1
|
Shen L, Han F, Pan L, Du L, Sun P, Zhang K, Wu X, Pang K, Zhu J. Construction of tissue engineered cornea with skin-derived corneal endothelial-like cell and mechanism research for the cell differentiation. Front Med (Lausanne) 2024; 11:1448248. [PMID: 39286645 PMCID: PMC11402686 DOI: 10.3389/fmed.2024.1448248] [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: 06/13/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Corneal endothelial transplantation accounts for most of corneal transplantation for treating corneal diseases, however severe shortage of corneal donors is the biggest obstacle. In our previous study, we differentiated human skin-derived precursors (SKPs) into corneal endothelial cell (CEC)-like cells with a co-culture system. In this study, we aimed to investigate cell differentiation molecular mechanism and evaluate the function of CEC-like cells by developing tissue-engineered corneas in order to improve cell production efficiency and provide basic research for clinical transformation. Methods We performed transcriptome sequencing of SKPs and CEC-like cells. Further, we focused on the possible enriching pathways, including PI3K/Akt, MAPK/Erk, WNT/β-catenin, and important transcription factors Pitx2 and Foxc1. The PI3K and β-catenin inhibitors were also added to the culture system to observe the differentiation alteration. We developed a graft for a tissue-engineered cornea (TEC) using CEC-like cells and acellular porcine cornea matrix scaffold. The tissue-engineered corneas were transplanted into rabbits via penetrating keratoplasty. Results The PI3K/Akt, MAPK/Erk, and WNT/β-catenin pathways play important roles during the differentiation of SKPs into CEC-like cells. Crosstalk existed between the PI3K/Akt and MAPK/Erk pathways. The PI3K/Akt and WNT/β-catenin pathways were connected. Pitx2 and Foxc1 were subject to temporal and spatial controls of the WNT/β-catenin pathway. The inhibition of the PI3K/Akt and WNT/β-catenin pathways both prevented cell differentiation. CEC-like cells grew well on the acellular porcine cornea matrix scaffold, and the tissue-engineered corneal graft performed well after transplantation into rabbits. Conclusion We provide experimental basis for CEC-like cell industrial production and drive the cells to be clinically applied in cellular replacement therapy or alternative graft substitution for treating corneal diseases in the future.
Collapse
Affiliation(s)
- Lin Shen
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Fang Han
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Lijie Pan
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Liqun Du
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Peng Sun
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kai Zhang
- Department of Ophthalmology, Shandong Second Provincial General Hospital, Jinan, China
| | - Xinyi Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Kunpeng Pang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Zhu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
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
|
3
|
Yam GHF, Pi S, Du Y, Mehta JS. Posterior corneoscleral limbus: Architecture, stem cells, and clinical implications. Prog Retin Eye Res 2023; 96:101192. [PMID: 37392960 DOI: 10.1016/j.preteyeres.2023.101192] [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: 04/19/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023]
Abstract
The limbus is a transition from the cornea to conjunctiva and sclera. In human eyes, this thin strip has a rich variation of tissue structures and composition, typifying a change from scleral irregularity and opacity to corneal regularity and transparency; a variation from richly vascularized conjunctiva and sclera to avascular cornea; the neural passage and drainage of aqueous humor. The limbal stroma is enriched with circular fibres running parallel to the corneal circumference, giving its unique role in absorbing small pressure changes to maintain corneal curvature and refractivity. It contains specific niches housing different types of stem cells for the corneal epithelium, stromal keratocytes, corneal endothelium, and trabecular meshwork. This truly reflects the important roles of the limbus in ocular physiology, and the limbal functionality is crucial for corneal health and the entire visual system. Since the anterior limbus containing epithelial structures and limbal epithelial stem cells has been extensively reviewed, this article is focused on the posterior limbus. We have discussed the structural organization and cellular components of the region beneath the limbal epithelium, the characteristics of stem cell types: namely corneal stromal stem cells, endothelial progenitors and trabecular meshwork stem cells, and recent advances leading to the emergence of potential cell therapy options to replenish their respective mature cell types and to correct defects causing corneal abnormalities. We have reviewed different clinical disorders associated with defects of the posterior limbus and summarized the available preclinical and clinical evidence about the developing topic of cell-based therapy for corneal disorders.
Collapse
Affiliation(s)
- Gary Hin-Fai Yam
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.
| | - Shaohua Pi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yiqin Du
- Department of Ophthalmology, University of South Florida, Tampa, FL, USA
| | - Jodhbir S Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore; Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-National University of Singapore (NUS) Medical School, Singapore.
| |
Collapse
|
4
|
Attico E, Galaverni G, Torello A, Bianchi E, Bonacorsi S, Losi L, Manfredini R, Lambiase A, Rama P, Pellegrini G. Comparison between Cultivated Oral Mucosa and Ocular Surface Epithelia for COMET Patients Follow-Up. Int J Mol Sci 2023; 24:11522. [PMID: 37511281 PMCID: PMC10380900 DOI: 10.3390/ijms241411522] [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: 05/16/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Total bilateral Limbal Stem Cell Deficiency is a pathologic condition of the ocular surface due to the loss of corneal stem cells. Cultivated oral mucosa epithelial transplantation (COMET) is the only autologous successful treatment for this pathology in clinical application, although abnormal peripheric corneal vascularization often occurs. Properly characterizing the regenerated ocular surface is needed for a reliable follow-up. So far, the univocal identification of transplanted oral mucosa has been challenging. Previously proposed markers were shown to be co-expressed by different ocular surface epithelia in a homeostatic or perturbated environment. In this study, we compared the transcriptome profile of human oral mucosa, limbal and conjunctival cultured holoclones, identifying Paired Like Homeodomain 2 (PITX2) as a new marker that univocally distinguishes the transplanted oral tissue from the other epithelia. We validated PITX2 at RNA and protein levels to investigate 10-year follow-up corneal samples derived from a COMET-treated aniridic patient. Moreover, we found novel angiogenesis-related factors that were differentially expressed in the three epithelia and instrumental in explaining the neovascularization in COMET-treated patients. These results will support the follow-up analysis of patients transplanted with oral mucosa and provide new tools to understand the regeneration mechanism of transplanted corneas.
Collapse
Affiliation(s)
- Eustachio Attico
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Galaverni
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Andrea Torello
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
- Holostem Terapie Avanzate s.r.l., 41125 Modena, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Susanna Bonacorsi
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Lorena Losi
- Unit of Pathology, Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | | | - Paolo Rama
- SC Ophathalmology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Graziella Pellegrini
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
- Holostem Terapie Avanzate s.r.l., 41125 Modena, Italy
| |
Collapse
|
5
|
Abstract
Corneal endothelium is the innermost layer of the cornea which has both barrier and pump function and very important to maintain cornea clarity. Unlike epithelium, endothelium does not have regenerative potential; hence, endothelial damage or dysfunction could lead to corneal edema and visual impairment. Advanced corneal transplantation which involves selective replacement of dysfunctional endothelium has led to improved and faster visual rehabilitation. But in recent times, alternative therapies in the management of corneal edema and endothelial diseases have been reported. In this review, we aim to give a comprehensive review of various strategies for the management of corneal endothelial dysfunction in order to give treatment which is precisely tailored for each individual patient. A review of all peer-reviewed publications on novel strategies for the management of endothelial dysfunction was performed. The various approaches to the management of endothelial dysfunction are compared and discussed. Shortage of human donor corneas globally is fuelling the search for keratoplasty alternatives. Corneal endothelial dysfunction can be caused following surgery, laser or corneal endothelial dystrophies which could be amenable to treatment with pharmacological, biological intervention and reverse the endothelial dysfunction in the early stages of endothelial failure. Pharmacological and surgical intervention are helpful in cases of good peripheral endothelial cell reserve, and advanced cases of endothelial cell dysfunction can be targeted with cell culture therapies, gene therapy and artificial implant. Treatment strategies which target endothelial dysfunction, especially FECD in its early stages, and gene therapy are rapidly evolving. Therapies which delay endothelial keratoplasty also are evolving like DSO and need more studies of long-term follow-up and patient selection criteria.
Collapse
Affiliation(s)
- Shalini Singh
- Academy of Eye Care Education, L V Prasad Eye Institute, Hyderabad, India.,The Cornea Institute, L V Prasad Eye Institute, Hyderabad, India
| | - Sunita Chaurasia
- The Cornea Institute, L V Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
6
|
Kopecny LR, Lee BWH, Coroneo MT. A systematic review on the effects of ROCK inhibitors on proliferation and/or differentiation in human somatic stem cells: A hypothesis that ROCK inhibitors support corneal endothelial healing via acting on the limbal stem cell niche. Ocul Surf 2023; 27:16-29. [PMID: 36586668 DOI: 10.1016/j.jtos.2022.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Rho kinase inhibitors (ROCKi) have attracted growing multidisciplinary interest, particularly in Ophthalmology where the question as to how they promote corneal endothelial healing remains unresolved. Concurrently, stem cell biology has rapidly progressed in unravelling drivers of stem cell (SC) proliferation and differentiation, where mechanical niche factors and the actin cytoskeleton are increasingly recognized as key players. There is mounting evidence from the study of the peripheral corneal endothelium that supports the likelihood of an internal limbal stem cell niche. The possibility that ROCKi stimulate the endothelial SC niche has not been addressed. Furthermore, there is currently a paucity of data that directly evaluates whether ROCKi promotes corneal endothelial healing by acting on this limbal SC niche located near the transition zone. Therefore, we performed a systematic review examining the effects ROCKi on the proliferation and differentiation of human somatic SC, to provide insight into its effects on various human SC populations. An appraisal of electronic searches of four databases identified 1 in vivo and 58 in vitro studies (36 evaluated proliferation while 53 examined differentiation). Types of SC studied included mesenchymal (n = 32), epithelial (n = 11), epidermal (n = 8), hematopoietic and other (n = 8). The ROCK 1/2 selective inhibitor Y-27632 was used in almost all studies (n = 58), while several studies evaluated ≥2 ROCKi (n = 4) including fasudil, H-1152, and KD025. ROCKi significantly influenced human somatic SC proliferation in 81% of studies (29/36) and SC differentiation in 94% of studies (50/53). The present systemic review highlights that ROCKi are influential in regulating human SC proliferation and differentiation, and provides evidence to support the hypothesis that ROCKi promotes corneal endothelial division and maintenance via acting on the inner limbal SC niche.
Collapse
Affiliation(s)
- Lloyd R Kopecny
- School of Clinical Medicine, University of New South Wales, Sydney, Australia.
| | - Brendon W H Lee
- Department of Ophthalmology, School of Clinical Medicine, University of New South Wales, Level 2 South Wing, Edmund Blacket Building, Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Minas T Coroneo
- Department of Ophthalmology, Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
7
|
Cvekl A, Camerino MJ. Generation of Lens Progenitor Cells and Lentoid Bodies from Pluripotent Stem Cells: Novel Tools for Human Lens Development and Ocular Disease Etiology. Cells 2022; 11:3516. [PMID: 36359912 PMCID: PMC9658148 DOI: 10.3390/cells11213516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
In vitro differentiation of human pluripotent stem cells (hPSCs) into specialized tissues and organs represents a powerful approach to gain insight into those cellular and molecular mechanisms regulating human development. Although normal embryonic eye development is a complex process, generation of ocular organoids and specific ocular tissues from pluripotent stem cells has provided invaluable insights into the formation of lineage-committed progenitor cell populations, signal transduction pathways, and self-organization principles. This review provides a comprehensive summary of recent advances in generation of adenohypophyseal, olfactory, and lens placodes, lens progenitor cells and three-dimensional (3D) primitive lenses, "lentoid bodies", and "micro-lenses". These cells are produced alone or "community-grown" with other ocular tissues. Lentoid bodies/micro-lenses generated from human patients carrying mutations in crystallin genes demonstrate proof-of-principle that these cells are suitable for mechanistic studies of cataractogenesis. Taken together, current and emerging advanced in vitro differentiation methods pave the road to understand molecular mechanisms of cataract formation caused by the entire spectrum of mutations in DNA-binding regulatory genes, such as PAX6, SOX2, FOXE3, MAF, PITX3, and HSF4, individual crystallins, and other genes such as BFSP1, BFSP2, EPHA2, GJA3, GJA8, LIM2, MIP, and TDRD7 represented in human cataract patients.
Collapse
Affiliation(s)
- Aleš Cvekl
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michael John Camerino
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
8
|
Català P, Thuret G, Skottman H, Mehta JS, Parekh M, Ní Dhubhghaill S, Collin RWJ, Nuijts RMMA, Ferrari S, LaPointe VLS, Dickman MM. Approaches for corneal endothelium regenerative medicine. Prog Retin Eye Res 2021; 87:100987. [PMID: 34237411 DOI: 10.1016/j.preteyeres.2021.100987] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
The state of the art therapy for treating corneal endothelial disease is transplantation. Advances in the reproducibility and accessibility of surgical techniques are increasing the number of corneal transplants, thereby causing a global deficit of donor corneas and leaving 12.7 million patients with addressable visual impairment. Approaches to regenerate the corneal endothelium offer a solution to the current tissue scarcity and a treatment to those in need. Methods for generating corneal endothelial cells into numbers that could address the current tissue shortage and the possible strategies used to deliver them have now become a therapeutic reality with clinical trials taking place in Japan, Singapore and Mexico. Nevertheless, there is still a long way before such therapies are approved by regulatory bodies and become clinical practice. Moreover, acellular corneal endothelial graft equivalents and certain drugs could provide a treatment option for specific disease conditions without the need of donor tissue or cells. Finally, with the emergence of gene modulation therapies to treat corneal endothelial disease, it would be possible to treat presymptomatic patients or those presenting early symptoms, drastically reducing the need for donor tissue. It is necessary to understand the most recent developments in this rapidly evolving field to know which conditions could be treated with which approach. This article provides an overview of the current and developing regenerative medicine therapies to treat corneal endothelial disease and provides the necessary guidance and understanding towards the treatment of corneal endothelial disease.
Collapse
Affiliation(s)
- Pere Català
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Gilles Thuret
- Laboratory of Biology, Engineering and Imaging of Corneal Graft, BiiGC, Faculty of Medicine, University of Saint Etienne, Saint Etienne, France; Institut Universitaire de France, Paris, France
| | - Heli Skottman
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jodhbir S Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore; Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-National University Singapore Medical School, Singapore; Singapore National Eye Centre, Singapore
| | - Mohit Parekh
- Institute of Ophthalmology, University College London, London, UK; The Veneto Eye Bank Foundation, Venice, Italy; Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sorcha Ní Dhubhghaill
- Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium; Ophthalmology, Visual Optics and Visual Rehabilitation, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium
| | - Rob W J Collin
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rudy M M A Nuijts
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | | | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Mor M Dickman
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
9
|
Directed Differentiation of Human Pluripotent Stem Cells towards Corneal Endothelial-Like Cells under Defined Conditions. Cells 2021; 10:cells10020331. [PMID: 33562615 PMCID: PMC7915025 DOI: 10.3390/cells10020331] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
The most crucial function of corneal endothelial cells (CEnCs) is to maintain optical transparency by transporting excess fluid out of stroma. Unfortunately, CEnCs are not able to proliferate in vivo in the case of trauma or dystrophy. Visually impaired patients with corneal endothelial deficiencies that are waiting for transplantation due to massive global shortage of cadaveric corneal transplants are in a great need of help. In this study, our goal was to develop a defined, clinically applicable protocol for direct differentiation of CEnCs from human pluripotent stem cells (hPSCs). To produce feeder-free hPSC-CEnCs, we used small molecule induction with transforming growth factor (TGF) beta receptor inhibitor SB431542, GSK-3-specific inhibitor CHIR99021 and retinoic acid to guide differentiation through the neural crest and periocular mesenchyme (POM). Cells were characterized by the morphology and expression of human (h)CEnC markers with immunocytochemistry and RT-qPCR. After one week of induction, we observed the upregulation of POM markers paired-like homeodomain transcription factor 2 (PITX2) and Forkhead box C1 (FOXC1) and polygonal-shaped cells expressing CEnC-associated markers Zona Occludens-1 (ZO-1), sodium-potassium (Na+/K+)-ATPase, CD166, sodium bicarbonate cotransporter 1 (SLC4A4), aquaporin 1 (AQP1) and N-cadherin (NCAD). Furthermore, we showed that retinoic acid induced a dome formation in the cell culture, with a possible indication of fluid transport by the differentiated cells. Thus, we successfully generated CEnC-like cells from hPSCs with a defined, simple and fast differentiation method.
Collapse
|
10
|
Ting DSJ, Peh GSL, Adnan K, Mehta JS. Translational and Regulatory Challenges of Corneal Endothelial Cell Therapy: A Global Perspective. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:52-62. [PMID: 33267724 DOI: 10.1089/ten.teb.2020.0319] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell therapies are emerging as a unique class of clinical therapeutics in medicine. In 2015, Holoclar (ex vivo expanded autologous human corneal epithelial cells containing stem cells) gained the regulatory approval for treating limbal stem cell deficiency after chemical eye burn. This has set a precedent in ophthalmology and in medicine, reinforcing the therapeutic promise of cell therapy. However, to generalize and commercialize cell therapies on a global scale, stringent translational and regulatory requirements need to be fulfilled at both local and international levels. Over the past decade, the Singapore group has taken significant steps in developing human corneal endothelial cell (HCEnC) therapy for treating corneal endothelial diseases, which are currently the leading indication for corneal transplantation in many countries. Successful development of HCEnC therapy may serve as a novel solution to the current global shortage of donor corneas. Based on the experience in Singapore, this review aims to provide a global perspective on the translational and regulatory challenges for bench-to-bedside translation of cell therapy. Specifically, we discussed about the characterization of the critical quality attributes (CQA), the challenges that can affect the CQA, and the variations in the regulatory framework embedded within different regions, including Singapore, Europe, and the United States. Impact statement Functional corneal endothelium is critical to normal vision. Corneal endothelial disease-secondary to trauma, surgery, or pathology-represents an important cause of visual impairment and blindness in both developed and developing countries. Currently, corneal transplantation serves as the current gold standard for treating visually significant corneal endothelial diseases, although limited by the shortage of donor corneas. Over the past decade, human corneal endothelial cell therapy has emerged as a promising treatment option for treating corneal endothelial diseases. To allow widespread application of this therapy, significant regulatory challenges will need to be systematically overcome.
Collapse
Affiliation(s)
- Darren Shu Jeng Ting
- Academic Ophthalmology, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom.,Department of Ophthalmology, Queen's Medical Centre, Nottingham, United Kingdom.,Singapore Eye Research Institute, Singapore, Singapore
| | - Gary S L Peh
- Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | - Jodhbir S Mehta
- Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore.,Schools of Material Science and Engineering, Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
11
|
Regenerative capacity of the corneal transition zone for endothelial cell therapy. Stem Cell Res Ther 2020; 11:523. [PMID: 33276809 PMCID: PMC7716425 DOI: 10.1186/s13287-020-02046-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
The corneal endothelium located on the posterior corneal surface is responsible for regulating stromal hydration. This is contributed by a monolayer of corneal endothelial cells (CECs), which are metabolically active in a continuous fluid-coupled efflux of ions from the corneal stroma into the aqueous humor, preventing stromal over-hydration and preserving the orderly arrangement of stromal collagen fibrils, which is essential for corneal transparency. Mature CECs do not have regenerative capacity and cell loss due to aging and diseases results in irreversible stromal edema and a loss of corneal clarity. The current gold standard of treatment for this worldwide blindness caused by corneal endothelial failure is the corneal transplantation using cadaveric donor corneas. The top indication is Fuchs corneal endothelial dystrophy/degeneration, which represents 39% of all corneal transplants performed. However, the global shortage of transplantable donor corneas has restricted the treatment outcomes, hence instigating a need to research for alternative therapies. One such avenue is the CEC regeneration from endothelial progenitors, which have been identified in the peripheral endothelium and the adjacent transition zone. This review examines the evidence supporting the existence of endothelial progenitors in the posterior limbus and summarizes the existing knowledge on the microanatomy of the transitional zone. We give an overview of the isolation and ex vivo propagation of human endothelial progenitors in the transition zone, and their growth and differentiation capacity to the corneal endothelium. Transplanting these bioengineered constructs into in vivo models of corneal endothelial degeneration will prove the efficacy and viability, and the long-term maintenance of functional endothelium. This will develop a novel regenerative therapy for the management of corneal endothelial diseases.
Collapse
|
12
|
Price MO, Mehta JS, Jurkunas UV, Price FW. Corneal endothelial dysfunction: Evolving understanding and treatment options. Prog Retin Eye Res 2020; 82:100904. [PMID: 32977001 DOI: 10.1016/j.preteyeres.2020.100904] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022]
Abstract
The cornea is exquisitely designed to protect the eye while transmitting and focusing incoming light. Precise control of corneal hydration by the endothelial cell layer that lines the inner surface of the cornea is required for optimal transparency, and endothelial dysfunction or damage can result in corneal edema and visual impairment. Advances in corneal transplantation now allow selective replacement of dysfunctional corneal endothelium, providing rapid visual rehabilitation. A series of technique improvements have minimized complications and various adaptations allow use even in eyes with complicated anatomy. While selective endothelial keratoplasty sets a very high standard for safety and efficacy, a shortage of donor corneas in many parts of the world restricts access, prompting a search for alternatives. Clinical trials are underway to evaluate the potential for self-recovery after removal of dysfunctional central endothelium in patients with healthy peripheral endothelium. Various approaches to using cultured human corneal endothelial cells are also in clinical trials; these aim to multiply cells from a single donor cornea for use in potentially hundreds of patients. Pre-clinical studies are underway with induced pluripotent stem cells, endothelial stem cell regeneration, gene therapy, anti-sense oligonucleotides, and various biologic/pharmacologic approaches designed to treat, prevent, or retard corneal endothelial dysfunction. The availability of more therapeutic options will hopefully expand access around the world while also allowing treatment to be more precisely tailored to each individual patient.
Collapse
Affiliation(s)
- Marianne O Price
- Cornea Research Foundation of America, 9002 N. Meridian St., Suite 212, Indianapolis, IN, USA.
| | - Jodhbir S Mehta
- Singapore National Eye Centre, 11 Third Hospital Ave #08-00, 168751, Singapore
| | - Ula V Jurkunas
- Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA, USA
| | - Francis W Price
- Price Vision Group, 9002 N. Meridian St., Suite 100, Indianapolis, IN, USA
| |
Collapse
|
13
|
Okubo T, Hayashi R, Shibata S, Kudo Y, Ishikawa Y, Inoue S, Kobayashi Y, Honda A, Honma Y, Kawasaki S, Nishida K. Generation and validation of a PITX2-EGFP reporter line of human induced pluripotent stem cells enables isolation of periocular mesenchymal cells. J Biol Chem 2020; 295:3456-3465. [PMID: 32034090 PMCID: PMC7076207 DOI: 10.1074/jbc.ra119.010713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/05/2020] [Indexed: 11/06/2022] Open
Abstract
PITX2 (Paired-like homeodomain transcription factor 2) plays important roles in asymmetric development of the internal organs and symmetric development of eye tissues. During eye development, cranial neural crest cells migrate from the neural tube and form the periocular mesenchyme (POM). POM cells differentiate into several ocular cell types, such as corneal endothelial cells, keratocytes, and some ocular mesenchymal cells. In this study, we used transcription activator-like effector nuclease technology to establish a human induced pluripotent stem cell (hiPSC) line expressing a fluorescent reporter gene from the PITX2 promoter. Using homologous recombination, we heterozygously inserted a PITX2-IRES2-EGFP sequence downstream of the stop codon in exon 8 of PITX2 Cellular pluripotency was monitored with alkaline phosphatase and immunofluorescence staining of pluripotency markers, and the hiPSC line formed normal self-formed ectodermal autonomous multizones. Using a combination of previously reported methods, we induced PITX2 in the hiPSC line and observed simultaneous EGFP and PITX2 expression, as indicated by immunoblotting and immunofluorescence staining. PITX2 mRNA levels were increased in EGFP-positive cells, which were collected by cell sorting, and marker gene expression analysis of EGFP-positive cells induced in self-formed ectodermal autonomous multizones revealed that they were genuine POM cells. Moreover, after 2 days of culture, EGFP-positive cells expressed the PITX2 protein, which co-localized with forkhead box C1 (FOXC1) protein in the nucleus. We anticipate that the PITX2-EGFP hiPSC reporter cell line established and validated here can be utilized to isolate POM cells and to analyze PITX2 expression during POM cell induction.
Collapse
Affiliation(s)
- Toru Okubo
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka 544-8666, Japan
| | - Ryuhei Hayashi
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| | - Shun Shibata
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka 544-8666, Japan
| | - Yuji Kudo
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka 544-8666, Japan
| | - Yuki Ishikawa
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Saki Inoue
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuki Kobayashi
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ai Honda
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yoichi Honma
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka 544-8666, Japan
| | - Satoshi Kawasaki
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
14
|
Zhang J, Patel DV, McGhee CNJ. The Rapid Transformation of Transplantation for Corneal Endothelial Diseases: An Evolution From Penetrating to Lamellar to Cellular Transplants. Asia Pac J Ophthalmol (Phila) 2019; 8:441-447. [PMID: 31789646 PMCID: PMC6903320 DOI: 10.1097/apo.0000000000000265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022] Open
Abstract
The cornea is the major focusing structure of the human eye and the corneal endothelium maintains the relatively dehydrated state of the cornea required for clarity. The endothelial cells respond to disease or injury by migration and cellular enlargement. Our current understanding is that there is a very limited degree of proliferative or regenerative capacity in the human corneal endothelium. Thus, corneal endothelial diseases may result in corneal edema, significantly impact vision and quality of life. Contemporary surgical transplantation options for treating moderate to advanced endothelial dysfunction include penetrating keratoplasty (PK), Descemet stripping endothelial keratoplasty (DSEK), and Descemet membrane endothelial keratoplasty. Advances in surgical techniques aim to bring faster visual recovery and improve visual outcomes; however, there is still a significant donor cornea shortage worldwide and alternative methods for treatment for corneal endothelial disease are rapidly evolving. Indeed, we are at a pivotal point in corneal transplantation for endothelial disease and novel surgical strategies include using 1 donor for multiple recipients, a minimally attached endothelial graft, and Descemet membrane stripping only. Crucially, forthcoming approaches include the use of Rho-Kinase (ROCK) inhibitors, endothelial cell therapy, tissue engineered grafts, and consideration of stem cell techniques. Ultimately, the choice of technique will be dependent on recipient factors such as age, type of endothelial disease, extent of the disease, and associated ocular disorders. The safety and efficacy of these rapidly developing treatments warrant further investigations. In time, some or all of these alternatives for corneal transplantation will alleviate the reliance on limited corneal donor tissue.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
15
|
Yam GHF, Seah X, Yusoff NZBM, Setiawan M, Wahlig S, Htoon HM, Peh GSL, Kocaba V, Mehta JS. Characterization of Human Transition Zone Reveals a Putative Progenitor-Enriched Niche of Corneal Endothelium. Cells 2019; 8:cells8101244. [PMID: 31614883 PMCID: PMC6829622 DOI: 10.3390/cells8101244] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
: The corneal endothelium regulates corneal hydration to maintain the transparency of cornea. Lacking regenerative capacity, corneal endothelial cell loss due to aging and diseases can lead to corneal edema and vision loss. There is limited information on the existence of corneal endothelial progenitors. We conducted ultrastructural examinations and expression analyses on the human transition zone (TZ) at the posterior limbus of corneal periphery, to elucidate if the TZ harbored progenitor-like cells, and to reveal their niche characteristics. Within the narrow TZ (~190 μm width), the inner TZ-adjacent to the peripheral endothelium (PE)-contained cells expressing stem/progenitor markers (Sox2, Lgr5, CD34, Pitx2, telomerase). They were located on the inner TZ surface and in its underlying stroma. Lgr5 positive cells projected as multicellular clusters into the PE. Under transmission electron microscopy and serial block face-scanning electron microscopy and three-dimensional (3D) reconstruction, the terminal margin of Descemet's membrane was inserted beneath the TZ surface, with the distance akin to the inner TZ breadth. Porcine TZ cells were isolated and proliferated into a confluent monolayer and differentiated to cells expressing corneal endothelial markers (ZO1, Na+K+ATPase) on cell surface. In conclusion, we have identified a novel inner TZ containing progenitor-like cells, which could serve the regenerative potential for corneal endothelium.
Collapse
Affiliation(s)
- Gary Hin-Fai Yam
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
- Eye-Academic Clinical Program, Duke-National University of Singapore (NUS) Graduate Medical School, Singapore 169857, Singapore.
| | - Xinyi Seah
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
| | | | - Melina Setiawan
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
| | - Stephen Wahlig
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27705, USA.
| | - Hla Myint Htoon
- Eye-Academic Clinical Program, Duke-National University of Singapore (NUS) Graduate Medical School, Singapore 169857, Singapore.
- Data Science Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
| | - Gary S L Peh
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
- Eye-Academic Clinical Program, Duke-National University of Singapore (NUS) Graduate Medical School, Singapore 169857, Singapore.
| | - Viridiana Kocaba
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
- Department of Ophthalmology, Claude Bernard Lyon 1 Université, 69622 Villeurbanne, France.
| | - Jodhbir S Mehta
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
- Eye-Academic Clinical Program, Duke-National University of Singapore (NUS) Graduate Medical School, Singapore 169857, Singapore.
- Singapore National Eye Centre, Singapore, Singapore 168751, Singapore.
- School of Material Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore.
| |
Collapse
|
16
|
The future of keratoplasty: cell-based therapy, regenerative medicine, bioengineering keratoplasty, gene therapy. Curr Opin Ophthalmol 2019; 30:286-291. [PMID: 31045881 DOI: 10.1097/icu.0000000000000573] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW To provide an update on the state of development of novel therapeutic modalities for the treatment of corneal diseases. RECENT FINDINGS Novel corneal therapeutics may be broadly classified as cell therapy, regenerative medicine, bioengineered corneal grafts and gene therapy. Cell therapy encompasses cultivation of cells, such as corneal endothelial cells (CECs) and keratocytes to replenish the depleted native cell population. Regenerative medicine is mainly applicable to the corneal endothelium, and is dependent on the ability of native, healthy CECs to restore the corneal endothelium following trauma or descemetorhexis; this approach may be effective for the treatment of Peter's anomaly and Fuchs endothelial corneal dystrophy (FECD). Bioengineered corneal grafts are synthetic constructs designed to replace cadaveric corneal grafts; tissue-engineered endothelial-keratoplasty grafts and bioengineered stromal grafts have been experimented in animal models with favourable results. Gene therapy with antisense oligonucleotide and CRISPR endonucleases, including deactivated Cas9, may potentially be used to treat FECD and TGFBI-related corneal dystrophies. SUMMARY These novel therapeutic modalities may potentially supersede keratoplasty as the standard of care in the future.
Collapse
|
17
|
Stage-dependent differential gene expression profiles of cranial neural crest-like cells derived from mouse-induced pluripotent stem cells. Med Mol Morphol 2019; 53:28-41. [PMID: 31297611 PMCID: PMC7033077 DOI: 10.1007/s00795-019-00229-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/26/2019] [Indexed: 12/13/2022]
Abstract
Cranial neural crest cells are multipotent cells that migrate into the pharyngeal arches of the vertebrate embryo and differentiate into various craniofacial organ derivatives. Therefore, migrating cranial neural crest cells are considered one of the most attractive candidate cell sources in regenerative medicine. We generated cranial neural crest like cell (cNCCs) using mouse-induced pluripotent stem cells cultured in neural crest-inducing medium for 14 days. Subsequently, we conducted RNA sequencing experiments to analyze gene expression profiles of cNCCs at different time points after induction. cNCCs expressed several neural crest specifier genes; however, some previously reported specifier genes such as paired box 3 and Forkhead box D3, which are essential for embryonic neural crest development, were not expressed. Moreover, ETS proto-oncogene 1, transcription factor and sex-determining region Y-box 10 were only expressed after 14 days of induction. Finally, cNCCs expressed multiple protocadherins and a disintegrin and metalloproteinase with thrombospondin motifs enzymes, which may be crucial for their migration.
Collapse
|
18
|
Wahlig S, Yam GHF, Chong W, Seah XY, Kocaba V, Ang M, Htoon HM, Tun TA, Ong HS, Mehta JS. Quantification of the Posterior Cornea Using Swept Source Optical Coherence Tomography. Transl Vis Sci Technol 2018; 7:2. [PMID: 30197834 PMCID: PMC6126962 DOI: 10.1167/tvst.7.5.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/15/2018] [Indexed: 12/13/2022] Open
Abstract
Purpose We define optical coherence tomography (OCT) measurement parameters of the corneal endothelium/Descemet's membrane (DM) complex and peripheral transition zone (TZ) and describe these measurements in an ethnically Chinese population. Methods OCT images of the anterior segment and iridocorneal angle were obtained from 129 healthy Chinese subjects (129 eyes), aged 40 to 81 years. The scleral spur (SS) and Schwalbe's line (SL) were identified in each image. Endothelium/DM diameter, referred to as endothelial arc length (EAL), is the SL-to-SL distance. The SS-to-SL distance encompasses the TZ and trabecular meshwork (TM). Since the TZ cannot be visualized by OCT, a ratio of TZ-to-TZ+TM width was calculated from scanning electron microscopy (SEM) images obtained from 5 cadaveric corneas. The SS-to-SL distance was multiplied by this ratio to approximate in vivo TZ width. Results From SEM measurements, the relationship TZ = 0.20*(TZ+TM) was determined. From OCT measurements, mean EAL was 12.15 ± 0.58 mm and mean TZ width was 156 ± 20 μm. For eyes with horizontal and vertical images, vertical EAL was significantly greater than horizontal EAL (P = 0.03). Conclusions Corneal endothelium/DM diameter and TZ width can be obtained from OCT images. Although only combined TZ+TM is visualized on OCT, TZ width can be reasonably approximated. Translational Relevance Emerging procedures, like endothelial cell injection and DM transplantation (DMT), require accurate measurements of endothelium/DM size for preoperative planning. Size of the TZ, which may contain progenitor cells, also could contribute to endothelial regeneration in these procedures.
Collapse
Affiliation(s)
- Stephen Wahlig
- Singapore Eye Research Institute (SERI), Singapore.,Duke University School of Medicine, Durham, NC, USA
| | - Gary Hin-Fai Yam
- Singapore Eye Research Institute (SERI), Singapore.,Singapore National Eye Center (SNEC), Singapore
| | | | - Xin-Yi Seah
- Singapore Eye Research Institute (SERI), Singapore
| | - Viridiana Kocaba
- Singapore Eye Research Institute (SERI), Singapore.,Department of Ophthalmology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, F-69000 Lyon, France; Université Lyon 1, F-69100 Villeurbanne, France
| | - Marcus Ang
- Singapore Eye Research Institute (SERI), Singapore.,Singapore National Eye Center (SNEC), Singapore.,Eye-ACP, Duke-NUS Graduate Medical School, Singapore.,Moorfields Eye Hospital, London, UK
| | - Hla Myint Htoon
- Singapore Eye Research Institute (SERI), Singapore.,Eye-ACP, Duke-NUS Graduate Medical School, Singapore
| | - Tin A Tun
- Singapore Eye Research Institute (SERI), Singapore.,Singapore National Eye Center (SNEC), Singapore
| | - Hon Shing Ong
- Singapore Eye Research Institute (SERI), Singapore.,Singapore National Eye Center (SNEC), Singapore.,Moorfields Eye Hospital, London, UK
| | - Jodhbir S Mehta
- Singapore Eye Research Institute (SERI), Singapore.,Singapore National Eye Center (SNEC), Singapore.,Eye-ACP, Duke-NUS Graduate Medical School, Singapore
| |
Collapse
|
19
|
Yamashita K, Inagaki E, Hatou S, Higa K, Ogawa A, Miyashita H, Tsubota K, Shimmura S. Corneal Endothelial Regeneration Using Mesenchymal Stem Cells Derived from Human Umbilical Cord. Stem Cells Dev 2018; 27:1097-1108. [PMID: 29929442 DOI: 10.1089/scd.2017.0297] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Corneal blindness is the third leading cause of blindness in the world, and one of the main etiologies is dysfunction of the corneal endothelium. Current treatment of corneal endothelial disease is allogenic corneal transplantation, which is limited by the global shortage of donor corneas and immunological rejection. The corneal endothelium consists of a monolayer of cells derived from the neural crest and mesoderm. Its main function is to prevent corneal edema by tight junctions formed by zonular occludens-1 (ZO-1) and Na, K-ATPase pump function. The human umbilical cord (UC) is a rich source of mesenchymal stem cells (MSCs). UC-MSCs that have multi-lineage potential may be an accessible allogenic source. After inducing differentiation with medium containing glycogen synthase kinase (GSK) 3-β inhibitor, UC-MSCs formed polygonal corneal endothelial-like cells that functioned as tissue-engineered corneal endothelium (UTECE). Expressions of major corneal endothelial markers were confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and quantitative RT-PCR (qRT-PCR). Western blotting confirmed the expression of Na,K-ATPase and PITX2, the functional and developmental markers of corneal endothelial cells. Immunohistochemistry revealed the localization of Na,K-ATPase and ZO-1 in cell-cell junctions, suggesting the presence of tight junctions. In vitro functional analysis revealed that UTECE had significantly high pump function compared with UC-MSCs. Moreover, UTECE transplanted into a rabbit model of bullous keratopathy successfully maintained corneal thickness and transparency. Our findings suggest that UTECE may be used as a source of allogenic cells for the treatment of corneal endothelial disease.
Collapse
Affiliation(s)
- Kazuya Yamashita
- 1 Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Emi Inagaki
- 1 Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Shin Hatou
- 1 Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Kazunari Higa
- 2 Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital , Ichikawa, Japan
| | - Akiko Ogawa
- 1 Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Hideyuki Miyashita
- 1 Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Kazuo Tsubota
- 1 Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Shigeto Shimmura
- 1 Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| |
Collapse
|