1
|
Enkhbat M, Mehta JS, Peh GSL, Yim EKF. Biomaterial-based strategies for primary human corneal endothelial cells for therapeutic applications: from cell expansion to transplantable carrier. Biomater Sci 2025; 13:1114-1130. [PMID: 39831824 DOI: 10.1039/d4bm00941j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The treatment of corneal blindness due to corneal diseases and injuries often requires the transplantation of healthy cadaveric corneal endothelial graft tissue to restore corneal clarity and visual function. However, the limited availability of donor corneas poses a significant challenge in meeting the demand for corneal transplantation. As a result, there is a growing interest in developing strategies alleviate this unmet need, and one of the postulated approaches is to isolate and expand primary human corneal endothelial cells (HCECs) in vitro for use in cell therapy. This review summarizes the recent advancements in the expansion of HCECs using biomaterials. Two principal biomaterial-based approaches, including extracellular matrix (ECM) coating and functionalized synthetic polymers, have been investigated to create an optimal microenvironment for the expansion and maintenance of corneal endothelial cells (CECs). This review highlights the challenges and opportunities in expanding primary HCECs using biomaterials. It emphasizes the importance of optimizing biomaterial properties, cell culture conditions, and the roles of biophysical cues to achieve efficient expansion and functional maintenance of CECs. Biomaterial-based strategies hold significant promise for expanding primary HCECs and improving the outcomes of CEC transplantation. The integration of biomaterials as cell culture substrates and transplantable carriers offers a comprehensive approach to address the limitations associated with current corneal tissue engineering techniques.
Collapse
Affiliation(s)
- Myagmartsend Enkhbat
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada.
| | - Jodhbir S Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
- Corneal & External Eye Disease Department, Singapore National Eye Centre, Singapore 168751, Singapore
- Singhealth Duke-NUS Ophthalmology & Visual Sciences Academic Clinical Programme, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Gary S L Peh
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore.
- Singhealth Duke-NUS Ophthalmology & Visual Sciences Academic Clinical Programme, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada.
- Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
- Center for Biotechnology and Bioengineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
2
|
De Miguel MP, Cadenas-Martin M, Stokking M, Martin-Gonzalez AI. Biomedical Application of MSCs in Corneal Regeneration and Repair. Int J Mol Sci 2025; 26:695. [PMID: 39859409 PMCID: PMC11766311 DOI: 10.3390/ijms26020695] [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: 11/29/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
The World Health Organization estimates that approximately 285 million people suffer from visual impairments, around 5% of which are caused by corneal pathologies. Currently, the most common clinical treatment consists of a corneal transplant (keratoplasty) from a human donor. However, worldwide demand for donor corneas amply exceeds the available supply. Lamellar keratoplasty (transplantation replacement of only one of the three layers of the cornea) is partially solving the problem of cornea undersupply. Obviously, cell therapy applied to every one of these layers will expand current therapeutic options, reducing the cost of ophthalmological interventions and increasing the effectiveness of surgery. Mesenchymal stem cells (MSCs) are adult stem cells with the capacity for self-renewal and differentiation into different cell lineages. They can be obtained from many human tissues, such as bone marrow, umbilical cord, adipose tissue, dental pulp, skin, and cornea. Their ease of collection and advantages over embryonic stem cells or induced pluripotent stem cells make them a very practical source for experimental and potential clinical applications. In this review, we focus on recent advances using MSCs from different sources to replace the damaged cells of the three corneal layers, at both the preclinical and clinical levels for specific corneal diseases.
Collapse
Affiliation(s)
- Maria P. De Miguel
- Cell Engineering Laboratory, La Paz University Hospital Health Research Institute, IdiPAZ, 28046 Madrid, Spain; (M.C.-M.); (M.S.); (A.I.M.-G.)
| | | | | | | |
Collapse
|
3
|
Chi J, Wang S, Ju R, Li S, Liu C, Zou M, Xu T, Wang Y, Jiang Z, Yang C, Han B. Repair effects of thermosensitive hydrogels combined with iPSC-derived corneal endothelial cells on rabbit corneal endothelial dysfunction. Acta Biomater 2025; 191:216-232. [PMID: 39551331 DOI: 10.1016/j.actbio.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/22/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Considering the limitations of human corneal endothelial cell proliferation as well as the severe shortage of corneal donations, it is imperative to develop improved methods of corneal endothelial cell transplantation. The purpose of this study was to construct a modified corneal endothelial cell transplantation approach using thermosensitive hydrogels combined with induced pluripotent stem cells (iPSCs)-derived human corneal endothelial cells (hCECs). In this study, thermosensitive hydrogels hydroxypropyl chitin/carboxymethyl chitosan (HPCH/CMCS) were fabricated, and their hydrogels properties and biocompatibility were investigated. Our results demonstrated that HPCH/CMCS hydrogels exhibited superior transparency, appropriate mechanical properties and favorable biocompatibility. A two-step induction method of small molecule compounds was employed, by which iPSCs were differentiated into hCECs via neural crest cells (NCCs). Additionally, a rabbit corneal endothelial dysfunction model was established in vivo, aiming to evaluate the safety and effectiveness of the combined method. Slit lamp microscope results indicated that significant transparency improvement could be noted in HPCH/CMCS/hCECs group (P = 0.006), whereas the corneal transparency was not homogeneous in different areas. Moreover, histological examinations and immunofluorescence analysis revealed that HPCH/CMCS/hCECs group showed a higher density of corneal endothelial cells and positive expressions of related markers. This study may provide ideas and experimental basis for the combined application of hydrogels and iPSC-derived corneal endothelial cells for corneal endothelial dysfunction. STATEMENT OF SIGNIFICANCE: Corneal transplantation is the most effective treatment for corneal endothelial dysfunction, which is challenged by issues such as corneal donor shortages and immune rejection. In this study, we proposed a combined transplantation method of cells and hydrogels for corneal endothelial dysfunction. We modified the protocols to obtain corneal endothelial cells from iPSCs by a two-step induction method. Besides, thermosensitive hydrogels with satisfactory biocompatibility and degradability were fabricated as fixation and support carriers of iPSC-derived corneal endothelial cells for in vivo transplantation. Experimental results demonstrated that this method could locally repair corneal endothelial dysfunction in rabbits, with the repaired corneas expressing relevant markers. This study presented a preliminary attempt to combine hydrogels and cells for corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Jinhua Chi
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shuo Wang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Ruibao Ju
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shanshan Li
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Chenqi Liu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Mingyu Zou
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Tianjiao Xu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yanting Wang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Zhiwen Jiang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Chaozhong Yang
- School of Medicine, Heze Medical College, Heze 274046, China
| | - Baoqin Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
4
|
Mommaerts K, Okawa S, Schmitt M, Kofanova O, Turner TR, Ben RN, Del Sol A, Mathieson W, Schwamborn JC, Acker JP, Betsou F. Ice recrystallization inhibitors enable efficient cryopreservation of induced pluripotent stem cells: A functional and transcriptomic analysis. Stem Cell Res 2024; 81:103583. [PMID: 39467374 DOI: 10.1016/j.scr.2024.103583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/28/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
The successful use of human induced pluripotent stem cells (iPSCs) for research or clinical applications requires the development of robust, efficient, and reproducible cryopreservation protocols. After cryopreservation, the survival rate of iPSCs is suboptimal and cell line-dependent. We assessed the use of ice recrystallization inhibitors (IRIs) for cryopreservation of human iPSCs. A toxicity screening study was performed to assess specific small-molecule carbohydrate-based IRIs and concentrations for further evaluation. Then, a cryopreservation study compared the cryoprotective efficiency of 15 mM IRIs in 5 % or 10 % DMSO-containing solutions and with CryoStor® CS10. Three iPSC lines were cryopreserved as single-cell suspensions in the cryopreservation solutions and post-thaw characteristics, including pluripotency and differential gene expression were assessed. We demonstrate the fitness-for-purpose of 15 mM IRI in 5 % DMSO as an efficient cryoprotective solution for iPSCs in terms of post-thaw recovery, viability, pluripotency, and transcriptomic changes. This mRNA sequencing dataset has the potential to be used for molecular mechanism analysis relating to cryopreservation. Use of IRIs can reduce DMSO concentrations and its associated toxicities, thereby improving the utility, effectiveness, and efficiency of cryopreservation.
Collapse
Affiliation(s)
- Kathleen Mommaerts
- Integrated Biobank of Luxembourg, Luxembourg Institute of Health, 1 rue Louis Rech, L-3555 Dudelange, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2 avenue de Université, L-4365 Esch-sur-Alzette, Luxembourg.
| | - Satoshi Okawa
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2 avenue de Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Margaux Schmitt
- Integrated Biobank of Luxembourg, Luxembourg Institute of Health, 1 rue Louis Rech, L-3555 Dudelange, Luxembourg
| | - Olga Kofanova
- Integrated Biobank of Luxembourg, Luxembourg Institute of Health, 1 rue Louis Rech, L-3555 Dudelange, Luxembourg
| | | | - Robert N Ben
- PanTHERA CryoSolutions Inc., Edmonton, Alberta, Canada; Department of Chemistry, University of Ottawa, Ottawa, Ontario, Canada
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2 avenue de Université, L-4365 Esch-sur-Alzette, Luxembourg; CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - William Mathieson
- Integrated Biobank of Luxembourg, Luxembourg Institute of Health, 1 rue Louis Rech, L-3555 Dudelange, Luxembourg
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2 avenue de Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Jason P Acker
- PanTHERA CryoSolutions Inc., Edmonton, Alberta, Canada; Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Fay Betsou
- Integrated Biobank of Luxembourg, Luxembourg Institute of Health, 1 rue Louis Rech, L-3555 Dudelange, Luxembourg
| |
Collapse
|
5
|
Takayanagi H, Hayashi R. Status and prospects for the development of regenerative therapies for corneal and ocular diseases. Regen Ther 2024; 26:819-825. [PMID: 39329098 PMCID: PMC11424903 DOI: 10.1016/j.reth.2024.09.001] [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: 06/24/2024] [Revised: 08/13/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Among the regenerative therapies being put into clinical use, the field of corneal regenerative therapy is one of the most advanced, with several regulatory approved products. This article describes the progress from initial development through to clinical application in the eye field, with a particular focus on therapies for corneal epithelial and endothelial diseases that have already been regulatory approved as regenerative therapy products. The applications of regenerative therapy to the corneal epithelium were attempted and confirmed earlier than other parts of the cornea, following advancements in basic research on corneal epithelial stem cells. Based on these advances, four regenerative therapy products for corneal epithelial disease, each employing distinct cell sources and culture techniques, have been commercialized since the regulatory approval of Holoclar® in Italy as a regenerative therapy product for corneal epithelial disease in 2015. Corneal endothelial regenerative therapy was started by the development of an in vitro method to expand corneal endothelial cells which do not proliferate in adults. The product was approved in Japan as Vyznova® in 2023. The development of regenerative therapies for retinal and ocular surface diseases is actively being pursued, and these therapies use somatic stem cells and pluripotent stem cells (PSCs), especially induced pluripotent stem cells (iPSCs). Accordingly, the eye field is anticipated to play a pioneering role in regenerative therapy development going forward.
Collapse
Affiliation(s)
- Hiroshi Takayanagi
- Research, Development and Production Department of RAYMEI Incorporated, Osaka, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryuhei Hayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
6
|
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
|
7
|
Dong C, Zou D, Duan H, Hu X, Zhou Q, Shi W, Li Z. Ex vivo cultivated retinal pigment epithelial cell transplantation for the treatment of rabbit corneal endothelial dysfunction. EYE AND VISION (LONDON, ENGLAND) 2023; 10:34. [PMID: 37528478 PMCID: PMC10394777 DOI: 10.1186/s40662-023-00351-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 07/04/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE Stem cell therapy is a promising strategy for the treatment of corneal endothelial dysfunction, and the need to find functional alternative seed cells of corneal endothelial cells (CECs) is urgent. Here, we determined the feasibility of using the retinal pigment epithelium (RPE) as an equivalent substitute for the treatment of corneal endothelial dysfunction. METHODS RPE cells and CECs in situ were obtained from healthy New Zealand male rabbits, and the similarities and differences between them were analyzed by electron microscopy, immunofluorescent staining, and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Rabbit primary RPE cells and CECs were isolated and cultivated ex vivo, and Na+/K+-ATPase activity and cellular permeability were detected at passage 2. The injection of cultivated rabbit primary RPE cells, CECs and human embryonic stem cell (hESC)-derived RPE cells was performed on rabbits with corneal endothelial dysfunction. Then, the therapeutic effects were evaluated by corneal transparency, central corneal thickness, enzyme linked immunosorbent assay (ELISA), qRT-PCR and immunofluorescent staining. RESULTS The rabbit RPE cells were similar in form to CECs in situ and ex vivo, showing a larger regular hexagonal shape and a lower cell density, with numerous tightly formed cell junctions and hemidesmosomes. Moreover, RPE cells presented a stronger barrier and ionic pumping capacity than CECs. When intracamerally injected into the rabbits, the transplanted primary RPE cells could dissolve corneal edema and decrease corneal thickness, with effects similar to those of CECs. In addition, the transplantation of hESC-derived RPE cells exhibited a similar therapeutic effect and restored corneal transparency and thickness within seven days. qRT-PCR results showed that the expressions of CEC markers, like CD200 and S100A4, increased, and the RPE markers OTX2, BEST1 and MITF significantly decreased in the transplanted RPE cells. Furthermore, we have demonstrated that rabbits transplanted with hESC-derived RPE cells maintained normal corneal thickness and exhibited slight pigmentation in the central cornea one month after surgery. Immunostaining results showed that the HuNu-positive transplanted cells survived and expressed ZO1, ATP1A1 and MITF. CONCLUSION RPE cells and CECs showed high structural and functional similarities in barrier and pump characteristics. Intracameral injection of primary RPE cells and hESC-derived RPE cells can effectively restore rabbit corneal clarity and thickness and maintain normal corneal function. This study is the first to report the effectiveness of RPE cells for corneal endothelial dysfunction, suggesting the feasibility of hESC-derived RPE cells as an equivalent substitute for CECs.
Collapse
Affiliation(s)
- Chunxiao Dong
- Department of Medicine, Qingdao University, Qingdao, 266071, China
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250000, China
- School of Ophthalmology, Shandong First Medical University, Jinan, 250000, China
| | - Dulei Zou
- Department of Medicine, Qingdao University, Qingdao, 266071, China
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250000, China
- School of Ophthalmology, Shandong First Medical University, Jinan, 250000, China
| | - Haoyun Duan
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Jinan, 250000, China
| | - Xiangyue Hu
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Jinan, 250000, China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- School of Ophthalmology, Shandong First Medical University, Jinan, 250000, China
| | - Weiyun Shi
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250000, China
- School of Ophthalmology, Shandong First Medical University, Jinan, 250000, China
| | - Zongyi Li
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China.
- School of Ophthalmology, Shandong First Medical University, Jinan, 250000, China.
| |
Collapse
|
8
|
Zhou Q, Li Z, Duan H. iPSC-Derived Corneal Endothelial Cells. Handb Exp Pharmacol 2023; 281:257-276. [PMID: 36882600 DOI: 10.1007/164_2023_644] [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: 03/09/2023]
Abstract
The corneal endothelium is the innermost monolayer of the cornea that maintains corneal transparency and thickness. However, adult human corneal endothelial cells (CECs) possess limited proliferative capacity, and injuries can only be repaired by migration and enlargement of resident cells. When corneal endothelial cell density is lower than the critical level (400-500 cells/mm2) due to disease or trauma, corneal endothelial dysfunction will occur and lead to corneal edema. Corneal transplantation remains the most effective clinical treatment therapy but is limited by the global shortage of healthy corneal donors. Recently, researchers have developed several alternative strategies for the treatment of corneal endothelial disease, including the transplantation of cultured human CECs and artificial corneal endothelial replacement. Early-stage results show that these strategies can effectively resolve corneal edema and restore corneal clarity and thickness, but the long-term efficacy and safety remain to be further validated. Induced pluripotent stem cells (iPSCs) represent an ideal cell source for the treatment and drug discovery of corneal endothelial diseases, which can avoid the ethical-related and immune-related problems of human embryonic stem cells (hESCs). At present, many approaches have been developed to induce the differentiation of corneal endothelial-like cells from human induced pluripotent stem cells (hiPSCs). Their safety and efficacy for the treatment of corneal endothelial dysfunction have been confirmed in rabbit and nonhuman primate animal models. Therefore, the iPSC-derived corneal endothelial cell model may provide a novel effective platform for basic and clinical research of disease modeling, drug screening, mechanistic investigation, and toxicology testing.
Collapse
Affiliation(s)
- Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Zongyi Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Haoyun Duan
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| |
Collapse
|
9
|
So S, Park Y, Kang SS, Han J, Sunwoo JH, Lee W, Kim J, Ye EA, Kim JY, Tchah H, Kang E, Lee H. Therapeutic Potency of Induced Pluripotent Stem-Cell-Derived Corneal Endothelial-like Cells for Corneal Endothelial Dysfunction. Int J Mol Sci 2022; 24:701. [PMID: 36614165 PMCID: PMC9821383 DOI: 10.3390/ijms24010701] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Corneal endothelial cells (CECs) do not proliferate or recover after illness or injury, resulting in decreased cell density and loss of pump/barrier function. Considering the shortage of donor cornea, it is vital to establish robust methods to generate CECs from induced pluripotent stem cells (iPSCs). We investigated the efficacy and safety of transplantation of iPSC-derived CECs into a corneal endothelial dysfunction (CED) rabbit model. iPSCs were generated from human fibroblasts. We characterized iPSCs by demonstrating the gene expression of the PSC markers OCT4, SOX2, TRA-1-60, and NANOG, teratoma formation, and differentiation into three germ layers. Differentiation of iPSCs into CECs was induced via neural crest cell (NCC) induction. CEC markers were detected using immunofluorescence and gene expression was analyzed using quantitative real-time PCR (qRT-PCR). After culturing iPSC-derived NCCs, we found the expression of zona occludens-1 (ZO-1) and Na+/K+ ATPase and a hexagonal morphology. ATP1A1, COL8A1, and AQP1 mRNA expression was higher in iPSC-derived CECs than in iPSCs and NCCs. We performed an injection of iPSC-derived CECs into the anterior chamber of a CED rabbit model and found improved levels of corneal transparency. We also found increased numbers of ZO-1- and ATP1A1-positive cells in rabbit corneas in the iPSC-derived CEC transplantation group. Usage of the coating material vitronectin (VTN) and fasudil resulted in good levels of CEC marker expression, demonstrated with Western blotting and immunocytochemistry. Combination of the VTN coating material and fasudil, instead of FNC mixture and Y27632, afforded the best results in terms of CEC differentiation's in vitro and in vivo efficacy. Successful transplantation of CEC-like cells into a CED animal model confirms the therapeutic efficacy of these cells, demonstrated by the restoration of corneal clarity. Our results suggest that iPSC-derived CECs can be a promising cellular resource for the treatment of CED.
Collapse
Affiliation(s)
- Seongjun So
- Department of Biomedical Science, CHA Advanced Research Institute, College of Life Science and Center for Embryo and Stem Cell Research, CHA University, Seongnam 13488, Republic of Korea
| | - Yoonkyung Park
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Soon Suk Kang
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jongsuk Han
- Department of Biomedical Science, CHA Advanced Research Institute, College of Life Science and Center for Embryo and Stem Cell Research, CHA University, Seongnam 13488, Republic of Korea
| | - Jeong Hye Sunwoo
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Whanseo Lee
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jin Kim
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Eun Ah Ye
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jae Yong Kim
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hungwon Tchah
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Eunju Kang
- Department of Biomedical Science, CHA Advanced Research Institute, College of Life Science and Center for Embryo and Stem Cell Research, CHA University, Seongnam 13488, Republic of Korea
| | - Hun Lee
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| |
Collapse
|
10
|
Serrano A, Osei KA, Huertas-Bello M, Sabater AL. The Potential of Stem Cells as Treatment for Ocular Surface Diseases. CURRENT OPHTHALMOLOGY REPORTS 2022. [DOI: 10.1007/s40135-022-00303-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
11
|
Corneal Regeneration Using Adipose-Derived Mesenchymal Stem Cells. Cells 2022; 11:cells11162549. [PMID: 36010626 PMCID: PMC9406486 DOI: 10.3390/cells11162549] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 11/28/2022] Open
Abstract
Adipose-derived stem cells are a subtype of mesenchymal stem cell that offers the important advantage of being easily obtained (in an autologous manner) from low invasive procedures, rendering a high number of multipotent stem cells with the potential to differentiate into several cellular lineages, to show immunomodulatory properties, and to promote tissue regeneration by a paracrine action through the secretion of extracellular vesicles containing trophic factors. This secretome is currently being investigated as a potential source for a cell-free based regenerative therapy for human tissues, which would significantly reduce the involved costs, risks and law regulations, allowing for a broader application in real clinical practice. In the current article, we will review the existing preclinical and human clinical evidence regarding the use of such adipose-derived mesenchymal stem cells for the regeneration of the three main layers of the human cornea: the epithelium (derived from the surface ectoderm), the stroma (derived from the neural crest mesenchyme), and the endothelium (derived from the neural crest cells).
Collapse
|
12
|
Induced Pluripotent Stem Cell-Derived Corneal Cells: Current Status and Application. Stem Cell Rev Rep 2022; 18:2817-2832. [PMID: 35913555 DOI: 10.1007/s12015-022-10435-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 10/16/2022]
Abstract
Deficiency and dysfunction of corneal cells leads to the blindness observed in corneal diseases such as limbal stem cell deficiency (LSCD) and bullous keratopathy. Regenerative cell therapies and engineered corneal tissue are promising treatments for these diseases [1]. However, these treatments are not yet clinically feasible due to inadequate cell sources. The discovery of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka has provided a multitude of opportunities in research because iPSCs can be generated from somatic cells, thus providing an autologous and unlimited source for corneal cells. Compared to other stem cell sources such as mesenchymal and embryonic, iPSCs have advantages in differentiation potential and ethical concerns, respectively. Efforts have been made to use iPSCs to model corneal disorders and diseases, drug testing [2], and regenerative medicine [1]. Autologous treatments based on iPSCs can be exorbitantly expensive and time-consuming, but development of stem cell banks with human leukocyte antigen (HLA)- homozygous cell lines can provide cost- and time-efficient allogeneic alternatives. In this review, we discuss the early development of the cornea because protocols differentiating iPSCs toward corneal lineages rely heavily upon recapitulating this development. Differentiation of iPSCs toward corneal cell phenotypes have been analyzed with an emphasis on feeder-free, xeno-free, and well-defined protocols, which have clinical relevance. The application, challenges, and potential of iPSCs in corneal research are also discussed with a focus on hurdles that prevent clinical translation.
Collapse
|
13
|
Chen J, Ou Q, Wang Z, Liu Y, Hu S, Liu Y, Tian H, Xu J, Gao F, Lu L, Jin C, Xu GT, Cui HP. Small-Molecule Induction Promotes Corneal Endothelial Cell Differentiation From Human iPS Cells. Front Bioeng Biotechnol 2021; 9:788987. [PMID: 34976977 PMCID: PMC8714889 DOI: 10.3389/fbioe.2021.788987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose: Corneal endothelial cells (CECs) serve as a barrier and foothold for the corneal stroma to maintain the function and transparency of the cornea. Loss of CECs during aging or disease states leads to blindness, and cell replacement therapy using either donated or artificially differentiated CECs remains the only curative approach. Methods: Human induced pluripotent stem cells (hiPSCs) that were cultured in chemically defined medium were induced with dual-SMAD inhibition to differentiate into neural crest cells (NCCs). A small-molecule library was screened to differentiate the NCCs into corneal endothelial-like cells. The characteristics of these cells were identified with real-time PCR and immunofluorescence. Western blotting was applied to detect the signaling pathways and key factors regulated by the small molecules. Results: We developed an effective protocol to differentiate hiPSCs into CECs with defined small molecules. The hiPSC-CECs were characterized by ZO-1, AQP1, Vimentin and Na+/K+-ATPase. Based on our small-molecule screen, we identified a small-molecule combination, A769662 and AT13148, that enabled the most efficient production of CECs. The combination of A769662 and AT13148 upregulated the PKA/AKT signaling pathway, FOXO1 and PITX2 to promote the conversion of NCCs to CECs. Conclusion: We established an efficient small molecule-based method to differentiate hiPSCs into corneal endothelial-like cells, which might facilitate drug discovery and the development of cell-based therapies for corneal diseases.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qingjian Ou
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhe Wang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yifan Liu
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuqin Hu
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yumeilan Liu
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haibin Tian
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingying Xu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Furong Gao
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lixia Lu
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Caixia Jin
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Guo-Tong Xu
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Hong-Ping Cui
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Improvement of an Effective Protocol for Directed Differentiation of Human Adipose Tissue-Derived Adult Mesenchymal Stem Cells to Corneal Endothelial Cells. Int J Mol Sci 2021; 22:ijms222111982. [PMID: 34769411 PMCID: PMC8585097 DOI: 10.3390/ijms222111982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Corneal disease affects 12.5 million individuals worldwide, with 2 million new cases each year. The standard treatment consists of a corneal transplantation from a human donor; however, the worldwide demand significantly exceeds the available supply. Lamellar endothelial keratoplasty, the replacement of only the endothelial layer of the cornea, can partially solve the problem. Progressive efforts have succeeded in expanding hCECs; however, the ability to expand hCECs is still limited, and new sources of CECs are being sought. Crucial advances have been achieved by the directed differentiation of embryonic or induced pluripotent stem cells, but these cells have disadvantages, such as the use of oncogenes, and are still difficult to establish. We aimed to transfer such knowledge to obtain hCECs from adipose tissue-derived adult mesenchymal stem cells (ADSC) by modifying four previously published procedures. We present several protocols capable of the directed differentiation of human ADSCs to hCECs. In our hands, the protocol by Ali et al. was the best adapted to such differentiation in terms of efficiency, time, and financial cost; however, the protocol by Wagoner et al. was the best for CEC marker expression. Our results broaden the type of cells of autologous extraocular origin that could be employed in the clinical setting for corneal endothelial deficiency.
Collapse
|
15
|
Ali M, Khan SY, Gottsch JD, Hutchinson EK, Khan A, Riazuddin SA. Pluripotent stem cell-derived corneal endothelial cells as an alternative to donor corneal endothelium in keratoplasty. Stem Cell Reports 2021; 16:2320-2335. [PMID: 34358452 PMCID: PMC8452521 DOI: 10.1016/j.stemcr.2021.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/07/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022] Open
Abstract
Here, we evaluate the efficacy of cryopreserved human embryonic stem cell (hESC)-derived corneal endothelial cells (CECs) to form a functional monolayer of corneal endothelium (CE) in rabbits and monkeys. We injected cryopreserved hESC-derived CECs into the anterior chamber of rabbits and monkeys either immediately after mechanical scraping of the central CE or a few days later when corneal edema developed. All preclinical models developed deturgesced and clear corneas following the injection of cryopreserved hESC-derived CECs and remained comparable to the corneas of the untreated eye. Confocal scanning microscopy confirmed an intact structure of hexagonal/polygonal cells and immunohistochemical analysis illustrated a monolayer expressing barrier and pump function proteins in the regenerated CE. The necropsy examination confirmed no remarkable change in multiple tissues assessed for teratoma formation. In conclusion, our data demonstrate the efficacy of cryopreserved hESC-derived CECs to form a functional CE on the denuded Descemet's membrane.
Collapse
Affiliation(s)
- Muhammad Ali
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shahid Y Khan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - John D Gottsch
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Eric K Hutchinson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - S Amer Riazuddin
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
16
|
Sun B, Bikkuzin T, Li X, Shi Y, Zhang H. Human-Induced Pluripotent Stem Cells-Derived Corneal Endothelial-Like Cells Promote Corneal Transparency in a Rabbit Model of Bullous Keratopathy. Stem Cells Dev 2021; 30:856-864. [PMID: 34128390 DOI: 10.1089/scd.2020.0205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The corneal endothelium (CE) is vital for the cornea to maintain its transparency. However, CE dysfunction occurs due to aging, intraocular surgery, trauma, dystrophy, etc. Corneal transplantation is the only method to clinically treat CE dysfunction; however, this treatment strategy faces the disadvantages of a global cornea shortage, graft failure, and severe side effects. There is a recognized need for a substitute for the CE. Stem cells are becoming increasingly common for the treatment of human diseases. In fact, several studies have documented the induction of corneal endothelial-like cells (CECs) from stem cells, but an ideal procedure has not yet been established. Thus, this study aimed at exploring a more efficient and robust differentiation method. We used a modified approach to differentiate induced pluripotent stem cells (iPSCs) into CECs. After the identification of differentiated CECs, the CECs were injected into the anterior chambers of the eyes of a rabbit model of bullous keratopathy. The rabbits were maintained in the eye-down position to ensure that the cells attached to the cornea. The results showed that corneal edema was alleviated in the rabbits injected with CECs compared with that in the rabbits belonging to the control group. This study extends the ability to differentiate iPSCs into CECs and provides a potential strategy for the treatment of reduced visual acuity caused by CE deficiency in the future.
Collapse
Affiliation(s)
- Baoqi Sun
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Timur Bikkuzin
- Department of Ophthalmology, Bashkir State Medical University, Ufa, Russia
| | - Xuran Li
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Shi
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Huang J, Xian B, Peng Y, Zeng B, Li W, Li Z, Xie Y, Zhao M, Zhang H, Zhou M, Yu H, Wu P, Liu X, Huang B. Migration of pre-induced human peripheral blood mononuclear cells from the transplanted to contralateral eye in mice. Stem Cell Res Ther 2021; 12:168. [PMID: 33691753 PMCID: PMC7945672 DOI: 10.1186/s13287-021-02180-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/20/2021] [Indexed: 11/26/2022] Open
Abstract
Background Retina diseases may lead to blindness as they often afflict both eyes. Stem cell transplantation into the affected eye(s) is a promising therapeutic strategy for certain retinal diseases. Human peripheral blood mononuclear cells (hPBMCs) are a good source of stem cells, but it is unclear whether pre-induced hPBMCs can migrate from the injected eye to the contralateral eye for bilateral treatment. We examine the possibility of bilateral cell transplantation from unilateral cell injection. Methods One hundred and sixty-one 3-month-old retinal degeneration 1 (rd1) mice were divided randomly into 3 groups: an untreated group (n = 45), a control group receiving serum-free Dulbecco’s modified Eagle’s medium (DMEM) injection into the right subretina (n = 45), and a treatment group receiving injection of pre-induced hPBMCs into the right subretina (n = 71). Both eyes were examined by full-field electroretinogram (ERG), immunofluorescence, flow cytometry, and quantitative real-time polymerase chain reaction (qRT-PCR) at 1 and 3 months post-injection. Results At both 1 and 3 months post-injection, labeled pre-induced hPBMCs were observed in the retinal inner nuclear layer of the contralateral (left untreated) eye as well as the treated eye as evidenced by immunofluorescence staining for a human antigen. Flow cytometry of fluorescently label cells and qRT-PCR of hPBMCs genes confirmed that transplanted hPBMCs migrated from the treated to the contralateral untreated eye and remained viable for up to 3 months. Further, full-field ERG showed clear light-evoked a and b waves in both treated and untreated eyes at 3 months post-transplantation. Labeled pre-induced hPBMCs were also observed in the contralateral optic nerve but not in the blood circulation, suggesting migration via the optic chiasm. Conclusion It may be possible to treat binocular eye diseases by unilateral stem cell injection. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02180-5.
Collapse
Affiliation(s)
- Jianfa Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Bikun Xian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.,The Second People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Yuting Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.,Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Baozhu Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Zhiquan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yaojue Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Hening Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Minyi Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Huan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Peixin Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Bing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
18
|
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
|
19
|
Faye PA, Poumeaud F, Chazelas P, Duchesne M, Rassat M, Miressi F, Lia AS, Sturtz F, Robert PY, Favreau F, Benayoun Y. Focus on cell therapy to treat corneal endothelial diseases. Exp Eye Res 2021; 204:108462. [PMID: 33493477 DOI: 10.1016/j.exer.2021.108462] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023]
Abstract
The cornea is a multi-layered structure which allows fine refraction and provides both resistance to external insults and adequate transparency. The corneal endothelium ensures stromal hydration, failure of which, such as in Fuchs endothelial corneal dystrophy, after trauma or in aging, may lead to loss of corneal transparency and induce blindness. Currently, no efficient therapeutic alternatives exist except for corneal grafting. Thus corneal tissue engineering represents a valuable alternative approach, which may overcome cornea donor shortage. Several studies describe protocols to isolate, differentiate, and cultivate corneal endothelial cells (CEnCs) in vitro. Two main in vitro strategies can be described: expansion of eye-native cell populations, such as CEnCs, or the production and expansion of CEnCs from non-eye native cell populations, such as induced Pluripotent Stem Cells (iPSCs). The challenge with these cells is to obtain a monolayer of CEnCs on a biocompatible carrier, with a specific morphology (flat hexagonal cells), and with specific functions such as programmed cell cycle arrest. Another issue for this cell culture methodology is to define the adapted protocol (media, trophic factors, timeframe) that can mimic physiological development. Additionally, contamination by other cell types still represents a huge problem. Thus, purification methods, such as Fluorescence Activated Cell Sorting (FACS), Magnetic Ativated Cell Sorting (MACS) or Sedimentation Field Flow Fractionation (SdFFF) are useful. Animal models are also crucial to provide a translational approach for these therapies, integrating macro- and microenvironment influences, systemic hormonal or immune responses, and exogenous interactions. Non-eye native cell graft protocols are constantly improving both in efficacy and safety, with the aim of being the most suitable candidate for corneal therapies in future routine practice. The aim of this work is to review these different aspects with a special focus on issues facing CEnC culture in vitro, and to highlight animal graft models adapted to screen the efficacy of these different protocols.
Collapse
Affiliation(s)
- Pierre Antoine Faye
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France.
| | - François Poumeaud
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Pauline Chazelas
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Mathilde Duchesne
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France; CHU de Limoges, Laboratoire de Neurologie, F-87000, Limoges, France; CHU de Limoges, Service d'Anatomie Pathologique, F-87000, Limoges, France
| | - Marion Rassat
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Federica Miressi
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Anne Sophie Lia
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France; CHU Limoges, UF de Bioinformatique, F-87000, Limoges France
| | - Franck Sturtz
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | | | - Frédéric Favreau
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Yohan Benayoun
- Chénieux Ophtalmologie, Polyclinique de Limoges ELSAN, F-87000, Limoges, France
| |
Collapse
|
20
|
Ali M, Kabir F, Raskar S, Renuse S, Na CH, Delannoy M, Khan SY, Riazuddin SA. Generation and proteome profiling of PBMC-originated, iPSC-derived lentoid bodies. Stem Cell Res 2020; 46:101813. [PMID: 32474394 DOI: 10.1016/j.scr.2020.101813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/18/2020] [Accepted: 04/09/2020] [Indexed: 01/13/2023] Open
Abstract
Here, we report proteome profiling of peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cell (iPSC)-derived, lens-like organoids termed lentoid bodies at two differentiation time points. A small aliquot of the blood sample was ascertained to collect PBMCs that were reprogrammed to iPSCs. The PBMC-originated, iPSCs were differentiated to lentoid bodies employing the "fried egg" method. Quantitative real-time PCR (qRT-PCR) analysis revealed increased expression levels of lens-associated markers in lentoid bodies while transmission electron microscopy identified closely packed lens epithelial- and differentiating fiber-like cells in lentoid bodies. Total cellular protein was extracted from lentoid bodies at differentiation day 25 and mass spectrometry identified a total of 9,473 proteins. The low counts of crystallin proteins at differentiation day 25 prompted us to re-examine the proteome at differentiation day 35 as we reasoned that 10 additional days of differentiation will increase the crystallin count. However, we did not detect any substantial increase in crystallin protein counts at differentiation day 35. In conclusion, we report generation and proteome profiles of PBMC-originated, iPSC-derived lentoid bodies at multiple differentiation time points.
Collapse
Affiliation(s)
- Muhammad Ali
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Firoz Kabir
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Snehal Raskar
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Santosh Renuse
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chan Hyun Na
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Delannoy
- Department of Cell Biology and Imaging Facility, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shahid Y Khan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - S Amer Riazuddin
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
21
|
Yan P, Zhou B, Ma Y, Wang A, Hu X, Luo Y, Yuan Y, Wei Y, Pang P, Mao J. Tracking the important role of JUNB in hepatocellular carcinoma by single-cell sequencing analysis. Oncol Lett 2019; 19:1478-1486. [PMID: 31966074 PMCID: PMC6956120 DOI: 10.3892/ol.2019.11235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most commonly diagnosed liver cancer, accounting for ~90% of all primary malignancy of the liver. Although various medical treatments have been used as systemic therapies, patient survival time may be extended by only a few months. Moreover, the underlying mechanisms of HCC development and progression remain poorly understood. In the present study, the single-cell transcriptome of one in vivo HCC tumor sample, two in vitro HCC cell lines and normal peripheral blood mononuclear cells were analysed in order to identify the potential mechanism underlying the development and progression of HCC. Interestingly, JunB proto-oncogene was identified to serve a role in the immune response and in development and progression of HCC, potentially contributing to the development of novel therapeutics for HCC patients.
Collapse
Affiliation(s)
- Peng Yan
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Bin Zhou
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Yingdong Ma
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Ani Wang
- Department of Cardiovascular Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Xiaojun Hu
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Youli Luo
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Yajun Yuan
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Yajun Wei
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Pengfei Pang
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Junjie Mao
- Center of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China.,Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
22
|
Brejchova K, Dudakova L, Skalicka P, Dobrovolny R, Masek P, Putzova M, Moosajee M, Tuft SJ, Davidson AE, Liskova P. IPSC-Derived Corneal Endothelial-like Cells Act as an Appropriate Model System to Assess the Impact of SLC4A11 Variants on Pre-mRNA Splicing. Invest Ophthalmol Vis Sci 2019; 60:3084-3090. [PMID: 31323090 PMCID: PMC6645617 DOI: 10.1167/iovs.19-26930] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose To report molecular genetic findings in six probands with congenital hereditary endothelial dystrophy (CHED) variably associated with hearing loss (also known as Harboyan syndrome). Furthermore, we developed a cellular model to determine if disease-associated variants induce aberrant SLC4A11 pre-mRNA splicing. Methods Direct sequencing of the entire SLC4A11 coding region was performed in five probands. In one individual, whole genome sequencing was undertaken. The effect of c.2240+5G>A on pre-mRNA splicing was evaluated in a corneal endothelial-like (CE-like) cell model expressing SLC4A11. CE-like cells were derived from autologous induced pluripotent stem cells (iPSCs) via neural crest cells exposed to B27, PDGF-BB, and DKK-2. Total RNA was extracted, and RT-PCR was performed followed by Sanger and a targeted next generation sequencing (NGS) approach to identify and quantify the relative abundance of alternatively spliced transcripts. Results In total, 11 different mutations in SLC4A11 evaluated as pathogenic were identified; of these, c.1237G>A, c.2003T>C, c.1216+1G>A, and c.2240+5G>A were novel. The c.2240+5G>A variant was demonstrated to result in aberrant pre-mRNA splicing. A targeted NGS approach confirmed that the variant introduces a leaky cryptic splice donor site leading to the production of a transcript containing an insertion of six base pairs with the subsequent introduction of a premature stop codon (p.Thr747*). Furthermore, a subset of transcripts comprising full retention of intron 16 also were observed, leading to the same functionally null allele. Conclusions This proof-of-concept study highlights the potential of using CE-like cells to investigate the pathogenic consequences of SLC4A11 disease–associated variants.
Collapse
Affiliation(s)
- Kristyna Brejchova
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Lubica Dudakova
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Pavlina Skalicka
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.,Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Robert Dobrovolny
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Petr Masek
- Clinic of Ophthalmology, University Hospital Ostrava, Ostrava, Czech Republic.,Department of Craniofacial Surgery, University of Ostrava, Ostrava, Czech Republic
| | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, London, United Kingdom.,Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom.,Great Ormond Street Hospital for Children, London, United Kingdom
| | - Stephen J Tuft
- UCL Institute of Ophthalmology, London, United Kingdom.,Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | | | - Petra Liskova
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.,Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,UCL Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
23
|
Comparative transcriptome analysis of hESC- and iPSC-derived lentoid bodies. Sci Rep 2019; 9:18552. [PMID: 31811247 PMCID: PMC6898283 DOI: 10.1038/s41598-019-54258-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 10/26/2019] [Indexed: 01/18/2023] Open
Abstract
The ocular lens serves as an excellent system to investigate the intricate details of development and differentiation. Generation of lentoid bodies or lens-like structures using pluripotent stem cells is important for understanding the processes critical for lens morphogenesis and the mechanism of cataractogenesis. We previously reported the generation of peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cells (iPSCs). Here, we report generation of lentoid bodies from human embryonic stem cells (hESCs) and (PBMC)-originated, iPSCs employing the “fried egg” method with brief modifications. The ultrastructure analysis of hESC- and iPSC-derived lentoid bodies identified closely packed lens epithelial- and differentiating fiber-like cells. In addition, we performed RNA sequencing (RNA-Seq) based transcriptome profiling of hESC- and iPSC-derived lentoid bodies at differentiation day 25. Next-generation RNA sequencing (RNA-Seq) of hESC- and iPSC-derived lentoid bodies detected expression (≥0.659 RPKM) of 13,975 and 14,003 genes, respectively. Comparative transcriptome analysis of hESC- and iPSC-derived lentoid bodies revealed 13,563 (>96%) genes common in both datasets. Among the genes common in both transcriptome datasets, 12,856 (~95%) exhibited a quantitatively similar expression profile. Next, we compared the mouse lens epithelial and fiber cell transcriptomes with hESC- and iPSC-derived lentoid bodies transcriptomes and identified > 96% overlap with lentoid body transcriptomes. In conclusion, we report first-time comparative transcriptome analysis of hESC- and iPSC-derived lentoid bodies at differentiation day 25.
Collapse
|
24
|
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
|
25
|
Ali M, Khan SY, Kabir F, Gottsch JD, Riazuddin SA. Comparative transcriptome analysis of hESC- and iPSC-derived corneal endothelial cells. Exp Eye Res 2018; 176:252-257. [DOI: 10.1016/j.exer.2018.08.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/27/2018] [Accepted: 08/30/2018] [Indexed: 12/14/2022]
|