub K, Kupferschmid S, Lotfi R. Disinfection of Human and Porcine Corneal Endothelial Cells by Far-UVC Irradiation. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:416. [PMID: 40142227 PMCID: PMC11943857 DOI: 10.3390/medicina61030416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: The cornea protects the eye from external influences and contributes to its refractive power. Corneas belong to the most frequently transplanted tissues, providing a last resort for preserving the patient's vision. There is a high demand for donor corneas worldwide, but almost 4% of these transplants are not eligible due to microbial contamination. The objective of this study is to ascertain the suitability of 222 nm Far-UVC irradiation for the decontamination of corneas without damaging corneal endothelial cells. Materials and Methods: To assess the destructive effect of irradiation and, thus, identify the applicable dose needed to decontaminate the cornea without interfering with its integrity, 141 porcine corneas were irradiated with 0, 60 or 150 mJ/cm2 at 222 nm. In the second step, a series of 13 human corneas were subjected to half-sided irradiation using 15 or 60 mJ/cm2 at 222 nm. After five days of in vitro culturing, the endothelial cell density of the non-irradiated area of each human cornea was compared to the irradiated area. Results: Irradiation with up to 60 mJ/cm2 had no detectably significant effect on the cell integrity of human corneas (p = 0.764), with only a minimal reduction in cell density of 3.7% observed. These findings were partially corroborated by tests on porcine corneas, wherein the variability between test groups was consistent, even at increased irradiation doses of up to 150 mJ/cm2, and no notable effects on the irradiated porcine endothelium were monitored. The efficacy of the antimicrobial treatment was evident in the disinfection tests conducted on corneas. Conclusions: These initial irradiation experiments demonstrated that 222 nm Far-UVC radiation has the potential to decontaminate the cornea without compromising sensitive endothelial cell viability.
Collapse
Affiliation(s)
- Ben Sicks
- Institute of Medical Engineering and Mechatronics, Ulm University of Applied Sciences, Albert-Einstein-Allee 55, 89081 Ulm, Germany; (M.H.); (K.S.-S.)
| | - Martin Hessling
- Institute of Medical Engineering and Mechatronics, Ulm University of Applied Sciences, Albert-Einstein-Allee 55, 89081 Ulm, Germany; (M.H.); (K.S.-S.)
| | - Kathrin Stucke-Straub
- Institute of Medical Engineering and Mechatronics, Ulm University of Applied Sciences, Albert-Einstein-Allee 55, 89081 Ulm, Germany; (M.H.); (K.S.-S.)
| | - Sebastian Kupferschmid
- Clinic of Ophthalmology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081 Ulm, Germany;
| | - Ramin Lotfi
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg-Hessen, Institute for Transfusion Medicine, University Hospital Ulm, Helmholtzstraße 10, 89081 Ulm, Germany;
| |
Collapse
|
15
|
Prašnikar E, Stunf Pukl S. How "Omics" Studies Contribute to a Better Understanding of Fuchs' Endothelial Corneal Dystrophy. Curr Issues Mol Biol 2025; 47:135. [PMID: 40136389 PMCID: PMC11941302 DOI: 10.3390/cimb47030135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Fuchs' endothelial corneal dystrophy (FECD) is a progressive eye disease characterized by accelerated loss of endothelial cells and the development of focal excrescence (guttae) on Descemet's membrane, resulting in cornea opacity and vision deterioration. The development of FECD is assumed to be due to the interplay between genetic and environmental factor risks, causing abnormal extracellular-matrix organization, increased oxidative stress, apoptosis and unfolded protein response. However, the molecular knowledge of FECD is limited. The development of genome-wide platforms and bioinformatics approaches has enabled us to identify numerous genetic loci that are associated with FECD. In this review, we gathered genome-wide studies (n = 31) and sorted them according to genomics (n = 9), epigenomics (n = 3), transcriptomics (n = 15), proteomics (n = 3) and metabolomics (n = 1) levels to characterize progress in understanding FECD. We also extracted validated differentially expressed/spliced genes and proteins identified through comparisons of FECD case and control groups. In addition, highlighted loci from each omics layer were combined according to a comparison with similar study groups from original studies for downstream gene-set enrichment analysis, which provided the most significant biological pathways related to extracellular-matrix organization. In the future, multiomics study approaches are needed to increase the sample size and statistical power to identify strong candidate genes for functional studies on animal models and cell lines for better understanding FECD.
Collapse
Affiliation(s)
- Erika Prašnikar
- Eye Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
| | - Spela Stunf Pukl
- Eye Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
16
|
Chow BJ, Liu C, Yu M, Xin Yu Lee I, Mehta JS, Wu QYS, Wong Kay Ting R, Lin K, Liu YC. The Application of Terahertz Technology in Corneas and Corneal Diseases: A Systematic Review. Bioengineering (Basel) 2025; 12:45. [PMID: 39851319 PMCID: PMC11762122 DOI: 10.3390/bioengineering12010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Terahertz (THz) waves reside in the electromagnetic spectrum between the microwave and infrared bands. In recent decades, THz technology has demonstrated its potential for biomedical applications. With the highly unique characteristics of THz waves, such as the high sensitivity to water and optimal spatial resolution coupled with the characteristics of the human cornea, such as its high water content, THz technology has been explored as a potential modality to assess corneas and corneal diseases. This systematic review provides an overview of the characteristics of THz waves, the safety profile of THz technology in the field of ophthalmology, and its clinical applications, including the objective evaluation of the corneal hydration, tear film, dry eye disease, corneal endothelium, corneal elasticity, and scarring. The paper also presents our viewpoint on the present challenges and future directions of THz technology prior to its broader integration into clinical practice.
Collapse
Affiliation(s)
- Bing Jie Chow
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK
| | - Chang Liu
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Mingyi Yu
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Isabelle Xin Yu Lee
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Jodhbir S. Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore 168751, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Qing Yang Steve Wu
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, Singapore 138634, Singapore
| | - Regina Wong Kay Ting
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Ke Lin
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, Singapore 138634, Singapore
| | - Yu-Chi Liu
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore 168751, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
17
|
Ho WT, Chang JS, Lei CJ, Chen TC, Wang JK, Chang SW, Yang MH, Jou TS, Wang IJ. ROCK Inhibitor Enhances Resilience Against Metabolic Stress Through Increasing Bioenergetic Capacity in Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2025; 66:51. [PMID: 39847368 PMCID: PMC11759620 DOI: 10.1167/iovs.66.1.51] [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: 07/09/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose To investigate the effect of Rho-associated protein kinase (ROCK) inhibitor Y27632 on bioenergetic capacity and resilience of corneal endothelial cells (CECs) under metabolic stress. Methods Bovine CECs (BCECs) were treated with Y27632 and subjected to bioenergetic profiling using the Seahorse XFp Analyzer. The effects on adenosine triphosphate (ATP) production through oxidative phosphorylation and glycolysis were measured. BCECs were also challenged with monensin to induce metabolic stress. Cell viability, apoptosis, intracellular sodium levels, and hexokinase localization were assessed using calcein AM assay, flow cytometry, fluorescence imaging, and immunostaining, respectively. Results Y27632 increased maximal ATP production rates via both oxidative phosphorylation and glycolysis, thereby expanding the overall bioenergetic capacity in BCECs. Under monensin-induced metabolic stress, ROCK inhibitor pretreatment significantly enhanced glycolytic ATP production and reduced apoptosis compared with untreated cells. Y27632 also facilitated sodium export by increasing Na/K-ATPase activity, as evidenced by lower intracellular sodium levels. Additionally, Y27632 promoted the translocation of hexokinase 2 to mitochondria under stress conditions, thereby enhancing glycolytic capacity. The effect of Y27632 on cell viability and sodium export was abrogated when cells were forced to rely on oxidative phosphorylation in galactose media, indicating that the protective effects of Y27632 are dependent on glycolytic ATP production under monensin stress. Conclusions ROCK inhibitor Y27632 enhances the bioenergetic capacity of BCECs, allowing the cells to better withstand metabolic stress by rapidly generating ATP to meet increased energy demands, maintaining ion homeostasis and reducing apoptosis.
Collapse
Affiliation(s)
- Wei-Ting Ho
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jung-Shen Chang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Jen Lei
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsan-Chi Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Jia-Kang Wang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Electrical Engineering, Yuan Ze University, Taoyuan, Taiwan
| | - Shu-Wen Chang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzuu-Shuh Jou
- College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taiwan
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
18
|
Sood S, Tiwari A, Sangwan J, Vohra M, Sinha NR, Tripathi R, Sangwan VS, Mohan RR. Role of epigenetics in corneal health and disease. Prog Retin Eye Res 2025; 104:101318. [PMID: 39547455 PMCID: PMC11710990 DOI: 10.1016/j.preteyeres.2024.101318] [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: 09/03/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
Epigenetics plays a vital role in corneal health and diseases. Epigenetic changes regulate the expression of genes by altering the accessibility of chromatin via histone modifications, DNA methylation and miRNAs without altering DNA sequence. Ocular trauma and infections are common causes of corneal damage, vision impairment, and mono/bilateral blindness worldwide. Mounting literature shows that epigenetic modifications can modulate corneal clarity, function, and pathogenesis including inflammation, wound healing, fibrosis, and neovascularization. Additionally, epigenetic modifications can be targeted to reverse corneal pathologies and develop interventional therapies. However, current understanding on how epigenetic modifications lead to corneal abnormalities and diseases is limited. This review provides in-depth knowledge and mechanistic understanding of epigenetics alterations in corneal pathogenesis, and information on potential epigenetic targets for treatment of corneal diseases.
Collapse
Affiliation(s)
- Swati Sood
- Departments of Veterinary Medicine & Surgery, College of Veterinary Medicine University of Missouri, Columbia, MO, USA
| | - Anil Tiwari
- Departments of Veterinary Medicine & Surgery, College of Veterinary Medicine University of Missouri, Columbia, MO, USA; Eicher-Shroff Centre for Stem Cells Research (ESC-SCR), Dr. Shroff Charity Eye Hospital, Delhi, India
| | - Jyoti Sangwan
- Eicher-Shroff Centre for Stem Cells Research (ESC-SCR), Dr. Shroff Charity Eye Hospital, Delhi, India
| | - Mehak Vohra
- Eicher-Shroff Centre for Stem Cells Research (ESC-SCR), Dr. Shroff Charity Eye Hospital, Delhi, India
| | - Nishant R Sinha
- Departments of Veterinary Medicine & Surgery, College of Veterinary Medicine University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA; Department of Ophthalmology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Ratnakar Tripathi
- Departments of Veterinary Medicine & Surgery, College of Veterinary Medicine University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Virender S Sangwan
- Eicher-Shroff Centre for Stem Cells Research (ESC-SCR), Dr. Shroff Charity Eye Hospital, Delhi, India
| | - Rajiv R Mohan
- Departments of Veterinary Medicine & Surgery, College of Veterinary Medicine University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA; Department of Ophthalmology, School of Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
19
|
Colabelli Gisoldi RAM, Pocobelli G, Rodella U, Giurgola L, Gatto C, Lodato G, Buffon G, Nucci C, D’Amato Tóthová J, Pocobelli A. Retrospective Clinical Outcomes of Keratoplasty Using Human Donor Corneas Preserved in Eusol-C Hypothermic Storage Medium. J Clin Med 2024; 13:7606. [PMID: 39768528 PMCID: PMC11677598 DOI: 10.3390/jcm13247606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/26/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Objective: To evaluate the clinical outcomes of cornea transplantation (penetrating keratoplasty, Descemet membrane endothelial keratoplasty, Descemet stripping automated endothelial keratoplasty, and deep anterior lamellar keratoplasty) using donor corneas stored in Eusol-C hypothermic storage medium compared to corneas stored in organ-culture. Methods: The clinical outcomes of 92 patients who underwent corneal transplantation with human donor corneas stored in Eusol-C medium at 2-8 °C were retrospectively evaluated. The control group consisted of 169 patients who received corneas organ-cultured at 31 °C. Donor age, sex, death-to-preservation time, and storage time were recorded. Endothelial cell (EC) density (ECD), EC mortality, and EC morphology scores were evaluated during storage in both groups. Complication rates, visual outcomes, and corneal transparency were monitored for up to six months. Results: The mean storage in Eusol-C time was 7.7 ± 2.5 days, while organ-culture time was 14.2 ± 4.0 days. In the Eusol-C group, ECD was 2398 ± 354 cells/mm2, with an average EC morphology score of 3.4 ± 0.7/4. Approximately 28% of the corneas in the Eusol-C group had no EC mortality. In the organ-culture group, ECD was 2256 ± 328 cells/mm2, with an average EC morphology score of 3.5 ± 0.5/4, and 42% were devoid of EC mortality. No complications, such as re-bubbling, were observed in both groups during surgery. Transparent corneas were achieved in 81.3% of the Eusol-C group the day after surgery. Mean corrected distance visual acuity (CDVA) at 3 and 6 months was 4.5 ± 4.0/10 and 5.4 ± 3.7/10 for the Eusol-C group and 5.0 ± 2.9/10 and 5.7 ± 2.8/10 for the organ-culture group, with no statistical differences observed between the groups. No graft failure was observed up to three months. Graft rejection occurred in the Eusol-C group and in the organ-culture group in, respectively, one and two cases at the six-month follow-up. Conclusions: Comparable surgical outcomes were achieved with donor corneas stored in both hypothermic Eusol-C and organ-culture media.
Collapse
Affiliation(s)
| | - Giulio Pocobelli
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Umberto Rodella
- Research and Development, AL.CHI.MI.A. S.R.L., 35020 Ponte San Nicolò, Italy
| | - Laura Giurgola
- Research and Development, AL.CHI.MI.A. S.R.L., 35020 Ponte San Nicolò, Italy
| | - Claudio Gatto
- Research and Development, AL.CHI.MI.A. S.R.L., 35020 Ponte San Nicolò, Italy
| | - Gemma Lodato
- Ophthalmology Unit–Eye Bank of Rome, San Giovanni Addolorata Hospital, 00184 Rome, Italy
| | - Giacinta Buffon
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carlo Nucci
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Augusto Pocobelli
- Ophthalmology Unit–Eye Bank of Rome, San Giovanni Addolorata Hospital, 00184 Rome, Italy
| |
Collapse
|
20
|
Ng XY, Peh G, Morales-Wong F, Gabriel R, Soong PL, Lin KH, Mehta JS. Towards Clinical Application: Calcium Waves for In Vitro Qualitative Assessment of Propagated Primary Human Corneal Endothelial Cells. Cells 2024; 13:2012. [PMID: 39682760 PMCID: PMC11640329 DOI: 10.3390/cells13232012] [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: 10/25/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Corneal endothelium cells (CECs) regulate corneal hydration between the leaky barrier of the corneal endothelium and the ionic pumps on the surface of CECs. As CECs do not regenerate, loss of CECs leads to poor vision and corneal blindness. Corneal transplant is the only treatment option; however, there is a severe shortage of donor corneas globally. Cell therapy using propagated primary human CECs is an alternative approach to corneal transplantations, and proof of functionality is crucial for validating such CECs. Expression markers like Na-K-ATPase and ZO-1 are typical but not specific to CECs. Assessing the barrier function of the expanded CECs via electrical resistance (i.e., TEER and Ussing's chamber) involves difficult techniques and is thus impractical for clinical application. Calcium has been demonstrated to affect the paracellular permeability of the corneal endothelium. Its absence alters morphology and disrupts apical junctions in bovine CECs, underscoring its importance. Calcium signaling patterns such as calcium waves affect the rate of wound healing in bovine CECs. Therefore, observing calcium waves in expanded CECs could provide valuable insights into their health and functional integrity. Mechanical or chemical stimulations, combined with Ca2+-sensitive fluorescent dyes and time-lapse imaging, can be used to visualize these waves, which could potentially be used to qualify expanded CECs.
Collapse
Affiliation(s)
- Xiao Yu Ng
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
| | - Gary Peh
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- Eye-Academic Clinical Program (ACP), Duke-National University of Singapore (NUS) Graduate Medical School, Singapore 169857, Singapore
| | - Fernando Morales-Wong
- Department of Ophthalmology, University Hospital and Faculty of Medicine, Autonomous University of Nuevo León (UANL), Monterrey 64460, Mexico;
| | - Rami Gabriel
- Department of Ophthalmology, Duke University Health Center, Durham, NC 27705, USA;
| | | | - Kun-Han Lin
- Ternion Biosciences, Singapore 574329, Singapore; (P.L.S.)
| | - Jodhbir S. Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- Eye-Academic Clinical Program (ACP), Duke-National University of Singapore (NUS) Graduate Medical School, Singapore 169857, Singapore
- Corneal and External Diseases Department, Singapore National Eye Centre, Singapore 168751, Singapore
| |
Collapse
|
21
|
Ryu Y, Seo JH, Kim HS, Nam YJ, Bo Noh K, Oh SH, Hwang JS, Shin YJ. COL8A2 activation enhances function of corneal endothelial cells through HIPPO signaling/mitochondria pathway. Matrix Biol 2024; 134:119-131. [PMID: 39395654 DOI: 10.1016/j.matbio.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
Corneal endothelial cells (CECs) are essential for maintaining corneal transparency and hydration through their barrier and pump functions. The COL8A2 gene encodes a component of the extracellular matrix of the cornea, which is crucial for the normal functioning of these cells. Mutations in COL8A2 are linked to corneal dystrophies, emphasizing the gene's importance in corneal health. The purpose of this research is to explore the effects of COL8A2 activation within CECs, to understand its contribution to cellular behavior and health. COL8A2 CRISPR/dCas9 activation system (aCOL8A2) was used to activate the COL8A2. In rats, wound healing and mitochondrial function were assessed after COL8A2 activation. As a result, aCOL8A2 promoted wound healing of rat corneal endothelium by increasing mitochondrial membrane potential. In cultured human CECs, proteomic analysis was performed to screen and identify the differential protein profiles between control and aCOL8A2 cells. Western blot was used to validate the differential proteins from both cells. Mitochondrial function and intracellular distribution were assessed by measuring ATP production and mitochondrial membrane potential. In cultured human CECs, aCOL8A2 increased COL8A2 and phospho-YAP levels. Transendothelial electrical resistance (TEER) was increased and actin cytoskeleton was attenuated by aCOL8A2. Gene ontology analysis revealed that the proteins were mainly involved in the regulation of folate biosynthesis, ECM-receptor interaction, cell differentiation, NADP activity and cytoskeleton. ATP production was increased, mitochondrial membrane potential was polarized and mitochondrial distribution was widespread in the aCOL8A2 group. In conclusion, aCOL8A2 induces a regulatory cascade affecting mitochondrial positioning and efficiency, mediated by alterations in the cytoskeletal architecture and the YAP signaling pathway. This sequence of events serves to bolster the functional capacities of corneal endothelial cells, including their pump and barrier functions, essential for corneal health and transparency.
Collapse
Affiliation(s)
- Yunkyoung Ryu
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Hak Su Kim
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Youn Joo Nam
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Kyung Bo Noh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Sun-Hee Oh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea; Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
22
|
McMullen AJ, Dantsoho ZA, Chamness S, Brunelle J, Martiz J, Khalifa YM, Buckley MR. Investigating the Tolerance of Corneal Endothelial Cells to Surgical Fluid Pressure Using Intact Porcine Eyes. Transl Vis Sci Technol 2024; 13:27. [PMID: 39570617 PMCID: PMC11585060 DOI: 10.1167/tvst.13.11.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/25/2024] [Indexed: 11/22/2024] Open
Abstract
Purpose It remains unclear if fluid pressures used during cataract surgeries contribute to iatrogenic corneal endothelial cell (CEC) loss. Methods A custom experimental platform was used to pressurize the anterior chamber of explanted porcine eyes to surgical fluid pressures of 60 mm Hg or 400 mm Hg for 5 minutes or 60 mm Hg for 45 minutes (n = 8 or 9 per group). The corneal endothelia were stained with a unique combination of nucleic acid viability dyes and were imaged using fluorescence microscopy without removing the cornea from the globe. The images were analyzed using custom code to quantify acute CEC loss as a percentage of CEC injury/death (PCI). The PCI values from the surgical pressure groups were compared to sham controls perfused to physiological pressures for each surgical duration. As a positive control, a group of eyes (n = 8) was perfused with deionized water to intentionally induce CEC injury/death. Results No significant differences were observed in mean PCI values between any of the surgical fluid pressure groups and sham control groups, at either duration. The mean PCI of the positive control group was significantly different from all other groups, indicating that the method is able to detect CEC injury/death. Conclusions Our results suggest that the magnitudes of fluid pressure used over the duration of a cataract surgery do not significantly contribute to acute iatrogenic CEC loss. Translational Relevance Not only do the findings of this study answer a longstanding clinical question related to cataract surgery, but the platform introduced will facilitate testing how new cataract surgery devices and techniques affect CEC viability.
Collapse
Affiliation(s)
- Alex J. McMullen
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Zaynab A. Dantsoho
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Scott Chamness
- Carl Zeiss Meditec Cataract Technology, Inc., Reno, NV, USA
| | - John Brunelle
- Carl Zeiss Meditec Cataract Technology, Inc., Reno, NV, USA
| | - Jaime Martiz
- Carl Zeiss Meditec Cataract Technology, Inc., Reno, NV, USA
| | - Yousuf M. Khalifa
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mark R. Buckley
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Visual Science, University of Rochester, Rochester, NY, USA
| |
Collapse
|
23
|
Giri BR, Jakka D, Sandoval MA, Kulkarni VR, Bao Q. Advancements in Ocular Therapy: A Review of Emerging Drug Delivery Approaches and Pharmaceutical Technologies. Pharmaceutics 2024; 16:1325. [PMID: 39458654 PMCID: PMC11511072 DOI: 10.3390/pharmaceutics16101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Eye disorders affect a substantial portion of the global population, yet the availability of efficacious ophthalmic drug products remains limited. This can be partly ascribed to a number of factors: (1) inadequate understanding of physiological barriers, treatment strategies, drug and polymer properties, and delivery systems; (2) challenges in effectively delivering drugs to the anterior and posterior segments of the eye due to anatomical and physiological constraints; and (3) manufacturing and regulatory hurdles in ocular drug product development. The present review discusses innovative ocular delivery and treatments, encompassing implants, liposomes, nanoparticles, nanomicelles, microparticles, iontophoresis, in situ gels, contact lenses, microneedles, hydrogels, bispecific antibodies, and gene delivery strategies. Furthermore, this review also introduces advanced manufacturing technologies such as 3D printing and hot-melt extrusion (HME), aimed at improving bioavailability, reducing therapeutic dosages and side effects, facilitating the design of personalized ophthalmic dosage forms, as well as enhancing patient compliance. This comprehensive review lastly offers insights into digital healthcare, market trends, and industry and regulatory perspectives pertaining to ocular product development.
Collapse
Affiliation(s)
- Bhupendra Raj Giri
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (B.R.G.); (M.A.S.); (V.R.K.)
| | - Deeksha Jakka
- School of Pharmacy, The University of Mississippi, University, MS 38677, USA;
| | - Michael A. Sandoval
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (B.R.G.); (M.A.S.); (V.R.K.)
| | - Vineet R. Kulkarni
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (B.R.G.); (M.A.S.); (V.R.K.)
| | - Quanying Bao
- Synthetic Product Development, Alexion, AstraZeneca Rare Disease, 101 College Street, New Haven, CT 06510, USA
| |
Collapse
|
24
|
Brunel LG, Cai B, Hull SM, Han U, Wungcharoen T, Fernandes-Cunha GM, Seo YA, Johansson PK, Heilshorn SC, Myung D. In Situ UNIversal Orthogonal Network (UNION) Bioink Deposition for Direct Delivery of Corneal Stromal Stem Cells to Corneal Wounds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613997. [PMID: 39386574 PMCID: PMC11463654 DOI: 10.1101/2024.09.19.613997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The scarcity of human donor corneal graft tissue worldwide available for corneal transplantation necessitates the development of alternative therapeutic strategies for treating patients with corneal blindness. Corneal stromal stem cells (CSSCs) have the potential to address this global shortage by allowing a single donor cornea to treat multiple patients. To directly deliver CSSCs to corneal defects within an engineered biomatrix, we developed a UNIversal Orthogonal Network (UNION) collagen bioink that crosslinks in situ with a bioorthogonal, covalent chemistry. This cell-gel therapy is optically transparent, stable against contraction forces exerted by CSSCs, and permissive to the efficient growth of corneal epithelial cells. Furthermore, CSSCs remain viable within the UNION collagen gel precursor solution under standard storage and transportation conditions. This approach promoted corneal transparency and re-epithelialization in a rabbit anterior lamellar keratoplasty model, indicating that the UNION collagen bioink serves effectively as an in situ -forming, suture-free therapy for delivering CSSCs to corneal wounds. TEASER. Corneal stem cells are delivered within chemically crosslinked collagen as a transparent, regenerative biomaterial therapy.
Collapse
|
25
|
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
|
26
|
Kecik M, Kropp M, Thumann G, Pajic B, Guber J, Guber I. Intradevice Repeatability and Interdevice Comparison of Two Specular Microscopy Devices in a Real-Life Setting: Tomey EM-4000 and Nidek CEM-530. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1110. [PMID: 39064539 PMCID: PMC11278872 DOI: 10.3390/medicina60071110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: The purpose of this study was to compare two commercially available specular microscopes (Tomey EM-4000 and Nidek CEM-530) in a real-life clinical setting in terms of intra- and interdevice variability. The study was conducted on all patients seen in a clinical practice specializing in anterior segment pathologies, regardless of the purpose of their visit. Materials and Methods: In total, 112 eyes of 56 patients (age 23-85 years old) were included in the study. Each eye was measured three times with each device (for a total of six measurements), and results for central corneal thickness (CCT) and corneal endothelial cell density (ECD) were recorded. The results were then evaluated with the D'Agostino-Pearson normality test and compared with a Wilcoxon signed-rank test, t-test, ANOVA or Mann-Whitney test for intra- and interdevice variability. Results: Both specular microscopes produced very reliable reproducible intradevice results: The Tomey EM-4000 measured an ECD of 2390 ± 49.57 cells/mm2 (mean ± standard error of mean); the range was 799-3010 cells/mm2. The determined CCT was 546 ± 5.104 µm (mean ± standard error of mean [SEM]); the range was 425-615 µm. The measurements with the Nidek CEM-530 revealed an ECD of 2417 ± 0.09 cells/mm2 (mean ± SEM); the range was 505-3461 cells/mm2 (mean ± SEM). The mean CCT detected was 546.3 ± 4.937 µm (mean ± SEM); the range was 431-621 µm. The interdevice differences were statistically significant for both parameters, ECD (p = 0.0175) and CCT (p = 0.0125) (p < 0.05). Conclusions: The Nidek CEM-530 and the Tomey EM-4000 both produced reliable and reproducible results in terms of ECD and CCT. The absolute measurements were statistically significantly different for CCT and ECD for both devices; the Nidek produces slightly higher values.
Collapse
Affiliation(s)
- Mateusz Kecik
- Department of Ophthalmology, University of Geneva, 1205 Geneva, Switzerland; (M.K.); (M.K.); (G.T.); (B.P.)
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
| | - Martina Kropp
- Department of Ophthalmology, University of Geneva, 1205 Geneva, Switzerland; (M.K.); (M.K.); (G.T.); (B.P.)
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
| | - Gabriele Thumann
- Department of Ophthalmology, University of Geneva, 1205 Geneva, Switzerland; (M.K.); (M.K.); (G.T.); (B.P.)
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
| | - Bojan Pajic
- Department of Ophthalmology, University of Geneva, 1205 Geneva, Switzerland; (M.K.); (M.K.); (G.T.); (B.P.)
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
- Eye Clinic ORASIS, Swiss Eye Research Foundation, 5734 Reinach, Switzerland
- Faculty of Sciences, Department of Physics, University of Novi Sad, Trg Dositeja Obradovica 4, 21000 Novi Sad, Serbia
- Faculty of Medicine of the Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Josef Guber
- Department of Ophthalmology, University of Basel, 4001 Basel, Switzerland;
| | - Ivo Guber
- Department of Ophthalmology, University of Geneva, 1205 Geneva, Switzerland; (M.K.); (M.K.); (G.T.); (B.P.)
| |
Collapse
|
27
|
Hashemi A, Nabovati P, Hashemi H, Mortazavi AG, Khabazkhoob M. Corneal densitometry and associated factors in an elderly population. Clin Exp Optom 2024; 107:522-529. [PMID: 37674266 DOI: 10.1080/08164622.2023.2242864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/26/2023] [Indexed: 09/08/2023] Open
Abstract
CLINICAL RELEVANCE Knowing normative corneal optical density (COD) values is important for identifying pathologic corneal changes. BACKGROUND The aim of this work is to determine the distribution and associated factors of COD in the elderly population. METHODS This report is a part of a cross-sectional population-based study conducted on the elderly population (≥60 years) of Tehran, Iran from Jan 2019 to Jan 2020. Using a multi-stage stratified random cluster sampling method, a total of 160 clusters were randomly selected from 22 districts of Tehran city. Study participants underwent a complete ocular examination including measurement of visual acuity, refraction, and slit-lamp biomicroscopy. Corneal densitometry was evaluated using the Pentacam HR. Generalized estimating equation (GEE) models were used to investigate associations of COD with study variables. RESULTS A total of 3633 eyes from 2068 individuals were analysed. Of these, 1256 (60.7%) were female, and the mean age of the individuals was 66.42 ± 5.28 (60 to 95) years. The mean entire COD was 21.96 ± 4.45 greyscale unit (GSU) (95% CI: 21.57-22.34). There was a statistically significant difference in the mean COD between the anterior, central, and posterior layers (p < 0.001); the highest and lowest average COD was related to the anterior and posterior corneal layers, respectively. The lowest and highest mean COD were observed in the 0-2 mm [17.21GSU (95% CI:16.87-17.55)], and 10-12 mm annular zones [31.4 GSU (95% CI: 30.89-31.91)], respectively (p < 0.001). According to the multiple GEE model, the COD had a statistically significant direct relationship with age, central corneal thickness, and mean keratometry, while it was significantly inversely associated with female sex, anterior chamber depth, white-to-white distance, and corneal volume. CONCLUSION These normal values of COD in the present study could be used as reference data in older adults. The associated factors of COD should be taken into account to avoid misinterpretation of physiologic changes as pathologic processes.
Collapse
Affiliation(s)
- Alireza Hashemi
- Noor Ophthalmology Research Center, Noor Eye Hospital, Tehran, Iran
| | - Payam Nabovati
- Rehabilitation Research Center, Department of Optometry, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Hashemi
- Noor Research Center for Ophthalmic Epidemiology, Noor Eye Hospital, Tehran, Iran
| | - Abol Ghasem Mortazavi
- Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Khabazkhoob
- Department of Basic Sciences, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Jin L, Zhang L, Yan C, Liu M, Dean DC, Liu Y. Corneal injury repair and the potential involvement of ZEB1. EYE AND VISION (LONDON, ENGLAND) 2024; 11:20. [PMID: 38822380 PMCID: PMC11143703 DOI: 10.1186/s40662-024-00387-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
The cornea, consisting of three cellular and two non-cellular layers, is the outermost part of the eyeball and frequently injured by external physical, chemical, and microbial insults. The epithelial-to-mesenchymal transition (EMT) plays a crucial role in the repair of corneal injuries. Zinc finger E-box binding homeobox 1 (ZEB1), an important transcription factor involved in EMT, is expressed in the corneal tissues. It regulates cell activities like migration, transformation, and proliferation, and thereby affects tissue inflammation, fibrosis, tumor metastasis, and necrosis by mediating various major signaling pathways, including transforming growth factor (TGF)-β. Dysfunction of ZEB1 would impair corneal tissue repair leading to epithelial healing delay, interstitial fibrosis, neovascularization, and squamous cell metaplasia. Understanding the mechanism underlying ZEB1 regulation of corneal injury repair will help us to formulate a therapeutic approach to enhance corneal injury repair.
Collapse
Affiliation(s)
- Lin Jin
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Lijun Zhang
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Chunxiao Yan
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Mengxin Liu
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, China
| | - Douglas C Dean
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| | - Yongqing Liu
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
29
|
Wu Y, Benson MA, Sun SX. Cell-Driven Fluid Dynamics: A Physical Model of Active Systemic Circulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.19.594862. [PMID: 38826192 PMCID: PMC11142051 DOI: 10.1101/2024.05.19.594862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Active fluid circulation and transport are key functions of living organisms, which drive efficient delivery of oxygen and nutrients to various physiological compartments. Because fluid circulation occurs in a network, the systemic flux and pressure are not simple outcomes of any given component. Rather, they are emergent properties of network elements and network topology. Moreover, consistent pressure and osmolarity gradients across compartments such as the kidney, interstitium, and vessels are known. How these gradients and network properties are established and maintained is an unanswered question in systems physiology. Previous studies have shown that epithelial cells are fluid pumps that actively generate pressure and osmolarity gradients. Polarization and activity of ion exchangers that drive fluid flux in epithelial cells are affected by pressure and osmolarity gradients. Therefore, there is an unexplored coupling between the pressure and osmolarity in the circulating network. Here we develop a mathematical theory that integrates the influence of pressure and osmolarity on solute transport and explores both cell fluid transport and systemic circulation. This model naturally generates pressure and osmolarity gradients across physiological compartments, and demonstrates how systemic transport properties can depend on cell properties, and how the cell state can depend on systemic properties. When epithelial and endothelial pumps are considered together, we predict how pressures at various points in the network depend on the overall osmolarity of the system. The model can be improved by including physiological geometries and expanding solute species, and highlights the interplay of fluid properties with cell function in living organisms.
Collapse
Affiliation(s)
- Yufei Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Morgan A. Benson
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sean X. Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Vercammen H, Ondra M, Kotulova J, De La Hoz EC, Witters C, Jecmenova K, Le Compte M, Deben C, Ní Dhubhghaill S, Koppen C, Hajdúch M, Van den Bogerd B. "Keep on ROCKIn": Repurposed ROCK inhibitors to boost corneal endothelial regeneration. Biomed Pharmacother 2024; 174:116435. [PMID: 38513591 DOI: 10.1016/j.biopha.2024.116435] [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: 01/10/2024] [Revised: 03/06/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
The global shortage of corneal endothelial graft tissue necessitates the exploration of alternative therapeutic strategies. Rho-associated protein kinase inhibitors (ROCKi), recognized for their regenerative potential in cardiology, oncology, and neurology, have shown promise in corneal endothelial regeneration. This study investigates the repurposing potential of additional ROCKi compounds. Through screening a self-assembled library of ROCKi on B4G12 corneal endothelial cells, we evaluated their dose-dependent effects on proliferation, migration, and toxicity using live-cell imaging. Nine ROCKi candidates significantly enhanced B4G12 proliferation compared to the basal growth rate. These candidates were further assessed for their potential to accelerate wound closure as another indicator for tissue regeneration capacity, with most demonstrating notable efficacy. To assess the potential impact of candidate ROCKi on key corneal endothelial cell markers related to cell proliferation, leaky tight junctions and ion efflux capacity, we analyzed the protein expression of cyclin E1, CDK2, p16, ZO-1 and Na+/K+-ATPase, respectively. Immunocytochemistry and western blot analysis confirmed the preservation of corneal endothelial markers post-treatment with ROCKi hits. However, notable cytoplasm enlargement and nuclear fragmentation were detected after the treatment with SR-3677 and Thiazovivin, indicating possible cellular stress. In compared parameters, Chroman-1 at a concentration of 10 nM outperformed other ROCKi, requiring significantly 1000-fold lower effective concentration than established ROCKi Y-27632 and Fasudil. Altogether, this study underscores the potential of repurposing ROCKi for treating corneal endothelial dysfunctions, offering a viable alternative to conventional grafting methods, and highlights Chroman-1 as a promising candidate structure for hit-to-lead development.
Collapse
Affiliation(s)
- Hendrik Vercammen
- 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; DrugVision Lab, University of Antwerp, Wilrijk, Belgium.
| | - Martin Ondra
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic; Czech Advanced Technology and Research Institute (CATRIN), Palacky University Olomouc, Olomouc, Czech Republic
| | - Jana Kotulova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | | | - Charissa Witters
- 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; DrugVision Lab, University of Antwerp, Wilrijk, Belgium
| | - Katerina Jecmenova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | | | | | | | - 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
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic; Czech Advanced Technology and Research Institute (CATRIN), Palacky University Olomouc, Olomouc, Czech Republic
| | - Bert Van den Bogerd
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic; Czech Advanced Technology and Research Institute (CATRIN), Palacky University Olomouc, Olomouc, Czech Republic.
| |
Collapse
|
31
|
Okurowska K, Monk PN, Karunakaran E. Increased tolerance to commonly used antibiotics in a Pseudomonas aeruginosa ex vivo porcine keratitis model. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001459. [PMID: 38739119 PMCID: PMC11165664 DOI: 10.1099/mic.0.001459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/26/2024] [Indexed: 05/14/2024]
Abstract
Introduction. Bacterial keratitis, particularly caused by Pseudomonas aeruginosa, is challenging to treat because of multi-drug tolerance, often associated with the formation of biofilms. Antibiotics in development are typically evaluated against planktonic bacteria in a culture medium, which may not accurately represent the complexity of infections in vivo.Hypothesis/Gap Statement. Developing a reliable, economic ex vivo keratitis model that replicates some complexity of tissue infections could facilitate a deeper understanding of antibiotic efficacy, thus aiding in the optimization of treatment strategies for bacterial keratitis.Methodology. Here we investigated the efficacy of three commonly used antibiotics (gentamicin, ciprofloxacin and meropenem) against Pseudomonas aeruginosa cytotoxic strain PA14 and invasive strain PA01 using an ex vivo porcine keratitis model.Results. Both strains of P. aeruginosa were susceptible to the MIC of the three tested antibiotics. However, significantly higher concentrations were necessary to inhibit bacterial growth in the minimum biofilm eradication concentration (MBEC) assay, with both strains tolerating concentrations greater than 512 mg l-1 of meropenem. When MIC and higher concentrations than MBEC (1024 mg l-1) of antibiotics were applied, ciprofloxacin exhibited the highest potency against both P. aeruginosa strains, followed by meropenem, while gentamicin showed the least potency. Despite this, none of the antibiotic concentrations used effectively cleared the infection, even after 18 h of continuous exposure.Conclusions. Further exploration of antibiotic concentrations and aligning dosing with clinical studies to validate the model is needed. Nonetheless, our ex vivo porcine keratitis model could be a valuable tool for assessing antibiotic efficacy.
Collapse
Affiliation(s)
- Katarzyna Okurowska
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S10 2TN, UK
- National Institute for Health and Care Research, University of Leeds, Leeds LS2 9JT, UK
| | - Peter N. Monk
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Esther Karunakaran
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
32
|
Barathi VA, Katz A, Chaudhary S, Li HL, Tal DM, Marcovich A, Do CW, Karlish SJD. A digoxin derivative that potently reduces intraocular pressure: efficacy and mechanism of action in different animal models. Am J Physiol Cell Physiol 2024; 326:C1505-C1519. [PMID: 38557355 PMCID: PMC11371363 DOI: 10.1152/ajpcell.00617.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
Glaucoma is a blinding disease. Reduction of intraocular pressure (IOP) is the mainstay of treatment, but current drugs show side effects or become progressively ineffective, highlighting the need for novel compounds. We have synthesized a family of perhydro-1,4-oxazepine derivatives of digoxin, the selective inhibitor of Na,K-ATPase. The cyclobutyl derivative (DcB) displays strong selectivity for the human α2 isoform and potently reduces IOP in rabbits. These observations appeared consistent with a hypothesis that in ciliary epithelium DcB inhibits the α2 isoform of Na,K-ATPase, which is expressed strongly in nonpigmented cells, reducing aqueous humor (AH) inflow. This paper extends assessment of efficacy and mechanism of action of DcB using an ocular hypertensive nonhuman primate model (OHT-NHP) (Macaca fascicularis). In OHT-NHP, DcB potently lowers IOP, in both acute (24 h) and extended (7-10 days) settings, accompanied by increased aqueous humor flow rate (AFR). By contrast, ocular normotensive animals (ONT-NHP) are poorly responsive to DcB, if at all. The mechanism of action of DcB has been analyzed using isolated porcine ciliary epithelium and perfused enucleated eyes to study AH inflow and AH outflow facility, respectively. 1) DcB significantly stimulates AH inflow although prior addition of 8-Br-cAMP, which raises AH inflow, precludes additional effects of DcB. 2) DcB significantly increases AH outflow facility via the trabecular meshwork (TM). Taken together, the data indicate that the original hypothesis on the mechanism of action must be revised. In the OHT-NHP, and presumably other species, DcB lowers IOP by increasing AH outflow facility rather than by decreasing AH inflow.NEW & NOTEWORTHY When applied topically, a cyclobutyl derivative of digoxin (DcB) potently reduces intraocular pressure in an ocular hypertensive nonhuman primate model (Macaca fascicularis), associated with increased aqueous humor (AH) flow rate (AFR). The mechanism of action of DcB involves increased AH outflow facility as detected in enucleated perfused porcine eyes and, in parallel, increased (AH) inflow as detected in isolated porcine ciliary epithelium. DcB might have potential as a drug for the treatment of open-angle human glaucoma.
Collapse
Affiliation(s)
- Veluchamy Amutha Barathi
- Translational Pre-Clinical Model Platform, Singapore Institute of Eye Research (SERI)
- ACP in Ophthalmology & Visual Sciences, DUKE-NUS Graduate Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Adriana Katz
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shashikant Chaudhary
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | - Hoi-Lam Li
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | - Daniel M Tal
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Arie Marcovich
- Opthalmology Department, Kaplan Medical Center, Rehovot, Israel
- Hebrew University Medical School, Jerusalem, Israel
| | - Chi-Wai Do
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
- Centre for Eye and Vision Research (CEVR), Hong Kong, People's Republic of China
- Research Institute for Smart Ageing (RISA), The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | - Steven J D Karlish
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
33
|
Kisiel FB, Gurumurthy GJ. Endothelial cell loss post-implantable collamer lens V4c: meta-analysis. J Cataract Refract Surg 2024; 50:420-423. [PMID: 38194352 DOI: 10.1097/j.jcrs.0000000000001389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/03/2023] [Indexed: 01/10/2024]
Abstract
Endothelial cell density (ECD) loss is a noted effect of implantable collamer lens (ICL) V4c surgery. Current literature provides a wide range of values for ECD loss postsurgery, which may not be helpful in advising patients and clinicians. A meta-analysis exploring ECD loss in ICL V4c for myopia correction was undertaken. 18 studies were included in this meta-analysis with 2 subgroup analyses to account for the variability in follow-up lengths. The average ECD loss 3 months, 12 months, and 21.25 (mean) months postsurgery were 1.32% ± 1.28% ( P < .001, 95% CI, -75.158 to -1.19), 1.75% ± 2.17% ( P < .001, 95% CI, -134.09 to 14.52), and 3.84% ± 1.78% ( P < .001, 95% CI, -156.04 to -54.26), respectively. ECD loss is most pronounced 3 months postsurgery, suggesting that acute surgical trauma was the primary contributor rather than long-term lens implantation. Overall, ICL V4c is for myopia correction exhibits similar ECD loss as seen in other ICL models, thereby affirming its safety.
Collapse
Affiliation(s)
- Filip Blazej Kisiel
- From the Department of Chemical Engineering, University of Manchester, Manchester, United Kingdom (Kisiel); Department of Medicine, University of Manchester, Manchester, United Kingdom (Gurumurthy)
| | | |
Collapse
|
34
|
Melnyk S, Bollag WB. Aquaporins in the Cornea. Int J Mol Sci 2024; 25:3748. [PMID: 38612559 PMCID: PMC11011575 DOI: 10.3390/ijms25073748] [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: 02/20/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The cornea is an avascular, transparent tissue that allows light to enter the visual system. Accurate vision requires proper maintenance of the cornea's integrity and structure. Due to its exposure to the external environment, the cornea is prone to injury and must undergo proper wound healing to restore vision. Aquaporins (AQPs) are a family of water channels important for passive water transport and, in some family members, the transport of other small molecules; AQPs are expressed in all layers of the cornea. Although their functions as water channels are well established, the direct function of AQPs in the cornea is still being determined and is the focus of this review. AQPs, primarily AQP1, AQP3, and AQP5, have been found to play an important role in maintaining water homeostasis, the corneal structure in relation to proper hydration, and stress responses, as well as wound healing in all layers of the cornea. Due to their many functions in the cornea, the identification of drug targets that modulate the expression of AQPs in the cornea could be beneficial to promote corneal wound healing and restore proper function of this tissue crucial for vision.
Collapse
Affiliation(s)
- Samuel Melnyk
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Wendy B. Bollag
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Charlie Norwood Department of Veterans Affairs Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
35
|
Shimmura S, Inagaki E, Hirayama M, Hatou S. The Cornea: An Ideal Tissue for Regenerative Medicine. Keio J Med 2024; 73:1-7. [PMID: 38369325 DOI: 10.2302/kjm.2023-0001-ir] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Regenerative medicine is a highly anticipated field with hopes to provide cures for previously uncurable diseases such as spinal cord injuries and retinal blindness. Most regenerative medical products use either autologous or allogeneic stem cells, which may or may not be genetically modified. The introduction of induced-pluripotent stem cells (iPSCs) has fueled research in the field, and several iPSC-derived cells/tissues are currently undergoing clinical trials. The cornea is one of the pioneering areas of regenerative medicine, and already four cell therapy products are approved for clinical use in Japan. There is one other government-approved cell therapy product approved in Europe, but none are approved in the USA at present. The cornea is transparent and avascular, making it unique as a target for stem cell therapy. This review discusses the unique properties of the cornea and ongoing research in the field.
Collapse
Affiliation(s)
- Shigeto Shimmura
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Emi Inagaki
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shin Hatou
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Cellusion Inc., Tokyo, Japan
| |
Collapse
|
36
|
Grönroos P, Mörö A, Puistola P, Hopia K, Huuskonen M, Viheriälä T, Ilmarinen T, Skottman H. Bioprinting of human pluripotent stem cell derived corneal endothelial cells with hydrazone crosslinked hyaluronic acid bioink. Stem Cell Res Ther 2024; 15:81. [PMID: 38486306 PMCID: PMC10941625 DOI: 10.1186/s13287-024-03672-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Human corneal endothelial cells lack regenerative capacity through cell division in vivo. Consequently, in the case of trauma or dystrophy, the only available treatment modality is corneal tissue or primary corneal endothelial cell transplantation from cadaveric donor which faces a high global shortage. Our ultimate goal is to use the state-of-the-art 3D-bioprint technology for automated production of human partial and full-thickness corneal tissues using human stem cells and functional bioinks. In this study, we explore the feasibility of bioprinting the corneal endothelium using human pluripotent stem cell derived corneal endothelial cells and hydrazone crosslinked hyaluronic acid bioink. METHODS Corneal endothelial cells differentiated from human pluripotent stem cells were bioprinted using optimized hydrazone crosslinked hyaluronic acid based bioink. Before the bioprinting process, the biocompatibility of the bioink with cells was first analyzed with transplantation on ex vivo denuded rat and porcine corneas as well as on denuded human Descemet membrane. Subsequently, the bioprinting was proceeded and the viability of human pluripotent stem cell derived corneal endothelial cells were verified with live/dead stainings. Histological and immunofluorescence stainings involving ZO1, Na+/K+-ATPase and CD166 were used to confirm corneal endothelial cell phenotype in all experiments. Additionally, STEM121 marker was used to identify human cells from the ex vivo rat and porcine corneas. RESULTS The bioink, modified for human pluripotent stem cell derived corneal endothelial cells successfully supported both the viability and printability of the cells. Following up to 10 days of ex vivo transplantations, STEM121 positive cells were confirmed on the Descemet membrane of rat and porcine cornea demonstrating the biocompatibility of the bioink. Furthermore, biocompatibility was validated on denuded human Descemet membrane showing corneal endothelial -like characteristics. Seven days post bioprinting, the corneal endothelial -like cells were viable and showed polygonal morphology with expression and native-like localization of ZO-1, Na+/K+-ATPase and CD166. However, mesenchymal-like cells were observed in certain areas of the cultures, spreading beneath the corneal endothelial-like cell layer. CONCLUSIONS Our results demonstrate the successful printing of human pluripotent stem cell derived corneal endothelial cells using covalently crosslinked hyaluronic acid bioink. This approach not only holds promise for a corneal endothelium transplants but also presents potential applications in the broader mission of bioprinting the full-thickness human cornea.
Collapse
Affiliation(s)
- Pyry Grönroos
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Anni Mörö
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Paula Puistola
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Karoliina Hopia
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Maija Huuskonen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
- Tays Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Taina Viheriälä
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Tanja Ilmarinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Heli Skottman
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland.
| |
Collapse
|
37
|
Zhang BN, Qi B, Dong C, Zhang B, Cheng J, Wang X, Li S, Zhuang X, Chen S, Duan H, Li D, Zhu S, Li G, Cao Y, Zhou Q, Xie L. The role of corneal endothelium in macular corneal dystrophy development and recurrence. SCIENCE CHINA. LIFE SCIENCES 2024; 67:332-344. [PMID: 37480470 DOI: 10.1007/s11427-023-2364-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/10/2023] [Indexed: 07/24/2023]
Abstract
Macular corneal dystrophy (MCD) is a progressive, bilateral stromal dystrophic disease that arises from mutations in carbohydrate sulfotransferase 6 (CHST6). Corneal transplantation is the ultimate therapeutic solution for MCD patients. Unfortunately, postoperative recurrence remains a significant challenge. We conducted a retrospective review of a clinical cohort comprising 102 MCD patients with 124 eyes that underwent either penetrating keratoplasty (PKP) or deep anterior lamellar keratoplasty (DALK). Our results revealed that the recurrence rate was nearly three times higher in the DALK group (39.13%, 9/23 eyes) compared with the PKP group (10.89%, 11/101 eyes), suggesting that surgical replacement of the corneal endothelium for treating MCD is advisable to prevent postoperative recurrence. Our experimental data confirmed the robust mRNA and protein expression of CHST6 in human corneal endothelium and the rodent homolog CHST5 in mouse endothelium. Selective knockdown of wild-type Chst5 in mouse corneal endothelium (ACsiChst5), but not in the corneal stroma, induced experimental MCD with similar extracellular matrix synthesis impairments and corneal thinning as observed in MCD patients. Mice carrying Chst5 point mutation also recapitulated clinical phenotypes of MCD, along with corneal endothelial abnormalities. Intracameral injection of wild-type Chst5 rescued the corneal impairments in ACsiChst5 mice and retarded the disease progression in Chst5 mutant mice. Overall, our study provides new mechanistic insights and therapeutic approaches for MCD treatment by high-lighting the role of corneal endothelium in MCD development.
Collapse
Affiliation(s)
- Bi-Ning Zhang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
| | - Benxiang Qi
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
| | - Chunxiao Dong
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
- Department of Medicine, Qingdao University, Qingdao, 266071, China
| | - Bin Zhang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
| | - Jun Cheng
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
| | - Xin Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021, China
| | - Suxia Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021, China
| | - Xiaoyun Zhuang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
- Department of Ophthalmology, School of Clinical Medicine, Weifang Medical University, Weifang, 261072, China
| | - Shijiu Chen
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
- Department of Medicine, Qingdao University, Qingdao, 266071, China
| | - Haoyun Duan
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
| | - Dewei Li
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China
| | - Sujie Zhu
- Institute of Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266071, China
| | - Guoyun Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266071, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China.
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China.
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China.
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China.
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China.
- School of Ophthalmology, Shandong First Medical University, Qingdao, 250021, China.
| |
Collapse
|
38
|
Hadrian K, Cursiefen C. The role of lymphatic vessels in corneal fluid homeostasis and wound healing. J Ophthalmic Inflamm Infect 2024; 14:4. [PMID: 38252213 PMCID: PMC10803698 DOI: 10.1186/s12348-023-00381-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/16/2023] [Indexed: 01/23/2024] Open
Abstract
The cornea, essential for vision, is normally avascular, transparent, and immune-privileged. However, injuries or infections can break this privilege, allowing blood and lymphatic vessels to invade, potentially impairing vision and causing immune responses. This review explores the complex role of corneal lymphangiogenesis in health and diseases. Traditionally, the cornea was considered devoid of lymphatic vessels, a phenomenon known as "corneal (lymph)angiogenic privilege." Recent advances in molecular markers have enabled the discovery of lymphatic vessels in the cornea under certain conditions. Several molecules contribute to preserving both immune and lymphangiogenic privileges. Lymphangiogenesis, primarily driven by VEGF family members, can occur directly or indirectly through macrophage recruitment. Corneal injuries and diseases disrupt these privileges, reducing graft survival rates following transplantation. However, modulation of lymphangiogenesis offers potential interventions to promote graft survival and expedite corneal edema resolution.This review underscores the intricate interplay between lymphatic vessels, immune privilege, and corneal pathologies, highlighting innovative therapeutic possibilities. Future investigations should explore the modulation of lymphangiogenesis to enhance corneal health and transparency, as well as corneal graft survival, and this benefits patients with various corneal conditions.
Collapse
Affiliation(s)
- Karina Hadrian
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
39
|
Zhang X, Kumar A, Sathe AA, Mootha VV, Xing C. Transcriptomic meta-analysis reveals ERRα-mediated oxidative phosphorylation is downregulated in Fuchs' endothelial corneal dystrophy. PLoS One 2023; 18:e0295542. [PMID: 38096202 PMCID: PMC10721014 DOI: 10.1371/journal.pone.0295542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/25/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Late-onset Fuchs' endothelial corneal dystrophy (FECD) is a degenerative disease of cornea and the leading indication for corneal transplantation. Genetically, FECD patients can be categorized as with (RE+) or without (RE-) the CTG trinucleotide repeat expansion in the transcription factor 4 gene. The molecular mechanisms underlying FECD remain unclear, though there are plausible pathogenic models proposed for RE+ FECD. METHOD In this study, we performed a meta-analysis on RNA sequencing datasets of FECD corneal endothelium including 3 RE+ datasets and 2 RE- datasets, aiming to compare the transcriptomic profiles of RE+ and RE- FECD. Gene differential expression analysis, co-expression networks analysis, and pathway analysis were conducted. RESULTS There was a striking similarity between RE+ and RE- transcriptomes. There were 1,184 genes significantly upregulated and 1,018 genes significantly downregulated in both RE+ and RE- cases. Pathway analysis identified multiple biological processes significantly enriched in both-mitochondrial functions, energy-related processes, ER-nucleus signaling pathway, demethylation, and RNA splicing were negatively enriched, whereas small GTPase mediated signaling, actin-filament processes, extracellular matrix organization, stem cell differentiation, and neutrophil mediated immunity were positively enriched. The translational initiation process was downregulated in the RE+ transcriptomes. Gene co-expression analysis identified modules with relatively distinct biological processes enriched including downregulation of mitochondrial respiratory chain complex assembly. The majority of oxidative phosphorylation (OXPHOS) subunit genes, as well as their upstream regulator gene estrogen-related receptor alpha (ESRRA), encoding ERRα, were downregulated in both RE+ and RE- cases, and the expression level of ESRRA was correlated with that of OXPHOS subunit genes. CONCLUSION Meta-analysis increased the power of detecting differentially expressed genes. Integrating differential expression analysis with co-expression analysis helped understand the underlying molecular mechanisms. FECD RE+ and RE- transcriptomic profiles are much alike with the hallmark of downregulation of genes in pathways related to ERRα-mediated OXPHOS.
Collapse
Affiliation(s)
- Xunzhi Zhang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Adwait A. Sathe
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - V. Vinod Mootha
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- O’Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
40
|
Sakakura S, Inagaki E, Sayano T, Yamazaki R, Fusaki N, Hatou S, Hirayama M, Tsubota K, Negishi K, Okano H, Shimmura S. Non-apoptotic regulated cell death in Fuchs endothelial corneal dystrophy. Regen Ther 2023; 24:592-601. [PMID: 38034859 PMCID: PMC10681880 DOI: 10.1016/j.reth.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/15/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Fuchs endothelial corneal dystrophy (FECD) is the leading cause of corneal blindness in developed countries. Corneal endothelial cells in FECD are susceptive to oxidative stress, leading to mitochondrial dysfunction and cell death. Oxidative stress causes many forms of cell death including parthanatos, which is characterized by translocation of apoptosis-inducing factor (AIF) to the nucleus with upregulation of poly (ADP-ribose) polymerase 1 (PARP-1) and poly (ADP-ribose) (PAR). Although cell death is an important aspect of FECD, previous reports have often analyzed immortalized cell lines, making the evaluation of cell death difficult. Therefore, we established a new in vitro FECD model to evaluate the pathophysiology of FECD. Methods Corneal endothelial cells were derived from disease-specific induced pluripotent stem cells (iPSCs). Hydrogen peroxide (H2O2) was used as a source for oxidative stress to mimic the pathophysiology of FECD. We investigated the responses to oxidative stress and the involvement of parthanatos in FECD-corneal endothelial cells. Results Cell death ratio and oxidative stress level were upregulated in FECD with H2O2 treatment compared with non-FECD control, indicating the vulnerability of oxidative stress in FECD. We also found that intracellular PAR, as well as PARP-1 and AIF in the nucleus were upregulated in FECD. Furthermore, PARP inhibition, but not pan-caspase inhibition, rescued cell death, DNA double-strand breaks, mitochondrial membrane potential depolarization and energy depletion, suggesting that cell death was mainly due to parthanatos. Conclusions We report that parthanatos may be involved in the pathophysiology of FECD and targeting this cell death pathway may be a potential therapeutic approach for FECD.
Collapse
Affiliation(s)
- Saki Sakakura
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Emi Inagaki
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
- Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda, Tokyo 102-0083, Japan
| | - Tomoko Sayano
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
- Cellusion Inc. 8-6 Nihonbashi-Kobunacho, Chuo-ku, Tokyo 103-0024, Japan
| | - Risa Yamazaki
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Noemi Fusaki
- University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center, 4-6-1 Shirokanedai, Minato, Tokyo 108-8639, Japan
| | - Shin Hatou
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
- Cellusion Inc. 8-6 Nihonbashi-Kobunacho, Chuo-ku, Tokyo 103-0024, Japan
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Shigeto Shimmura
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Haneda Innovation City Zone A, 1-1-4, Hanedakuko, Ota-ku, Tokyo 144-0041, Japan
| |
Collapse
|
41
|
Tarvestad-Laise KE, Ceresa BP. Modulating Growth Factor Receptor Signaling to Promote Corneal Epithelial Homeostasis. Cells 2023; 12:2730. [PMID: 38067157 PMCID: PMC10706396 DOI: 10.3390/cells12232730] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
The corneal epithelium is the first anatomical barrier between the environment and the cornea; it is critical for proper light refraction onto the retina and prevents pathogens (e.g., bacteria, viruses) from entering the immune-privileged eye. Trauma to the highly innervated corneal epithelium is extremely painful and if not resolved quickly or properly, can lead to infection and ultimately blindness. The healthy eye produces its own growth factors and is continuously bathed in tear fluid that contains these proteins and other nutrients to maintain the rapid turnover and homeostasis of the ocular surface. In this article, we review the roles of growth factors in corneal epithelial homeostasis and regeneration and some of the limitations to their use therapeutically.
Collapse
Affiliation(s)
- Kate E. Tarvestad-Laise
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Brian P. Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
- Department of Ophthalmology and Vision Sciences, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
42
|
Yu J, Yu N, Tian Y, Fang Y, An B, Feng G, Wu J, Wang L, Hao J, Wang L, Zhou Q, Li W, Wang Y, Hu B. Safety and efficacy of human ESC-derived corneal endothelial cells for corneal endothelial dysfunction. Cell Biosci 2023; 13:201. [PMID: 37932828 PMCID: PMC10629087 DOI: 10.1186/s13578-023-01145-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/12/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Research on human pluripotent stem cells (hPSCs) has shown tremendous progress in cell-based regenerative medicine. Corneal endothelial dysfunction is associated with the loss and degeneration of corneal endothelial cells (CECs), rendering cell replacement a promising therapeutic strategy. However, comprehensive preclinical assessments of hPSC-derived CECs for this cell therapy remain a challenge. RESULTS Here we defined an adapted differentiation protocol to generate induced corneal endothelial cells (iCECs) consistently and efficiently from clinical-grade human embryonic stem cells (hESCs) with xeno-free medium and manufactured cryopreserved iCECs. Cells express high levels of typical CECs markers and exhibit transendothelial potential properties in vitro typical of iCECs. After rigorous quality control measures, cells meeting all release criteria were available for in vivo studies. We found that there was no overgrowth or tumorigenicity of grafts in immunodeficient mice. After grafting into rabbit models, the surviving iCECs ameliorated edema and recovered corneal opacity. CONCLUSIONS Our work provides an efficient approach for generating iCECs and demonstrates the safety and efficacy of iCECs in disease modeling. Therefore, clinical-grade iCECs are a reliable source for future clinical treatment of corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Juan Yu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Nianye Yu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Yao Tian
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Yifan Fang
- Department of Ophthalmology, The First Center of the PLA General Hospital, Beijing, China
| | - Bin An
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jie Hao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
| | - Liqiang Wang
- Department of Ophthalmology, The First Center of the PLA General Hospital, Beijing, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- National Stem Cell Resource Center, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Baoyang Hu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| |
Collapse
|
43
|
Moon CE, Kim CH, Jung JH, Cho YJ, Choi KY, Han K, Seo KY, Lee HK, Ji YW. Integrated Analysis of Transcriptome and Proteome of the Human Cornea and Aqueous Humor Reveal Novel Biomarkers for Corneal Endothelial Cell Dysfunction. Int J Mol Sci 2023; 24:15354. [PMID: 37895034 PMCID: PMC10607268 DOI: 10.3390/ijms242015354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Earlier studies have reported that elevated protein levels in the aqueous humor (AH) are associated with corneal endothelial cell dysfunction (CECD), but the details of the underlying mechanism as well as specific biomarkers for CECD remain elusive. In the present study, we aimed to identify protein markers in AH directly associated with changes to corneal endothelial cells (CECs), as AH can be easily obtained for analysis. We carried out an in-depth proteomic analysis of patient-derived AH as well as transcriptomic analysis of CECs from the same patients with bullous keratopathy (BK) resulting from CECD. We first determined differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) from CECs and AH in CECD, respectively. By combining transcriptomic and proteomic analyses, 13 shared upregulated markers and 22 shared downregulated markers were observed between DEGs and DEPs. Among these 35 candidates from biomarker profiling, three upregulated markers were finally verified via data-independent acquisition (DIA) proteomic analysis using additional individual AH samples, namely metallopeptidase inhibitor 1 (TIMP1), Fc fragment of IgG binding protein (FCGBP), and angiopoietin-related protein 7 (ANGPTL7). Furthermore, we confirmed these AH biomarkers for CECD using individual immunoassay validation. Conclusively, our findings may provide valuable insights into the disease process and identify biofluid markers for the assessment of CEC function during BK development.
Collapse
Affiliation(s)
- Chae-Eun Moon
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (C.-E.M.)
| | - Chang Hwan Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (C.-E.M.)
- Department of Ophthalmology, Yongin Severance Hospital, Yongin 16995, Republic of Korea
| | - Jae Hun Jung
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Young Joo Cho
- The Yonsei Eye Clinic, Seoul 06289, Republic of Korea
- Department of Ophthalmology, HanGil Eye Hospital, Incheon 21388, Republic of Korea
| | - Kee Yong Choi
- Department of Ophthalmology, HanGil Eye Hospital, Incheon 21388, Republic of Korea
| | - Kyusun Han
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (C.-E.M.)
| | - Kyoung Yul Seo
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (C.-E.M.)
| | - Hyung Keun Lee
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (C.-E.M.)
- Department of Ophthalmology, Gangnam Severance Hospital, Seoul 06273, Republic of Korea
- College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Yong Woo Ji
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (C.-E.M.)
- Department of Ophthalmology, Yongin Severance Hospital, Yongin 16995, Republic of Korea
| |
Collapse
|
44
|
Wu YF, Chang NW, Chu LA, Liu HY, Zhou YX, Pai YL, Yu YS, Kuan CH, Wu YC, Lin SJ, Tan HY. Single-Cell Transcriptomics Reveals Cellular Heterogeneity and Complex Cell-Cell Communication Networks in the Mouse Cornea. Invest Ophthalmol Vis Sci 2023; 64:5. [PMID: 37792336 PMCID: PMC10565710 DOI: 10.1167/iovs.64.13.5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 06/30/2023] [Indexed: 10/05/2023] Open
Abstract
Purpose To generate a single-cell RNA-sequencing (scRNA-seq) map and construct cell-cell communication networks of mouse corneas. Methods C57BL/6 mouse corneas were dissociated to single cells and subjected to scRNA-seq. Cell populations were clustered and annotated for bioinformatic analysis using the R package "Seurat." Differential expression patterns were validated and spatially mapped with whole-mount immunofluorescence staining. Global intercellular signaling networks were constructed using CellChat. Results Unbiased clustering of scRNA-seq transcriptomes of 14,732 cells from 40 corneas revealed 17 cell clusters of six major cell types: nine epithelial cell, three keratocyte, two corneal endothelial cell, and one each of immune cell, vascular endothelial cell, and fibroblast clusters. The nine epithelial cell subtypes included quiescent limbal stem cells, transit-amplifying cells, and differentiated cells from corneas and two minor conjunctival epithelial clusters. CellChat analysis provided an atlas of the complex intercellular signaling communications among all cell types. Conclusions We constructed a complete single-cell transcriptomic map and the complex signaling cross-talk among all cell types of the cornea, which can be used as a foundation atlas for further research on the cornea. This study also deepens the understanding of the cellular heterogeneity and heterotypic cell-cell interaction within corneas.
Collapse
Affiliation(s)
- Yueh-Feng Wu
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Nai-Wen Chang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-An Chu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsin-Yu Liu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Ophthalmology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Yu-Xian Zhou
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Yun-Lin Pai
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Sheng Yu
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Chen-Hsiang Kuan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ching Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Hsin-Yuan Tan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
45
|
Català P, Vivensang F, van Beek D, Adriaens ME, Dickman MM, LaPointe VLS, Kutmon M. Elucidating the Corneal Endothelial Cell Proliferation Capacity through an Interspecies Transcriptome Comparison. Adv Biol (Weinh) 2023; 7:e2300065. [PMID: 37062753 DOI: 10.1002/adbi.202300065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/20/2023] [Indexed: 04/18/2023]
Abstract
The regenerative capacity of corneal endothelial cells (CECs) differs between species; in bigger mammals, CECs are arrested in a non-proliferative state. Damage to these cells can compromise their function causing corneal opacity. Corneal transplantation is the current treatment for the recovery of clear eyesight, but the donor tissue demand is higher than the availability and there is a need to develop novel treatments. Interestingly, rabbit CECs retain a high proliferative profile and can repopulate the endothelium. There is a lack of fundamental knowledge to explain these differences. Gaining information on their transcriptomic variances could allow the identification of CEC proliferation drivers. In this study, human, sheep, and rabbit CECs are analyzed at the transcriptomic level. To understand the differences across each species, a pipeline for the analysis of pathways with different activities is generated. The results reveal that 52 pathways have different activity when comparing species with non-proliferative CECs (human and sheep) to species with proliferative CECs (rabbit). The results show that Notch and TGF-β pathways have increased activity in species with non-proliferative CECs, which might be associated with their low proliferation. Overall, this study illustrates transcriptomic pathway-level differences that can provide leads to develop novel therapies to regenerate the corneal endothelium.
Collapse
Affiliation(s)
- Pere Català
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, 6229ER, The Netherlands
- University Eye Clinic Maastricht, Maastricht University Medical Center+, Maastricht, 6229HX, The Netherlands
| | - Flora Vivensang
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, 6229ER, The Netherlands
| | - Daan van Beek
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6229EN, The Netherlands
| | - Michiel E Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6229EN, The Netherlands
| | - Mor M Dickman
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, 6229ER, The Netherlands
- University Eye Clinic Maastricht, Maastricht University Medical Center+, Maastricht, 6229HX, The Netherlands
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, 6229ER, The Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6229EN, The Netherlands
| |
Collapse
|
46
|
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
|
47
|
Clahsen T, Hadrian K, Notara M, Schlereth SL, Howaldt A, Prokosch V, Volatier T, Hos D, Schroedl F, Kaser-Eichberger A, Heindl LM, Steven P, Bosch JJ, Steinkasserer A, Rokohl AC, Liu H, Mestanoglu M, Kashkar H, Schumacher B, Kiefer F, Schulte-Merker S, Matthaei M, Hou Y, Fassbender S, Jantsch J, Zhang W, Enders P, Bachmann B, Bock F, Cursiefen C. The novel role of lymphatic vessels in the pathogenesis of ocular diseases. Prog Retin Eye Res 2023; 96:101157. [PMID: 36759312 DOI: 10.1016/j.preteyeres.2022.101157] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 02/10/2023]
Abstract
Historically, the eye has been considered as an organ free of lymphatic vessels. In recent years, however, it became evident, that lymphatic vessels or lymphatic-like vessels contribute to several ocular pathologies at various peri- and intraocular locations. The aim of this review is to outline the pathogenetic role of ocular lymphatics, the respective molecular mechanisms and to discuss current and future therapeutic options based thereon. We will give an overview on the vascular anatomy of the healthy ocular surface and the molecular mechanisms contributing to corneal (lymph)angiogenic privilege. In addition, we present (i) current insights into the cellular and molecular mechanisms occurring during pathological neovascularization of the cornea triggered e.g. by inflammation or trauma, (ii) the role of lymphatic vessels in different ocular surface pathologies such as dry eye disease, corneal graft rejection, ocular graft versus host disease, allergy, and pterygium, (iii) the involvement of lymphatic vessels in ocular tumors and metastasis, and (iv) the novel role of the lymphatic-like structure of Schlemm's canal in glaucoma. Identification of the underlying molecular mechanisms and of novel modulators of lymphangiogenesis will contribute to the development of new therapeutic targets for the treatment of ocular diseases associated with pathological lymphangiogenesis in the future. The preclinical data presented here outline novel therapeutic concepts for promoting transplant survival, inhibiting metastasis of ocular tumors, reducing inflammation of the ocular surface, and treating glaucoma. Initial data from clinical trials suggest first success of novel treatment strategies to promote transplant survival based on pretransplant corneal lymphangioregression.
Collapse
Affiliation(s)
- Thomas Clahsen
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Karina Hadrian
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Maria Notara
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Simona L Schlereth
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Antonia Howaldt
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Verena Prokosch
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Volatier
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Hos
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Falk Schroedl
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Ludwig M Heindl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Steven
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany
| | - Jacobus J Bosch
- Centre for Human Drug Research and Leiden University Medical Center, Leiden, the Netherlands
| | | | - Alexander C Rokohl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hanhan Liu
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Mert Mestanoglu
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Institute for Molecular Immunology, Center for Molecular Medicine Cologne (CMMC), CECAD Research Center, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Björn Schumacher
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany
| | - Friedemann Kiefer
- European Institute for Molecular Imaging (EIMI), University of Münster, 48149, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster, Germany
| | - Mario Matthaei
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Yanhong Hou
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, China
| | - Sonja Fassbender
- IUF‒Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; Immunology and Environment, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wei Zhang
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philip Enders
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Björn Bachmann
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Felix Bock
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany.
| |
Collapse
|
48
|
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
|
49
|
Ng XY, Peh GSL, Yam GHF, Tay HG, Mehta JS. Corneal Endothelial-like Cells Derived from Induced Pluripotent Stem Cells for Cell Therapy. Int J Mol Sci 2023; 24:12433. [PMID: 37569804 PMCID: PMC10418878 DOI: 10.3390/ijms241512433] [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: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Corneal endothelial dysfunction is one of the leading causes of corneal blindness, and the current conventional treatment option is corneal transplantation using a cadaveric donor cornea. However, there is a global shortage of suitable donor graft material, necessitating the exploration of novel therapeutic approaches. A stem cell-based regenerative medicine approach using induced pluripotent stem cells (iPSCs) offers a promising solution, as they possess self-renewal capabilities, can be derived from adult somatic cells, and can be differentiated into all cell types including corneal endothelial cells (CECs). This review discusses the progress and challenges in developing protocols to induce iPSCs into CECs, focusing on the different media formulations used to differentiate iPSCs to neural crest cells (NCCs) and subsequently to CECs, as well as the characterization methods and markers that define iPSC-derived CECs. The hurdles and solutions for the clinical application of iPSC-derived cell therapy are also addressed, including the establishment of protocols that adhere to good manufacturing practice (GMP) guidelines. The potential risks of genetic mutations in iPSC-derived CECs associated with long-term in vitro culture and the danger of potential tumorigenicity following transplantation are evaluated. In all, this review provides insights into the advancement and obstacles of using iPSC in the treatment of corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Xiao Yu Ng
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore; (X.Y.N.); (G.S.L.P.); (G.H.-F.Y.)
| | - Gary S. L. Peh
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore; (X.Y.N.); (G.S.L.P.); (G.H.-F.Y.)
- Ophthalmology and Visual Sciences Academic Clinical Program, SingHealth and Duke-NUS Medical School, Singapore 169857, Singapore;
| | - Gary Hin-Fai Yam
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore; (X.Y.N.); (G.S.L.P.); (G.H.-F.Y.)
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh, 6614, Pittsburgh, PA 15260, USA
| | - Hwee Goon Tay
- Ophthalmology and Visual Sciences Academic Clinical Program, SingHealth and Duke-NUS Medical School, Singapore 169857, Singapore;
- Centre for Vision Research, DUKE-NUS Medical School, Singapore 169857, Singapore
| | - Jodhbir S. Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore; (X.Y.N.); (G.S.L.P.); (G.H.-F.Y.)
- Ophthalmology and Visual Sciences Academic Clinical Program, SingHealth and Duke-NUS Medical School, Singapore 169857, Singapore;
- Centre for Vision Research, DUKE-NUS Medical School, Singapore 169857, Singapore
- Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore 168751, Singapore
| |
Collapse
|
50
|
Zhu YT, Tighe S, Chen SL, Zhang Y, Chen SY, Kao WWY, Tseng SCG. Manufacturing of human corneal endothelial grafts. Ocul Surf 2023; 29:301-310. [PMID: 37268293 PMCID: PMC10529356 DOI: 10.1016/j.jtos.2023.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/04/2023]
Abstract
PURPOSE Human corneal endothelial cells (HCECs) play a significant role in maintaining visual function. However, these cells are notorious for their limited proliferative capacity in vivo. Current treatment of corneal endothelial dysfunction resorts to corneal transplantation. Herein we describe an ex vivo engineering method to manufacture HCEC grafts suitable for transplantation through reprogramming into neural crest progenitors. METHODS HCECs were isolated by collagenase A from stripped Descemet membrane of cadaveric corneoscleral rims, and induced reprogramming via knockdown with p120 and Kaiso siRNAs on collagen IV-coated atelocollagen. Engineered HCEC grafts were released after assessing their identity, potency, viability, purity and sterility. Phase contrast was used for monitoring cell shape, graft size, and cell density. Immunostaining was used to determine the normal HCEC phenotype with expression of N-cadherin, ZO-1, ATPase, acetyl-α-tubulin, γ-tubulin, p75NTR, α-catenin, β-catenin, and F-actin. Stability of manufactured HCEC graft was evaluated after transit and storage for up to 3 weeks. The pump function of HCEC grafts was measured by lactate efflux. RESULTS One HCEC graft suitable for corneal transplantation was generated from 1/8th of the donor corneoscleral rim with normal hexagonal cell shape, density, and phenotype. The manufactured grafts were stable for up to 3 weeks at 37 °C or up to 1 week at 22 °C in MESCM medium and after transcontinental shipping at room temperature by retaining normal morphology (hexagonal, >2000 cells/mm2, >8 mm diameter), phenotype, and pump function. CONCLUSIONS This regenerative strategy through knockdown with p120 and Kaiso siRNAs can be used to manufacture HCEC grafts with normal phenotype, morphology and pump function following prolonged storage and shipping.
Collapse
Affiliation(s)
| | - Sean Tighe
- R&D Department, BioTissue, Miami, FL, 33126, USA
| | | | - Yuan Zhang
- R&D Department, BioTissue, Miami, FL, 33126, USA
| | - Szu-Yu Chen
- R&D Department, BioTissue, Miami, FL, 33126, USA
| | - Winston W Y Kao
- Department of Ophthalmology, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH, 45220, USA
| | | |
Collapse
|