1
|
Tsedilina TR, Sharova EI, Kanygina AV, Malyugin BE, Antonova OP, Belodedova AV, Tkachenko IS, Gelyastanov AM, Zolotarev AV, Klokov AV, Murashev AO, Fedyushkina IV, Generozov EV, Skorodumova LO. From Genes to Disease: Reassessing LOXHD1 and AGBL1's Contribution to Fuchs' Dystrophy. Int J Mol Sci 2025; 26:3343. [PMID: 40244234 PMCID: PMC11989410 DOI: 10.3390/ijms26073343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Fuchs' endothelial corneal dystrophy (FECD) is a genetically complex eye disease associated with multiple genes. A recent systematic review has raised concerns about the causal role of variants in the LOXHD1 and AGBL1 genes in the development of FECD. Conflicting data have been reported on the expression of the LOXHD1 and AGBL1 genes in the corneal endothelium. Furthermore, only partial segregation of the variants was observed in familial cases. An analysis of published datasets was conducted to examine the expression of LOXHD1 and AGBL1 genes in normal and FECD-affected corneal endothelia and progenitor cells. Neither LOXHD1 nor AGBL1 genes were expressed in normal or FECD corneal endothelia or progenitor cells. In-house cohorts were screened for carriers of previously reported LOXHD1 and AGBL1 variants. Carriers and their first-degree relatives were invited for an ophthalmological examination to reassess the causal relationship of these variants with FECD phenotype. Three carriers of LOXHD1 variants (one carrier of rs200242497 and two carriers of rs192376005) and two carriers of AGBL1 variants (rs181958589 and rs185919705) were recruited. None of the carriers or first-degree relatives over 50 years exhibited phenotypic signs of FECD via ophthalmic examination. The causal role of the AGBL1 and LOXHD1 variants found in the carriers was not confirmed. Taken together, our findings do not support a causal role for AGBL1 and LOXHD1 in the development of FECD.
Collapse
Affiliation(s)
- Tatiana Romanovna Tsedilina
- Medical Genomics Laboratory, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, 119435 Moscow, Russia; (T.R.T.); (E.I.S.)
| | - Elena Ivanovna Sharova
- Medical Genomics Laboratory, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, 119435 Moscow, Russia; (T.R.T.); (E.I.S.)
| | - Alexandra Vasilevna Kanygina
- Medical Genomics Laboratory, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, 119435 Moscow, Russia; (T.R.T.); (E.I.S.)
| | | | - Olga Pavlovna Antonova
- Department of Anterior Segment Transplant and Optical Reconstructive Surgery, S. Fyodorov Eye Microsurgery Complex Federal State Institution, 127486 Moscow, Russia
| | - Alexandra Vladimirovna Belodedova
- Department of Anterior Segment Transplant and Optical Reconstructive Surgery, S. Fyodorov Eye Microsurgery Complex Federal State Institution, 127486 Moscow, Russia
| | - Ivan Sergeevich Tkachenko
- Department of Anterior Segment Transplant and Optical Reconstructive Surgery, S. Fyodorov Eye Microsurgery Complex Federal State Institution, 127486 Moscow, Russia
| | - Aslan Mukhtarovich Gelyastanov
- Department of Anterior Segment Transplant and Optical Reconstructive Surgery, S. Fyodorov Eye Microsurgery Complex Federal State Institution, 127486 Moscow, Russia
| | | | | | | | - Irina Viktorovna Fedyushkina
- Laboratory of Human Molecular Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, 119435 Moscow, Russia (E.V.G.)
| | - Edward Viktorovich Generozov
- Laboratory of Human Molecular Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, 119435 Moscow, Russia (E.V.G.)
| | - Liubov Olegovna Skorodumova
- Medical Genomics Laboratory, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, 119435 Moscow, Russia; (T.R.T.); (E.I.S.)
| |
Collapse
|
2
|
Sun F, Xi LWQ, Luu W, Enkhbat M, Neo D, Mehta JS, Peh GSL, Yim EKF. Preclinical Models for Studying Fuchs Endothelial Corneal Dystrophy. Cells 2025; 14:505. [PMID: 40214459 PMCID: PMC11988011 DOI: 10.3390/cells14070505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Fuchs Endothelial Corneal Dystrophy (FECD) is a corneal endothelial disease that causes microenvironment alterations and endothelial cell loss, which leads to vision impairment. It has a high global prevalence, especially in elderly populations. FECD is also one of the leading indications of corneal transplantation globally. Currently, there is no clearly defined canonical pathway for this disease, and it has been proposed that the combinatorial effects of genetic mutations and exogenous factors cause FECD. Clinical studies and observations have provided valuable knowledge and understanding of FECD, while preclinical studies are essential for gaining insights into disease progression and mechanisms for the development and testing of regenerative medicine therapies. In this review, we first introduce the proposed genetic and molecular pathologies of FECD. Notably, we discuss the impact of abnormal extracellular matrix deposition (guttata), endothelial-to-mesenchymal transition, cell senescence, and oxidative stress on the pathology and etiology of FECD. We review and summarize the in vitro cell models, ex vivo tissues, and in vivo animal models used to study FECD. The benefits and challenges of each model are also discussed.
Collapse
Affiliation(s)
- Fancheng Sun
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Lexie W. Q. Xi
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Wesley Luu
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Myagmartsend Enkhbat
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
| | - Dawn Neo
- Singapore Eye Research Institute, Singapore 169856, Singapore; (D.N.); (J.S.M.)
| | - Jodhbir S. Mehta
- Singapore Eye Research Institute, Singapore 169856, Singapore; (D.N.); (J.S.M.)
- Ophthalmology Academic Clinical Program, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Gary S. L. Peh
- Singapore Eye Research Institute, Singapore 169856, Singapore; (D.N.); (J.S.M.)
| | - Evelyn K. F. Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada; (F.S.); (L.W.Q.X.); (W.L.); (M.E.)
- Centre for Biotechnology and Bioengineering, University of Waterloo, Waterloo, ON NL2 3G1, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON NL2 3G1, Canada
| |
Collapse
|
3
|
Yang GN, Sun YBY, Roberts PK, Moka H, Sung MK, Gardner-Russell J, El Wazan L, Toussaint B, Kumar S, Machin H, Dusting GJ, Parfitt GJ, Davidson K, Chong EW, Brown KD, Polo JM, Daniell M. Exploring single-cell RNA sequencing as a decision-making tool in the clinical management of Fuchs' endothelial corneal dystrophy. Prog Retin Eye Res 2024; 102:101286. [PMID: 38969166 DOI: 10.1016/j.preteyeres.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has enabled the identification of novel gene signatures and cell heterogeneity in numerous tissues and diseases. Here we review the use of this technology for Fuchs' Endothelial Corneal Dystrophy (FECD). FECD is the most common indication for corneal endothelial transplantation worldwide. FECD is challenging to manage because it is genetically heterogenous, can be autosomal dominant or sporadic, and progress at different rates. Single-cell RNA sequencing has enabled the discovery of several FECD subtypes, each with associated gene signatures, and cell heterogeneity. Current FECD treatments are mainly surgical, with various Rho kinase (ROCK) inhibitors used to promote endothelial cell metabolism and proliferation following surgery. A range of emerging therapies for FECD including cell therapies, gene therapies, tissue engineered scaffolds, and pharmaceuticals are in preclinical and clinical trials. Unlike conventional disease management methods based on clinical presentations and family history, targeting FECD using scRNA-seq based precision-medicine has the potential to pinpoint the disease subtypes, mechanisms, stages, severities, and help clinicians in making the best decision for surgeries and the applications of therapeutics. In this review, we first discuss the feasibility and potential of using scRNA-seq in clinical diagnostics for FECD, highlight advances from the latest clinical treatments and emerging therapies for FECD, integrate scRNA-seq results and clinical notes from our FECD patients and discuss the potential of applying alternative therapies to manage these cases clinically.
Collapse
Affiliation(s)
- Gink N Yang
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Yu B Y Sun
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Philip Ke Roberts
- Department of Ophthalmology, Medical University Vienna, 18-20 Währinger Gürtel, Vienna, Austria
| | - Hothri Moka
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Min K Sung
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Jesse Gardner-Russell
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Layal El Wazan
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Bridget Toussaint
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Satheesh Kumar
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Heather Machin
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Lions Eye Donation Service, Level 7, Smorgon Family Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia
| | - Gregory J Dusting
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Geraint J Parfitt
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Kathryn Davidson
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Elaine W Chong
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Department of Ophthalmology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Karl D Brown
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Jose M Polo
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Mark Daniell
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Lions Eye Donation Service, Level 7, Smorgon Family Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
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
|
5
|
Alonso-Alonso S, Vázquez N, Chacón M, Caballero-Sánchez N, Del Olmo-Aguado S, Suárez C, Alfonso-Bartolozzi B, Fernández-Vega-Cueto L, Nagy L, Merayo-Lloves J, Meana A. An effective method for culturing functional human corneal endothelial cells using a xenogeneic free culture medium. Sci Rep 2023; 13:19492. [PMID: 37945668 PMCID: PMC10636196 DOI: 10.1038/s41598-023-46590-2] [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: 07/12/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
Endothelial dysfunction is a leading cause of corneal blindness in developed countries and the only available treatment is the endothelial transplantation. However, the limited availability of suitable donors remains a significant challenge, driving the exploration of alternative regenerative therapies. Advanced Therapy Medicinal Products show promise but must adhere to strict regulations that prohibit the use of animal-derived substances. This study investigates a novel culture methodology using Plasma Rich in Growth Factors (PRGF) as the only source of growth factors for primary cultures of human corneal endothelial cells (CECs). CECs were obtained from discarded corneas or endothelial rings and cultured in two different media: one supplemented with xenogeneic factors and other xenogeneic-free, using PRGF. Comprehensive characterization through immunofluorescence, morphological analyses, trans-endothelial electrical resistance measurements, RNA-seq, and qPCR was conducted on the two groups. Results demonstrate that CECs cultured in the xenogeneic-free medium exhibit comparable gene expression, morphology, and functionality to those cultured in the xenogeneic medium. Notably, PRGF-expanded CECs share 46.9% of the gene expression profile with native endothelium and express all studied endothelial markers. In conclusion, PRGF provides an effective source of xenogeneic-free growth factors for the culture of CECs from discarded corneal tissue. Further studies will be necessary to demonstrate the applicability of these cultures to cell therapies that make clinical translation possible.
Collapse
Affiliation(s)
- S Alonso-Alonso
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Avenida Doctores Fernández Vega, 33012, Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
| | - N Vázquez
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Avenida Doctores Fernández Vega, 33012, Oviedo, Asturias, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain.
| | - M Chacón
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Avenida Doctores Fernández Vega, 33012, Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
| | - N Caballero-Sánchez
- Doctoral School of Molecular Cell and Immunobiology. Faculty of Medicine, University of Debrecen, Nagyerdei Krt, Debrecen, 4032, Hungary
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Nagyerdei Krt, Debrecen, 4032, Hungary
| | - S Del Olmo-Aguado
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Avenida Doctores Fernández Vega, 33012, Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
| | - C Suárez
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Avenida Doctores Fernández Vega, 33012, Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
| | - B Alfonso-Bartolozzi
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
- Instituto Oftalmológico Fernández-Vega. Avenida Doctores Fernández-Vega, 33012, Oviedo, Asturias, Spain
| | - L Fernández-Vega-Cueto
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
- Instituto Oftalmológico Fernández-Vega. Avenida Doctores Fernández-Vega, 33012, Oviedo, Asturias, Spain
| | - L Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Nagyerdei Krt, Debrecen, 4032, Hungary
- Department of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, 6Th Ave S, St. Petersburg, FL, 33701, USA
| | - J Merayo-Lloves
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Avenida Doctores Fernández Vega, 33012, Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
| | - A Meana
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, Avenida Doctores Fernández Vega, 33012, Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avenida del Hospital Universitario, 33011, Oviedo, Asturias, Spain
- Unidad de Ingeniería Tisular, Centro Comunitario Sangre y Tejidos de Asturias (CCST), Unidad 714 CIBERER, Calle Emilio Rodríguez Vigil, 33006, Oviedo, Asturias, Spain
| |
Collapse
|
6
|
Tsedilina TR, Sharova E, Iakovets V, Skorodumova LO. Systematic review of SLC4A11, ZEB1, LOXHD1, and AGBL1 variants in the development of Fuchs' endothelial corneal dystrophy. Front Med (Lausanne) 2023; 10:1153122. [PMID: 37441688 PMCID: PMC10333596 DOI: 10.3389/fmed.2023.1153122] [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: 01/28/2023] [Accepted: 03/30/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction The pathogenic role of variants in TCF4 and COL8A2 in causing Fuchs' endothelial corneal dystrophy (FECD) is not controversial and has been confirmed by numerous studies. The causal role of other genes, SLC4A11, ZEB1, LOXHD1, and AGBL1, which have been reported to be associated with FECD, is more complicated and less obvious. We performed a systematic review of the variants in the above-mentioned genes in FECD cases, taking into account the currently available population frequency information, transcriptomic data, and the results of functional studies to assess their pathogenicity. Methods Search for articles published in 2005-2022 was performed manually between July 2022 and February 2023. We searched for original research articles in peer-reviewed journals, written in English. Variants in the genes of interest identified in patients with FECD were extracted for the analysis. We classified each presented variant by pathogenicity status according to the ACMG criteria implemented in the Varsome tool. Diagnosis, segregation data, presence of affected relatives, functional analysis results, and gene expression in the corneal endothelium were taken into account. Data on the expression of genes of interest in the corneal endothelium were extracted from articles in which transcriptome analysis was performed. The identification of at least one variant in a gene classified as pathogenic or significantly associated with FECD was required to confirm the causal role of the gene in FECD. Results The analysis included 34 articles with 102 unique ZEB1 variants, 20 articles with 64 SLC4A11 variants, six articles with 26 LOXHD1 variants, and five articles with four AGBL1 variants. Pathogenic status was confirmed for seven SLC4A11 variants found in FECD. No variants in ZEB1, LOXHD1, and AGBL1 genes were classified as pathogenic for FECD. According to the transcriptome data, AGBL1 and LOXHD1 were not expressed in the corneal endothelium. Functional evidence for the association of LOXHD1, and AGBL1 with FECD was conflicting. Conclusion Our analysis confirmed the causal role of SLC4A11 variants in the development of FECD. The causal role of ZEB1, LOXHD1, and AGBL1 variants in FECD has not been confirmed. Further evidence from familial cases and functional analysis is needed to confirm their causal roles in FECD.
Collapse
Affiliation(s)
- Tatiana Romanovna Tsedilina
- Laboratory of Human Molecular Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Elena Sharova
- Laboratory of Human Molecular Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Valeriia Iakovets
- Laboratory of Human Molecular Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Liubov Olegovna Skorodumova
- Laboratory of Human Molecular Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| |
Collapse
|
7
|
Català P, Groen N, LaPointe VLS, Dickman MM. A single-cell RNA-seq analysis unravels the heterogeneity of primary cultured human corneal endothelial cells. Sci Rep 2023; 13:9361. [PMID: 37291161 PMCID: PMC10249941 DOI: 10.1038/s41598-023-36567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/06/2023] [Indexed: 06/10/2023] Open
Abstract
The cornea is a transparent and avascular tissue located in front of the eye. Its inner surface is lined by a monolayer of corneal endothelial cells (CECs), which maintain the cornea transparency. CECs remain arrested in a non-proliferative state and damage to these cells can compromise their function leading to corneal opacity. The primary culture of donor-derived CECs is a promising cell therapy. It confers the potential to treat multiple patients from a single donor, alleviating the global donor shortage. Nevertheless, this approach has limitations preventing its adoption, particularly culture protocols allow limited expansion of CECs and there is a lack of clear parameters to identify therapy-grade CECs. To address this limitation, a better understanding of the molecular changes arising from the primary culture of CECs is required. Using single-cell RNA sequencing on primary cultured CECs, we identify their variable transcriptomic fingerprint at the single cell level, provide a pseudo-temporal reconstruction of the changes arising from primary culture, and suggest markers to assess the quality of primary CEC cultures. This research depicts a deep transcriptomic understanding of the cellular heterogeneity arising from the primary expansion of CECs and sets the basis for further improvement of culture protocols and therapies.
Collapse
Affiliation(s)
- Pere Català
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- University Eye Clinic Maastricht, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | | | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Mor M Dickman
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- University Eye Clinic Maastricht, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| |
Collapse
|
8
|
Talpan D, Salla S, Meusel L, Walter P, Kuo CC, Franzen J, Fuest M. Cytoprotective Effects of Human Platelet Lysate during the Xeno-Free Culture of Human Donor Corneas. Int J Mol Sci 2023; 24:ijms24032882. [PMID: 36769200 PMCID: PMC9917909 DOI: 10.3390/ijms24032882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
We evaluated the suitability of 2% human platelet lysate medium (2%HPL) as a replacement for 2% fetal bovine serum medium (2%FBS) for the xeno-free organ culture of human donor corneas. A total of 32 corneas from 16 human donors were cultured in 2%FBS for 3 days (TP1), then evaluated using phase contrast microscopy (endothelial cell density (ECD) and cell morphology). Following an additional 25-day culture period (TP2) in either 2%FBS or 2%HPL, the pairs were again compared using microscopy; then stroma and Descemet membrane/endothelium (DmE) were processed for next generation sequencing (NGS). At TP2 the ECD was higher in the 2%HPL group (2179 ± 288 cells/mm2) compared to 2%FBS (2113 ± 331 cells/mm2; p = 0.03), and endothelial cell loss was lower (ECL HPL = -0.7% vs. FBS = -3.8%; p = 0.01). There were no significant differences in cell morphology between TP1 and 2, or between 2%HPL and 2%FBS. NGS showed the differential expression of 1644 genes in endothelial cells and 217 genes in stromal cells. It was found that 2%HPL led to the upregulation of cytoprotective, anti-inflammatory and anti-fibrotic genes (HMOX1, SERPINE1, ANGPTL4, LEFTY2, GADD45B, PLIN2, PTX3, GFRA1/2), and the downregulation of pro-inflammatory/apoptotic genes (e.g., CXCL14, SIK1B, PLK5, PPP2R3B, FABP5, MAL, GATA3). 2%HPL is a suitable xeno-free substitution for 2%FBS in human cornea organ culture, inducing less ECL and producing potentially beneficial alterations in gene expression.
Collapse
Affiliation(s)
- Delia Talpan
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
| | - Sabine Salla
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Linus Meusel
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Peter Walter
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Chao-Chung Kuo
- Genomics Facility, Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Julia Franzen
- Genomics Facility, Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| | - Matthias Fuest
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
- Correspondence:
| |
Collapse
|
9
|
Pizzuto S, Duffey G, Weant J, Eveleth D. Acceleration of Regeneration of the Corneal Endothelial Layer After Descemet Stripping Induced by the Engineered FGF TTHX1114 in Human Corneas in Organ Culture. Cornea 2023; 42:232-242. [PMID: 35942526 PMCID: PMC9797199 DOI: 10.1097/ico.0000000000003098] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/07/2022] [Accepted: 05/23/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Descemet stripping only (DSO, descemetorhexis without endothelial keratoplasty) is increasing in clinical use but can impose long recovery times. The objective of this research was to determine whether TTHX1114, an engineered analog of FGF1, could accelerate healing in corneas after DSO. METHODS Corneas obtained from eye banks were placed into suspension culture and subjected to DSO with a procedure comparable with that used clinically. The healing of the stripped area and the regeneration of the corneal endothelial cell (CEC) layer were evaluated intermittently for 14 days using trypan blue staining, alizarin red staining, and immunohistochemistry. RESULTS Corneas subjected to DSO showed about 30% of the stripped area healed after 14 days in culture while those treated with TTHX1114 healed 81%. The healed area was similar in both normal corneas and corneas judged by the eye banks to be dystrophic. The regeneration of the endothelial layer in the stripped area was substantially more complete in TTHX1114-treated corneas, most of which demonstrated a contiguous monolayer of CECs expressing ZO-1 at the cell-cell junctions. In corneas not subject to DSO, incorporation of EdU, a marker of proliferation, was stimulated by TTHX1114 treatment. CONCLUSIONS The corneal organ culture model recapitulated clinical observations of DSO, only with much more rapid recovery. Within the immediate postsurgical time frame of 2 weeks, treatment with TTHX1114 stimulated near-total regeneration of the CEC layer, suggesting that TTHX1114 may be useful as an adjunct to DSO.
Collapse
|
10
|
Ying PX, Fu M, Huang C, Li ZH, Mao QY, Fu S, Jia XH, Cao YC, Hong LB, Cai LY, Guo X, Liu RB, Meng FK, Yi GG. Profile of biological characterizations and clinical application of corneal stem/progenitor cells. World J Stem Cells 2022; 14:777-797. [PMID: 36483848 PMCID: PMC9724387 DOI: 10.4252/wjsc.v14.i11.777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Corneal stem/progenitor cells are typical adult stem/progenitor cells. The human cornea covers the front of the eyeball, which protects the eye from the outside environment while allowing vision. The location and function demand the cornea to maintain its transparency and to continuously renew its epithelial surface by replacing injured or aged cells through a rapid turnover process in which corneal stem/progenitor cells play an important role. Corneal stem/progenitor cells include mainly corneal epithelial stem cells, corneal endothelial cell progenitors and corneal stromal stem cells. Since the discovery of corneal epithelial stem cells (also known as limbal stem cells) in 1971, an increasing number of markers for corneal stem/progenitor cells have been proposed, but there is no consensus regarding the definitive markers for them. Therefore, the identification, isolation and cultivation of these cells remain challenging without a unified approach. In this review, we systematically introduce the profile of biological characterizations, such as anatomy, characteristics, isolation, cultivation and molecular markers, and clinical applications of the three categories of corneal stem/progenitor cells.
Collapse
Affiliation(s)
- Pei-Xi Ying
- Department of Ophthalmology, Zhujiang Hospital, The Second Clinical School, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Chang Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200030, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200030, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200030, China
| | - Zhi-Hong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510550, Guangdong Province, China
| | - Qing-Yi Mao
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Sheng Fu
- Hengyang Medical School, The University of South China, Hengyang 421001, Hunan Province, China
| | - Xu-Hui Jia
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Yu-Chen Cao
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Li-Bing Hong
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Li-Yang Cai
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xi Guo
- Medical College of Rehabilitation, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Ru-Bing Liu
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Fan-ke Meng
- Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Guo-Guo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| |
Collapse
|
11
|
Hazra S, Sneha IV, Chaurasia S, Ramachandran C. In Vitro Expansion of Corneal Endothelial Cells for Clinical Application: Current Update. Cornea 2022; 41:1313-1324. [PMID: 36107851 DOI: 10.1097/ico.0000000000003080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/08/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT Endothelial dysfunction is one of the leading causes of corneal blindness and one of the common indications for keratoplasty. At present, the standard of treatment involves the replacement of the dysfunctional endothelium with healthy tissue taken from a donor. Because there is a paucity of healthy donor tissues, research on the corneal endothelium has focused primarily on expanding these cells in the laboratory for transplantation in an attempt to reduce the gap between the demand and supply of donor tissues for transplantation. To expand these cells, which are nonmitotic in vivo, various mitogens, substrates, culture systems, and alternate strategies have been tested with varying success. The biggest challenge has been the limited proliferative capacity of these cells compounded with endothelial to mesenchymal transition that alters the functioning of these cells and renders them unsuitable for human transplantation. This review aims to give a comprehensive overview of the most common and successful techniques used in the culture of the cells, the current available evidence in support of epithelial to mesenchymal transition (EMT), alternate sources for deriving the corneal endothelial cells, and advances made in transplantation of these cells.
Collapse
Affiliation(s)
- Swatilekha Hazra
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
- Manipal University, Manipal, Karnataka, India ; and
| | - Iskala V Sneha
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | | | | |
Collapse
|
12
|
Looking into the Eyes—In Vitro Models for Ocular Research. Int J Mol Sci 2022; 23:ijms23169158. [PMID: 36012421 PMCID: PMC9409455 DOI: 10.3390/ijms23169158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Animal research undoubtedly provides scientists with virtually unlimited data but inflicts pain and suffering on animals. Currently, legislators and scientists alike are promoting alternative in vitro approaches allowing for an accurate evaluation of processes occurring in the body without animal sacrifice. Historically, one of the most infamous animal tests is the Draize test, mainly performed on rabbits. Even though this test was considered the gold standard for around 50 years, the Draize test fails to mimic human response mainly due to human and rabbit eye physiological differences. Therefore, many alternative assays were developed to evaluate ocular toxicity and drug effectiveness accurately. Here we review recent achievements in tissue engineering of in vitro 2D, 2.5D, 3D, organoid and organ-on-chip ocular models, as well as in vivo and ex vivo models in terms of their advantages and limitations.
Collapse
|
13
|
Nakai H, Tsuchiya Y, Koike N, Asano T, Ueno M, Umemura Y, Sasawaki Y, Ono R, Hamuro J, Sotozono C, Yagita K. Comprehensive Analysis Identified the Circadian Clock and Global Circadian Gene Expression in Human Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2022; 63:16. [PMID: 35579906 PMCID: PMC9123520 DOI: 10.1167/iovs.63.5.16] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate circadian clock oscillation and circadian global gene expression in cultured human corneal endothelial cells (cHCECs) to elucidate and assess the potential function of circadian regulation in HCECs. Methods In this study, we introduced a circadian bioluminescence reporter, Bmal1:luciferase (Bmal1:luc), into cHCECs and subsequently monitored real-time bioluminescence rhythms. RNA-sequencing data analysis was then performed using sequential time-course samples of the cHCECs to obtain a comprehensive understanding of the circadian gene expression rhythms. The potential relevance of rhythmically expressed genes was then assessed by systematic approaches using functional clustering and individual gene annotations. Results Bmal1:luc bioluminescence exhibited clear circadian oscillation in the cHCECs. The core clock genes and clock-related genes showed high-amplitude robust circadian messenger RNA (mRNA) expression rhythms in cHCECs after treatment with dexamethasone, and 329 genes that exhibited circadian mRNA expression rhythms were identified (i.e., genes involved in various physiological processes including glycolysis, mitochondrial function, antioxidative systems, hypoxic responses, apoptosis, and extracellular matrix regulation, which represent the physiological functions of HCECs). Conclusions Our findings revealed that cHCECs have a robust and functional circadian clock, and our discovery that a large number of genes exhibit circadian mRNA expression rhythms in cHCECs suggests a potential contribution of circadian regulation to fine-tune HCEC functions for daily changes in the environment.
Collapse
Affiliation(s)
- Hiroko Nakai
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshiki Tsuchiya
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuya Koike
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taiki Asano
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiro Umemura
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuh Sasawaki
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryutaro Ono
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junji Hamuro
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
14
|
Spinozzi D, Miron A, Bruinsma M, Dapena I, Kocaba V, Jager MJ, Melles GRJ, Ni Dhubhghaill S, Oellerich S. New developments in corneal endothelial cell replacement. Acta Ophthalmol 2021; 99:712-729. [PMID: 33369235 DOI: 10.1111/aos.14722] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022]
Abstract
Corneal transplantation is currently the most effective treatment to restore corneal clarity in patients with endothelial disorders. Endothelial transplantation, either by Descemet membrane endothelial keratoplasty (DMEK) or by Descemet stripping (automated) endothelial keratoplasty (DS(A)EK), is a surgical approach that replaces diseased Descemet membrane and endothelium with tissue from a healthy donor eye. Its application, however, is limited by the availability of healthy donor tissue. To increase the pool of endothelial grafts, research has focused on developing new treatment options as alternatives to conventional corneal transplantation. These treatment options can be considered as either 'surgery-based', that is tissue-efficient modifications of the current techniques (e.g. Descemet stripping only (DSO)/Descemetorhexis without endothelial keratoplasty (DWEK) and Quarter-DMEK), or 'cell-based' approaches, which rely on in vitro expansion of human corneal endothelial cells (hCEC) (i.e. cultured corneal endothelial cell sheet transplantation and cell injection). In this review, we will focus on the most recent developments in the field of the 'cell-based' approaches. Starting with the description of aspects involved in the isolation of hCEC from donor tissue, we then describe the different natural and bioengineered carriers currently used in endothelial cell sheet transplantation, and finally, we discuss the current 'state of the art' in novel therapeutic approaches such as endothelial cell injection.
Collapse
Affiliation(s)
- Daniele Spinozzi
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
| | - Alina Miron
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
| | - Marieke Bruinsma
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
| | - Isabel Dapena
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
- Melles Cornea Clinic Rotterdam The Netherlands
| | - Viridiana Kocaba
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
- Melles Cornea Clinic Rotterdam The Netherlands
- Tissue Engineering and Stem Cell Group Singapore Eye Research Institute Singapore Singapore
| | - Martine J. Jager
- Department of Ophthalmology Leiden University Medical Center Leiden The Netherlands
| | - Gerrit R. J. Melles
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
- Melles Cornea Clinic Rotterdam The Netherlands
- Amnitrans EyeBank Rotterdam The Netherlands
| | - Sorcha Ni Dhubhghaill
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
- Melles Cornea Clinic Rotterdam The Netherlands
- Antwerp University Hospital (UZA) Edegem Belgium
| | - Silke Oellerich
- Netherlands Institute for Innovative Ocular Surgery Rotterdam The Netherlands
| |
Collapse
|
15
|
Smeringaiova I, Paaske Utheim T, Jirsova K. Ex vivo expansion and characterization of human corneal endothelium for transplantation: a review. Stem Cell Res Ther 2021; 12:554. [PMID: 34717745 PMCID: PMC8556978 DOI: 10.1186/s13287-021-02611-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
The corneal endothelium plays a key role in maintaining corneal transparency. Its dysfunction is currently treated with penetrating or lamellar keratoplasty. Advanced cell therapy methods seek to address the persistent global deficiency of donor corneas by enabling the renewal of the endothelial monolayer with tissue-engineered grafts. This review provides an overview of recently published literature on the preparation of endothelial grafts for transplantation derived from cadaveric corneas that have developed over the last decade (2010–2021). Factors such as the most suitable donor parameters, culture substrates and media, endothelial graft storage conditions, and transplantation methods are discussed. Despite efforts to utilize alternative cellular sources, such as induced pluripotent cells, cadaveric corneas appear to be the best source of cells for graft preparation to date. However, native endothelial cells have a limited natural proliferative capacity, and they often undergo rapid phenotype changes in ex vivo culture. This is the main reason why no culture protocol for a clinical-grade endothelial graft prepared from cadaveric corneas has been standardized so far. Currently, the most established ex vivo culture protocol involves the peel-and-digest method of cell isolation and cell culture by the dual media method, including the repeated alternation of high and low mitogenic conditions. Culture media are enriched by additional substances, such as signaling pathway (Rho-associated protein kinase, TGF-β, etc.) inhibitors, to stimulate proliferation and inhibit unwanted morphological changes, particularly the endothelial-to-mesenchymal transition. To date, this promising approach has led to the development of endothelial grafts for the first in-human clinical trial in Japan. In addition to the lack of a standard culture protocol, endothelial-specific markers are still missing to confirm the endothelial phenotype in a graft ready for clinical use. Because the corneal endothelium appears to comprise phenotypically heterogeneous populations of cells, the genomic and proteomic expression of recently proposed endothelial-specific markers, such as Cadherin-2, CD166, or SLC4A11, must be confirmed by additional studies. The preparation of endothelial grafts is still challenging today, but advances in tissue engineering and surgery over the past decade hold promise for the successful treatment of endothelial dysfunctions in more patients worldwide.
Collapse
Affiliation(s)
- Ingrida Smeringaiova
- Laboratory of the Biology and Pathology of the Eye, Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic
| | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
| | - Katerina Jirsova
- Laboratory of the Biology and Pathology of the Eye, Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic.
| |
Collapse
|
16
|
Altered gene expression in slc4a11 -/- mouse cornea highlights SLC4A11 roles. Sci Rep 2021; 11:20885. [PMID: 34686736 PMCID: PMC8536660 DOI: 10.1038/s41598-021-98921-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/13/2021] [Indexed: 11/17/2022] Open
Abstract
SLC4A11 is a H+/NH3/water transport protein, of corneal endothelial cells. SLC4A11 mutations cause congenital hereditary endothelial dystrophy and some cases of Fuchs endothelial corneal dystrophy. To probe SLC4A11’s roles, we compared gene expression in RNA from corneas of 17-week-old slc4a11−/− (n = 3) and slc4a11+/+ mice (n = 3) and subjected to RNA sequencing. mRNA levels for a subset of genes were also assessed by quantitative real-time reverse transcription PCR (qRT RT-PCR). Cornea expressed 13,173 genes, which were rank-ordered for their abundance. In slc4a11−/− corneas, 100 genes had significantly altered expression. Abundant slc14a1 expression, encoding the urea transporter UT-A, suggests a significant role in the cornea. The set of genes with altered expression was subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, revealing that alterations clustered into extracellular region, cytoskeleton, cell adhesion and plasma membrane functions. Gene expression changes further clustered into classes (with decreasing numbers of genes): cell fate and development, extracellular matrix and cell adhesion, cytoskeleton, ion homeostasis and energy metabolism. Together these gene changes confirm earlier suggestions of a role of SLC4A11 in ion homeostasis, energy metabolism, cell adhesion, and reveal an unrecognized SLC4A11 role in cytoskeletal organization.
Collapse
|
17
|
Ligocki AJ, Fury W, Gutierrez C, Adler C, Yang T, Ni M, Bai Y, Wei Y, Lehmann GL, Romano C. Molecular characteristics and spatial distribution of adult human corneal cell subtypes. Sci Rep 2021; 11:16323. [PMID: 34381080 PMCID: PMC8357950 DOI: 10.1038/s41598-021-94933-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Bulk RNA sequencing of a tissue captures the gene expression profile from all cell types combined. Single-cell RNA sequencing identifies discrete cell-signatures based on transcriptomic identities. Six adult human corneas were processed for single-cell RNAseq and 16 cell clusters were bioinformatically identified. Based on their transcriptomic signatures and RNAscope results using representative cluster marker genes on human cornea cross-sections, these clusters were confirmed to be stromal keratocytes, endothelium, several subtypes of corneal epithelium, conjunctival epithelium, and supportive cells in the limbal stem cell niche. The complexity of the epithelial cell layer was captured by eight distinct corneal clusters and three conjunctival clusters. These were further characterized by enriched biological pathways and molecular characteristics which revealed novel groupings related to development, function, and location within the epithelial layer. Moreover, epithelial subtypes were found to reflect their initial generation in the limbal region, differentiation, and migration through to mature epithelial cells. The single-cell map of the human cornea deepens the knowledge of the cellular subsets of the cornea on a whole genome transcriptional level. This information can be applied to better understand normal corneal biology, serve as a reference to understand corneal disease pathology, and provide potential insights into therapeutic approaches.
Collapse
Affiliation(s)
- Ann J Ligocki
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Wen Fury
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | | | - Tao Yang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Yu Bai
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | - Carmelo Romano
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA.
| |
Collapse
|
18
|
Pei W, Chen J, Wu W, Wei W, Yu Y, Feng Y. Comparison of the rabbit and human corneal endothelial proteomes regarding proliferative capacity. Exp Eye Res 2021; 209:108629. [PMID: 34029595 DOI: 10.1016/j.exer.2021.108629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/29/2021] [Accepted: 05/16/2021] [Indexed: 02/05/2023]
Abstract
The shortage of human donor corneas has raised important concerns about engineering of corneal endothelial cells (CECs) for clinical use. However, due to the limited proliferative capacity of human CECs, driving them into proliferation and regeneration may be difficult. Unlike human CECs, rabbit CECs have a marked proliferative capacity. To clarify the potential reason for this difference, we analysed the proteomes of four human corneal endothelium samples and four rabbit corneal endothelium samples with quantitative label-free proteomics and downstream analysis. We discovered that vitamin and selenocompound metabolism and some signaling pathways such as NF-kappa B signaling pathway differed between the samples. Moreover, TGFβ, PITX2 and keratocan were distinctively expressed in rabbit samples, which might be associated with active proliferation in rabbit CECs. This study illustrates the proteomic differences between human and rabbit CECs and might promote CEC engineering strategies.
Collapse
Affiliation(s)
- Wendi Pei
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China
| | - Jun Chen
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenyu Wu
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Wei Wei
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Yang Yu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Yun Feng
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
19
|
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
|
20
|
Van Meenen J, Ní Dhubhghaill S, Van den Bogerd B, Koppen C. An Overview of Advanced In Vitro Corneal Models: Implications for Pharmacological Testing. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:506-516. [PMID: 33878935 DOI: 10.1089/ten.teb.2021.0031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cornea is an important barrier to consider when developing ophthalmic formulations, but proper modeling of this multilayered tissue remains a challenge. This is due to the varying properties associated with each layer in addition to the dynamics of the tear film. Hence, the most representative models to date rely on animals. Animal models, however, differ from humans in several aspects and are subject to ethical limitations. Consequently, in vitro approaches are being developed to address these issues. This review focuses on the barrier properties of the cornea and evaluates the most advanced three-dimensional cultures of human corneal equivalents in literature. Their application potential is subsequently assessed and discussed in the context of preclinical testing along with our perspective toward the future. Impact statement Most ocular drugs are applied topically, with the transcorneal pathway as the main administration route. Animal corneas are currently the only advanced models available, contributing to the drug attrition rate. Anatomical and physiological interspecies differences might account for a poor translatability of preclinical results to clinical trials, urging researchers to devise better corneal equivalents. This review elaborates on the emerging generation of three-dimensional in vitro models, which comprises spheroids, organoids, and organs-on-chips, which can serve as a stepping stone for advancements in this field.
Collapse
Affiliation(s)
- Joris Van Meenen
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium
| | - Sorcha Ní Dhubhghaill
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium.,Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium
| | - Bert Van den Bogerd
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium
| | - Carina Koppen
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium.,Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
21
|
Current development of alternative treatments for endothelial decompensation: Cell-based therapy. Exp Eye Res 2021; 207:108560. [PMID: 33811914 DOI: 10.1016/j.exer.2021.108560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/03/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
Current treatment for corneal endothelial dysfunction consists in the replacement of corneal endothelium by keratoplasty. Owing to the scarcity of donor corneas and the increasing number of transplants, alternative treatments such as cell-based therapies are necessary. In this article, we highlight the biological aspects of the cornea and the corneal endothelium, as well as the context that surrounds the need for new alternatives to conventional keratoplasty. We then review some of those experimental treatments in more detail, focusing on the development of the in vitro and preclinical phases of two cell-based therapies: tissue-engineered endothelial keratoplasty (TE-EK) and cell injection. In the case of TE-EK graft construction, we analyse the current progress, considering all the requirements it must meet in order to be functional. Moreover, we discuss the inherent drawbacks of endothelial keratoplasties, which TE-EK grafts should overcome in order to make surgical intervention easier and to improve the outcomes of current endothelial keratoplasties. Finally, we analyse the development of preclinical trials and their limitations in terms of performing an optimal functional evaluation of cell-based therapy, and we conclude by discussing early clinical trials in humans.
Collapse
|
22
|
Ong Tone S, Wylegala A, Böhm M, Melangath G, Deshpande N, Jurkunas UV. Increased Corneal Endothelial Cell Migration in Fuchs Endothelial Corneal Dystrophy: A Live Cell Imaging Study. OPHTHALMOLOGY SCIENCE 2021; 1:100006. [PMID: 36246012 PMCID: PMC9559113 DOI: 10.1016/j.xops.2021.100006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/09/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Purpose To investigate if corneal endothelial cells (CECs) in Fuchs endothelial corneal dystrophy (FECD) have altered cellular migration compared with normal controls. Design Comparative analysis. Materials Descemet's membrane and CECs derived from patients with FECD undergoing endothelial keratoplasty or normal cadaveric donors. Methods Ex vivo specimens were used for live cell imaging and generation of immortalized cell lines. Live imaging was performed on FECD and normal CECs and on ex vivo specimens transfected with green fluorescent protein. Migration speeds were determined as a function of cellular density using automated cell tracking. Ex vivo specimens were classified as either FECD or normal low cell density (nonconfluent) or high cell density (confluent). Scratch assay was performed on CECs seeded at high confluence to determine migration speed. Genetic analysis from blood samples or CECs was performed to detect a CTG repeat expansion in the TCF4 gene. Main Outcome Measures Mean cell migration speed. Results Fuchs endothelial corneal dystrophy CECs in low cell density areas displayed increased mean speed (0.391 ± 0.005 μm/minute vs. 0.364 ± 0.005 μm/minute; P < 0.001) and mean maximum speed (0.961 ± 0.010 μm/minute vs. 0.787 ± 0.011 μm/minute; P < 0.001) compared with normal CECs, and increased mean maximum speed (0.778 ± 0.014 μm/minute vs. 0.680 ± 0.011 μm/minute; P < 0.001) in high cell density areas ex vivo. Similarly, FECD CECs displayed increased mean speed compared with normal CECs (1.958 ± 0.020 μm/minute vs. 2.227 ± 0.021 μm/minute vs. 1.567 ± 0.019 μm/minute; P < 0.001) under nonconfluent conditions in vitro. Moreover, FECD CECs also displayed increased mean speed compared with normal CECs under high confluent conditions as detected by scratch assay (37.2 ± 1.1% vs. 44.3 ± 4.1% vs. 70.7 ± 5.2%; P < 0.001). Morphologic analysis showed that FECD CECs displayed an increased fibroblastic phenotype as detected by filamentous-actin labeling. Conclusions Fuchs endothelial corneal dystrophy CECs demonstrated increased migration speed compared with normal CECs. Further investigation into the mechanisms of heightened cell migration in FECD is needed and may provide insight into its pathogenesis, as well as having implications on descemetorhexis without endothelial keratoplasty.
Collapse
Key Words
- CE, corneal endothelium
- CEC, corneal endothelial cell
- Cell migration
- Corneal endothelium
- DM, Descemet’s membrane
- DMEK, Descemet's membrane endothelial keratoplasty
- DWEK, descemetorhexis without endothelial keratoplasty
- Descemetorhexis without endothelial keratoplasty
- Descemet’s stripping only
- ECD, endothelial cell density
- ECM, extracellular matrix
- EMT, endothelial-to-mesenchymal transition
- FECD, Fuchs endothelial corneal dystrophy
- Fuchs endothelial corneal dystrophy
- GFP, green fluorescent protein
- LNP, lipid nanoparticle
- PBS, phosphate-buffered saline
- TCF4, transcription factor 4
Collapse
Affiliation(s)
- Stephan Ong Tone
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Canada
- Department of Ophthalmology, University of Toronto, Toronto, Canada
| | - Adam Wylegala
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Myriam Böhm
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Geetha Melangath
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Neha Deshpande
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Ula V. Jurkunas
- Cornea Center of Excellence, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Zhang W, Frausto R, Chung DD, Griffis CG, Kao L, Chen A, Azimov R, Sampath AP, Kurtz I, Aldave AJ. Energy Shortage in Human and Mouse Models of SLC4A11-Associated Corneal Endothelial Dystrophies. Invest Ophthalmol Vis Sci 2021; 61:39. [PMID: 32721020 PMCID: PMC7425690 DOI: 10.1167/iovs.61.8.39] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose To elucidate the molecular events in solute carrier family 4 member 11 (SLC4A11)-deficient corneal endothelium that lead to the endothelial dysfunction that characterizes the dystrophies associated with SLC4A11 mutations, congenital hereditary endothelial dystrophy (CHED) and Fuchs endothelial corneal dystrophy 4. Methods Comparative transcriptomic analysis (CTA) was performed in primary human corneal endothelial cells (pHCEnC) and murine corneal endothelial cells (MCEnC) with normal and reduced levels of SLC4A11 (SLC4A11 KD pHCEnC) and Slc4a11 (Slc4a11−/− MCEnC), respectively. Validation of differentially expressed genes was performed using immunofluorescence staining of CHED corneal endothelium, as well as western blot and quantitative PCR analysis of SLC4A11 KD pHCEnC and Slc4a11−/− MCEnC. Functional analyses were performed to investigate potential functional changes associated with the observed transcriptomic alterations. Results CTA revealed inhibition of cell metabolism and ion transport function as well as mitochondrial dysfunction, leading to reduced adenosine triphosphate (ATP) production, in SLC4A11 KD pHCEnC and Slc4a11−/− MCEnC. Co-localization of SNARE protein STX17 with mitochondria marker COX4 was observed in CHED corneal endothelium, as was activation of AMPK–p53/ULK1 in both SLC4A11 KD pHCEnC and Slc4a11−/− MCEnC, providing additional evidence of mitochondrial dysfunction and mitophagy. Reduced Na+-dependent HCO3− transport activity and altered NH4Cl-induced membrane potential changes were observed in Slc4a11−/− MCEnC. Conclusions Reduced steady-state ATP levels and subsequent activation of the AMPK–p53 pathway provide a link between the metabolic functional deficit and transcriptome alterations, as well as evidence of insufficient ATP to maintain the Na+/K+-ATPase corneal endothelial pump as the cause of the edema that characterizes SLC4A11-associated corneal endothelial dystrophies.
Collapse
|
24
|
Sun K, Gu L, Ma L, Duan Y. Atlas of ACE2 gene expression reveals novel insights into transmission of SARS-CoV-2. Heliyon 2021; 7:e05850. [PMID: 33392409 PMCID: PMC7762714 DOI: 10.1016/j.heliyon.2020.e05850] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/03/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
The recent pandemic, COVID-19, is caused by a novel coronavirus, SARS-CoV-2, with elusive origin. SARS-CoV-2 infects mammalian cells via ACE2, a transmembrane protein. Therefore, the conservation and expression patterns of ACE2 may provide valuable insights into tracing the carriers of SARS-CoV-2. In this work, we analyzed the conservation of ACE2 and its expression pattern among various mammalian species that are close to human beings. We show that mammalian ACE2 gene is deeply conserved at both DNA and peptide levels, suggesting that a broad range of mammals can potentially host SARS-CoV-2. We further report that ACE2 expression in certain human tissues are consistent with clinical symptoms of COVID-19 patients. Furthermore, we have built the first atlas of ACE2 expression in various common mammals, which shows that ACE2 expresses in mammalian tissues in a species-specific manner. Most notably, we observe exceptionally high expression of ACE2 in external body parts of cats and dogs, suggesting that these household pet animals could be vulnerable to viral infections and/or may serve as intermediate hosts, thus yielding novel insights into the transmission of SARS-CoV-2.
Collapse
Affiliation(s)
- Kun Sun
- Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Liuqi Gu
- Department of Ecology and Evolutionary Biology, The University of Kansas, Lawrence, KS, 66045, USA
- Beijing Huayuan Academy of Biotechnology, Beijing, 100192, China
| | - Li Ma
- Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yunfeng Duan
- Beijing Huayuan Academy of Biotechnology, Beijing, 100192, China
| |
Collapse
|
25
|
Transcriptome dataset of human corneal endothelium based on ribosomal RNA-depleted RNA-Seq data. Sci Data 2020; 7:407. [PMID: 33219220 PMCID: PMC7680133 DOI: 10.1038/s41597-020-00754-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
The corneal endothelium maintains corneal transparency; consequently, damage to this endothelium by a number of pathological conditions results in severe vision loss. Publicly available expression databases of human tissues are useful for investigating the pathogenesis of diseases and for developing new therapeutic modalities; however, databases for ocular tissues, and especially the corneal endothelium, are poor. Here, we have generated a transcriptome dataset from the ribosomal RNA-depleted total RNA from the corneal endothelium of eyes from seven Caucasians without ocular diseases. The results of principal component analysis and correlation coefficients (ranged from 0.87 to 0.96) suggested high homogeneity of our RNA-Seq dataset among the samples, as well as sufficient amount and quality. The expression profile of tissue-specific marker genes indicated only limited, if any, contamination by other layers of the cornea, while the Smirnov-Grubbs test confirmed the absence of outlier samples. The dataset presented here should be useful for investigating the function/dysfunction of the cornea, as well as for extended transcriptome analyses integrated with expression data for non-coding RNAs. Measurement(s) | RNA | Technology Type(s) | RNA sequencing | Factor Type(s) | sex | Sample Characteristic - Organism | Homo sapiens |
Machine-accessible metadata file describing the reported data: 10.6084/m9.figshare.13186868
Collapse
|
26
|
Eveleth D, Pizzuto S, Weant J, Jenkins-Eveleth J, Bradshaw RA. Proliferation of Human Corneal Endothelia in Organ Culture Stimulated by Wounding and the Engineered Human Fibroblast Growth Factor 1 Derivative TTHX1114. J Ocul Pharmacol Ther 2020; 36:686-696. [PMID: 32735473 PMCID: PMC7703086 DOI: 10.1089/jop.2019.0119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose: Corneal endothelial dystrophies are characterized by endothelial cell loss and dysfunction. Recent evidence suggests that corneal endothelial cells (CECs) can regenerate although they do not do so under normal conditions. This work sought to test whether CECs can be stimulated to proliferate in organ culture by wounding and/or by treatment with the engineered human fibroblast growth factor 1 (FGF1) derivative TTHX1114. Methods: Human donor corneas obtained from eye banks were maintained in organ culture in the presence or absence of TTHX1114. Wounds in the corneas were created by quartering the corneas. The CEC monolayer was identified as a regular layer by Hoechst staining of the nuclear DNA with cell outlines delineated by immunohistochemical identification of ZO-1. Nuclei and nuclei incorporating 5-ethynyl-2′-deoxyuridine (EdU) were counted using ImageJ. Results: CECs in normal corneas in undisturbed monolayers had low, but measurable, rates of proliferation. CECs at the edge of a wound had higher rates of proliferation, probably due to the release of contact inhibition. TTHX1114 increased proliferation at wound edges. After 7 days of culture, proliferating CECs formed contiguous groups of labeled cells that did not migrate away from one another. TTHX1114-treated cells, including the EdU labeled proliferating cells, retained normal morphology, including cell/cell junction ZO-1 staining. Conclusions: Proliferation of CECs in organ-cultured corneas is low, but can be stimulated by wounding or by the administration of TTHX1114 with the effects of each being additive. The CEC monolayer appears to have a population of progenitor cells that are susceptible to stimulation.
Collapse
Affiliation(s)
- David Eveleth
- Trefoil Therapeutics, Inc., San Diego, California, USA
| | - Sarah Pizzuto
- Trefoil Therapeutics, Inc., San Diego, California, USA
| | - Jessica Weant
- Trefoil Therapeutics, Inc., San Diego, California, USA
| | | | | |
Collapse
|
27
|
Phenotypic and functional characterization of corneal endothelial cells during in vitro expansion. Sci Rep 2020; 10:7402. [PMID: 32366916 PMCID: PMC7198491 DOI: 10.1038/s41598-020-64311-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
The advent of cell culture-based methods for the establishment and expansion of human corneal endothelial cells (CEnC) has provided a source of transplantable corneal endothelium, with a significant potential to challenge the one donor-one recipient paradigm. However, concerns over cell identity remain, and a comprehensive characterization of the cultured CEnC across serial passages has not been performed. To this end, we compared two established CEnC culture methods by assessing the transcriptomic changes that occur during in vitro expansion. In confluent monolayers, low mitogenic culture conditions preserved corneal endothelial cell state identity better than culture in high mitogenic conditions. Expansion by continuous passaging induced replicative cell senescence. Transcriptomic analysis of the senescent phenotype identified a cell senescence signature distinct for CEnC. We identified activation of both classic and new cell signaling pathways that may be targeted to prevent senescence, a significant barrier to realizing the potential clinical utility of in vitro expansion.
Collapse
|
28
|
Alió Del Barrio JL, Chung DD, Al-Shymali O, Barrington A, Jatavallabhula K, Swamy VS, Yébana P, Angélica Henríquez-Recine M, Boto-de-Los-Bueis A, Alió JL, Aldave AJ. Punctiform and Polychromatic Pre-Descemet Corneal Dystrophy: Clinical Evaluation and Identification of the Genetic Basis. Am J Ophthalmol 2020; 212:88-97. [PMID: 31782998 DOI: 10.1016/j.ajo.2019.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE This study reports the clinical features and genetic bases of 3 previously unreported families with punctiform and polychromatic pre-Descemet corneal dystrophy (PPPCD). DESIGN Observational case series. METHODS Full ophthalmic assessment was performed for members of 3 unreported families with PPPCD. Structural and biomechanical alterations of the cornea were screened. Whole exome sequencing (WES) was performed in the first family. Novel or rare variants that segregated with the affected status were screened in the other 2 families using Sanger sequencing. Identified variants that segregated with the affected status in all families were characterized by using in silico prediction tools and/or in vitro splice assays. Additionally, 2 previously reported PPPCD families were screened for variants identified in the 3 unreported PPPCD families. RESULTS PPPCD was diagnosed in 12 of the 21 examined members of the 3 unreported families. The only refractive, topographic, or biomechanical abnormality associated with PPPCD was a significantly increased corneal stiffness. WES and Sanger sequencing identified 2 variants that segregated with the affected status in all 3 families: a rare intronic PDZD8 c.872+10A>T variant and a novel missense PRDX3 c.568G>C (p.Asp190His) variant. The same PRDX3 variant was identified in the previously reported PPPCD family expressing the common PPPCD phenotype and was predicted by in silico prediction tools to be damaging to protein function. CONCLUSIONS PPPCD is associated with an alteration of corneal biomechanics and a novel missense variant in PRDX3. Screening of additional families will determine whether all families demonstrate a PRDX3 variant or whether locus heterogeneity may exist for PPPCD.
Collapse
Affiliation(s)
- Jorge L Alió Del Barrio
- Cornea, Cataract and Refractive Surgery Unit, Vissum Corporación, Alicante, Spain; Division of Ophthalmology, School of Medicine, Universidad Miguel Hernández, Alicante, Spain
| | - Doug D Chung
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Olena Al-Shymali
- Cornea, Cataract and Refractive Surgery Unit, Vissum Corporación, Alicante, Spain
| | - Alice Barrington
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Kavya Jatavallabhula
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Vinay S Swamy
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Pilar Yébana
- Cornea, Cataract and Refractive Surgery Unit, Vissum Corporación, Alicante, Spain
| | | | | | - Jorge L Alió
- Cornea, Cataract and Refractive Surgery Unit, Vissum Corporación, Alicante, Spain; Division of Ophthalmology, School of Medicine, Universidad Miguel Hernández, Alicante, Spain
| | - Anthony J Aldave
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
29
|
Wahlig S, Peh GSL, Adnan K, Ang HP, Lwin CN, Morales-Wong F, Ong HS, Lovatt M, Mehta JS. Optimisation of Storage and Transportation Conditions of Cultured Corneal Endothelial Cells for Cell Replacement Therapy. Sci Rep 2020; 10:1681. [PMID: 32015414 PMCID: PMC6997453 DOI: 10.1038/s41598-020-58700-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/20/2020] [Indexed: 12/29/2022] Open
Abstract
As the cornea is one of the most transplanted tissues in the body it has placed a burden on the provision of corneas from cadaveric donors. Corneal endothelial dysfunction is the leading indication for cornea transplant. Therefore, tissue engineering is emerging as an alternative approach to overcome the global shortage of transplant-grade corneas. The propagation and expansion of corneal endothelial cells has been widely reported. However, one obstacle to overcome is the transport and storage of corneal endothelial cells. In this study we investigated whether tissue engineered corneal endothelial cells can be preserved in hypothermic conditions. Human corneal endothelial cells (HCEnCs) were exposed to various temperatures (4 °C, 23 °C, and 37 °C) in both adherent and suspension storage models. Optimal storage media and storage duration was tested along with post-storage viability. Following storage and subsequent recovery at 37 °C, cell phenotype was assessed by immunofluorescence, gene and protein expression, and proliferative capacity analysis. Functionality was also assessed within a rabbit model of bullous keratopathy. Our data support our hypothesis that functional HCEnCs can be preserved in hypothermic conditions.
Collapse
Affiliation(s)
- Stephen Wahlig
- Duke University School of Medicine, Durham, NC, USA.,Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Gary S L Peh
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Khadijah Adnan
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Heng-Pei Ang
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Chan N Lwin
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore
| | - F Morales-Wong
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore.,Singapore National Eye Centre, Singapore, Singapore.,Autonomous University of Nuevo Leon (UANL), University Hospital, Monterrey, Mexico
| | - Hon Shing Ong
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore.,Singapore National Eye Centre, Singapore, Singapore
| | - Matthew Lovatt
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore. .,Duke-NUS Graduate Medical School, Singapore, Singapore.
| | - Jodhbir S Mehta
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore, Singapore. .,Duke-NUS Graduate Medical School, Singapore, Singapore. .,Singapore National Eye Centre, Singapore, Singapore. .,School of Material Science and Engineering, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
30
|
Abstract
Penetrating keratoplasty was the only therapeutic choice for the treatment of corneal endothelial decompensation until the introduction of evolutional endothelial keratoplasties, namely Descemet's stripping automated endothelial keratoplasty (DSAEK) and Descemet's membrane endothelial keratoplasty (DMEK). Although now in widespread use, DSAEK and DMEK still have associated problems, such as difficulty of the surgical technique, acute and chronic cell loss, and shortage of donor corneas. Therefore, regeneration of the corneal endothelium by tissue engineering techniques is being researched to overcome these problems. The concept of transplantation of cultured corneal endothelial cells (CECs) was proposed in the 1970s. However, cultivation of human CECs (HCECs) in sufficient quantity and with acceptable quality for clinical use has proven surprisingly difficult, and the development of methods for transplanting cultured HCECs has been necessary. Numerous research groups have developed culture protocols and techniques that are now bringing corneal endothelial regeneration closer to real-world therapy. For instance, we started a clinical trial in 2013 involving the injection of cultured HCECs into the anterior chamber of patients with corneal endothelial decompensation. This review outlines the rapid progression of this research field, including clinical trial results, and is also intended to identify topics that still require further research or discussion.
Collapse
Affiliation(s)
- Naoki Okumura
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Noriko Koizumi
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| |
Collapse
|
31
|
Swamy V, McGaughey D. Eye in a Disk: eyeIntegration Human Pan-Eye and Body Transcriptome Database Version 1.0. Invest Ophthalmol Vis Sci 2019; 60:3236-3246. [PMID: 31343654 PMCID: PMC6660187 DOI: 10.1167/iovs.19-27106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose We develop an accessible and reliable RNA sequencing (RNA-seq) transcriptome database of healthy human eye tissues and a matching reactive web application to query gene expression in eye and body tissues. Methods We downloaded the raw sequence data for 1375 RNA-seq samples across 54 tissues in the Genotype-Tissue Expression (GTEx) project as a noneye reference set. We then queried several public repositories to find all healthy, nonperturbed, human eye-related tissue RNA-seq samples. The 916 eye and 1375 GTEx samples were sent into a Snakemake-based reproducible pipeline we wrote to quantify all known transcripts and genes, removes samples with poor sequence quality and mislabels, normalizes expression values across each tissue, perform 882 differential expression tests, calculate GO term enrichment, and output all as a single SQLite database file: the Eye in a Disk (EiaD) dataset. Furthermore, we rewrote the web application eyeIntegration (available in the public domain at https://eyeIntegration.nei.nih.gov) to display EiaD. Results The new eyeIntegration portal provides quick visualization of human eye-related transcriptomes published to date by database version, gene/transcript, 19 eye tissues, and 54 body tissues. As a test of the value of this unified pan-eye dataset, we showed that fetal and organoid retina are highly similar at a pan-transcriptome level, but display distinct differences in certain pathways and gene families, such as protocadherin and HOXB family members. Conclusions The eyeIntegration v1.0 web app serves the pan-human eye and body transcriptome dataset, EiaD. This offers the eye community a powerful and quick means to test hypotheses on human gene and transcript expression across 54 body and 19 eye tissues.
Collapse
Affiliation(s)
- Vinay Swamy
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - David McGaughey
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
32
|
Homeostasis of SLC4A11 protein is mediated by endoplasmic reticulum-associated degradation. Exp Eye Res 2019; 188:107782. [DOI: 10.1016/j.exer.2019.107782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/17/2019] [Accepted: 08/28/2019] [Indexed: 01/12/2023]
|
33
|
ZEB1 insufficiency causes corneal endothelial cell state transition and altered cellular processing. PLoS One 2019; 14:e0218279. [PMID: 31194824 PMCID: PMC6564028 DOI: 10.1371/journal.pone.0218279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
The zinc finger e-box binding homeobox 1 (ZEB1) transcription factor is a master regulator of the epithelial to mesenchymal transition (EMT), and of the reverse mesenchymal to epithelial transition (MET) processes. ZEB1 plays an integral role in mediating cell state transitions during cell lineage specification, wound healing and disease. EMT/MET are characterized by distinct changes in molecular and cellular phenotype that are generally context-independent. Posterior polymorphous corneal dystrophy (PPCD), associated with ZEB1 insufficiency, provides a new biological context in which to understand and evaluate the classic EMT/MET paradigm. PPCD is characterized by a cadherin-switch and transition to an epithelial-like transcriptomic and cellular phenotype, which we study in a cell-based model of PPCD generated using CRISPR-Cas9-mediated ZEB1 knockout in corneal endothelial cells (CEnCs). Transcriptomic and functional studies support the hypothesis that CEnC undergo a MET-like transition in PPCD, termed endothelial to epithelial transition (EnET), and lead to the conclusion that EnET may be considered a corollary to the classic EMT/MET paradigm.
Collapse
|
34
|
Bryan JM, Fufa TD, Bharti K, Brooks BP, Hufnagel RB, McGaughey DM. Identifying core biological processes distinguishing human eye tissues with precise systems-level gene expression analyses and weighted correlation networks. Hum Mol Genet 2019; 27:3325-3339. [PMID: 30239781 DOI: 10.1093/hmg/ddy239] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
The human eye is built from several specialized tissues which direct, capture and pre-process information to provide vision. The gene expression of the different eye tissues has been extensively profiled with RNA-seq across numerous studies. Large consortium projects have also used RNA-seq to study gene expression patterning across many different human tissues, minus the eye. There has not been an integrated study of expression patterns from multiple eye tissues compared with other human body tissues. We have collated all publicly available healthy human eye RNA-seq datasets as well as dozens of other tissues. We use this fully integrated dataset to probe the biological processes and pan expression relationships between the cornea, retina, retinal pigment epithelium (RPE)-choroid complex, and the rest of the human tissues with differential expression, clustering and gene ontology term enrichment tools. We also leverage our large collection of retina and RPE-choroid tissues to build the first human weighted gene correlation networks and use them to highlight known biological pathways and eye gene disease enrichment. We also have integrated publicly available single-cell RNA-seq data from mouse retina into our framework for validation and discovery. Finally, we make all these data, analyses and visualizations available via a powerful interactive web application (https://eyeintegration.nei.nih.gov/).
Collapse
Affiliation(s)
- John M Bryan
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Temesgen D Fufa
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kapil Bharti
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - David M McGaughey
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Owen N, Moosajee M. RNA-sequencing in ophthalmology research: considerations for experimental design and analysis. Ther Adv Ophthalmol 2019; 11:2515841419835460. [PMID: 30911735 PMCID: PMC6421592 DOI: 10.1177/2515841419835460] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
High-throughput, massively parallel sequence analysis has revolutionized the way that researchers design and execute scientific investigations. Vast amounts of sequence data can be generated in short periods of time. Regarding ophthalmology and vision research, extensive interrogation of patient samples for underlying causative DNA mutations has resulted in the discovery of many new genes relevant to eye disease. However, such analysis remains functionally limited. RNA-sequencing accurately snapshots thousands of genes, capturing many subtypes of RNA molecules, and has become the gold standard for transcriptome gene expression quantification. RNA-sequencing has the potential to advance our understanding of eye development and disease; it can reveal new candidates to improve our molecular diagnosis rates and highlight therapeutic targets for intervention. But with a wide range of applications, the design of such experiments can be problematic, no single optimal pipeline exists, and therefore, several considerations must be undertaken for optimal study design. We review the key steps involved in RNA-sequencing experimental design and the downstream bioinformatic pipelines used for differential gene expression. We provide guidance on the application of RNA-sequencing to ophthalmology and sources of open-access eye-related data sets.
Collapse
Affiliation(s)
- Nicholas Owen
- Development, Ageing and Disease Theme, UCL Institute of Ophthalmology, University College London, London, UK
| | | |
Collapse
|
36
|
Ali M, Khan SY, Vasanth S, Ahmed MR, Chen R, Na CH, Thomson JJ, Qiu C, Gottsch JD, Riazuddin SA. Generation and Proteome Profiling of PBMC-Originated, iPSC-Derived Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2019; 59:2437-2444. [PMID: 29847650 PMCID: PMC5957521 DOI: 10.1167/iovs.17-22927] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Corneal endothelial cells (CECs) are critical in maintaining clarity of the cornea. This study was initiated to develop peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cell (iPSC)-derived CECs. Methods We isolated PBMCs and programmed the mononuclear cells to generate iPSCs, which were differentiated to CECs through the neural crest cells (NCCs). The morphology of differentiating iPSCs was examined at regular intervals by phase contrast microscopy. In parallel, the expression of pluripotent and corneal endothelium (CE)-associated markers was investigated by quantitative real-time PCR (qRT-PCR). The molecular architecture of the iPSC-derived CECs and human corneal endothelium (hCE) was examined by mass spectrometry–based proteome sequencing. Results The PBMC-originated, iPSC-derived CECs were tightly adherent, exhibiting a hexagonal-like shape, one of the cardinal characteristics of CECs. The CE-associated markers expressed at significantly higher levels in iPSC-derived CECs at days 13, 20, and 30 compared with their respective levels in iPSCs. It is of importance that only residual expression levels of pluripotency markers were detected in iPSC-derived CECs. Cryopreservation of iPSC-derived CECs did not affect the tight adherence of CECs and their hexagonal-like shape while expressing high levels of CE-associated markers. Mass spectrometry–based proteome sequencing identified 10,575 proteins in the iPSC-derived CEC proteome. In parallel, we completed proteome profiling of the hCE identifying 6345 proteins. Of these, 5763 proteins were identified in the iPSC-derived CECs, suggesting that 90.82% of the hCE proteome overlaps with the iPSC-derived CEC proteome. Conclusions We have successfully developed a personalized approach to generate CECs that closely mimic the molecular architecture of the hCE. To the best of our knowledge, this is the first report describing the development of PBMC-originated, iPSC-derived CECs.
Collapse
Affiliation(s)
- Muhammad Ali
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Shahid Y Khan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Shivakumar Vasanth
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Mariya R Ahmed
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Ruiqiang Chen
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Chan Hyun Na
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jason J Thomson
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Caihong Qiu
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, United States
| | - John D Gottsch
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - S Amer Riazuddin
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
37
|
Kruse M, Walter P, Bauer B, Rütten S, Schaefer K, Plange N, Gries T, Jockenhoevel S, Fuest M. Electro-spun Membranes as Scaffolds for Human Corneal Endothelial Cells. Curr Eye Res 2019; 43:1-11. [PMID: 29281419 DOI: 10.1080/02713683.2017.1377258] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Corneal endothelial dysfunction remains the most frequent indication for corneal transplantation, limited by donor material shortage, poor long-term graft survival, or allogeneic graft rejection. Therefore, tissue-engineered endothelial grafts (TEEG) represent a promising alternative to human donor tissue. In this study, we generated electro-spun scaffolds and tested these for their suitability for human corneal endothelial cell (hCEC) cultivation. METHODS The polymers poly(methyl-methacrylate) (PMMA), poly(lactic-co-glycolic acid) (PLGA), and polycaprolactone (PCL) were spun with equal parameters. HCEC-12 was cultured on the scaffolds for 3 to 7 days. Scaffolds were evaluated by light microscopy, porometry, light transmission, scanning electron microscopy (SEM), live/dead staining and cell viability assay. RESULTS Electro-spun fibers from PMMA (2.99 ± 0.24 µm) showed significantly higher diameters than PCL (2.29 ± 0.11 µm; p = 0.003) and PLGA (1.84 ± 0.21 µm; p < 0.001), while fibers from PCL also showed larger diameters than those from PLGA (p = 0.002). PMMA scaffolds (26.77 ± 17.48 µm) had significantly larger interstitial spaces than those from PCL (13.30 ± 5.47 µm; p = 0.04) and PLGA (10.42 ± 6.15 µm; p = 0.002), while PCL and PLGA did not differ significantly (p = 0.26). SEM analysis revealed that only PLGA fibers preserved a normal HCEC-12 morphology. PLGA and PCL did not differ in cell number, death, or viability after 7 days of HCEC-12 cultivation. PMMA showed significantly higher cytotoxicity (p < 0.001; PLGA: 1626.2 ± 183.8 RLU; PMMA: 841.9 ± 92.7 RLU; PCL: 1580.2 ± 171.02 RLU). CONCLUSIONS The biodegradable PLGA and PCL electro-spun scaffolds resulted in equal biocompatibility, while PMMA showed cytotoxicity. Only PLGA preserved hCEC morphology and consequently seems to be a promising candidate for TEEG construction.
Collapse
Affiliation(s)
- Magnus Kruse
- a Department of Biohybrid & Medical Textiles (BioTex) , AME-Helmholtz Institute for Biomedical Engineering & ITA-Institut für Textiltechnik Aachen, RWTH Aachen University , Aachen , Germany
| | - Peter Walter
- b Department of Ophthalmology , RWTH Aachen University , Aachen , Germany
| | - Benedict Bauer
- a Department of Biohybrid & Medical Textiles (BioTex) , AME-Helmholtz Institute for Biomedical Engineering & ITA-Institut für Textiltechnik Aachen, RWTH Aachen University , Aachen , Germany
| | - Stephan Rütten
- c Department of Electron Microscopy , University Hospital RWTH , Aachen , Germany
| | - Karola Schaefer
- d DWI - Leibniz Institute for Interactive Materials e.V. and Institute of Technical and Macromolecular Chemistry (ITMC) , RWTH Aachen University , Aachen , Germany
| | - Niklas Plange
- b Department of Ophthalmology , RWTH Aachen University , Aachen , Germany
| | - Thomas Gries
- a Department of Biohybrid & Medical Textiles (BioTex) , AME-Helmholtz Institute for Biomedical Engineering & ITA-Institut für Textiltechnik Aachen, RWTH Aachen University , Aachen , Germany
| | - Stefan Jockenhoevel
- a Department of Biohybrid & Medical Textiles (BioTex) , AME-Helmholtz Institute for Biomedical Engineering & ITA-Institut für Textiltechnik Aachen, RWTH Aachen University , Aachen , Germany
| | - Matthias Fuest
- b Department of Ophthalmology , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
38
|
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]
|
39
|
Van den Bogerd B, Ní Dhubhghaill S, Zakaria N. Characterizing human decellularized crystalline lens capsules as a scaffold for corneal endothelial tissue engineering. J Tissue Eng Regen Med 2018; 12:e2020-e2028. [PMID: 29430874 PMCID: PMC5947733 DOI: 10.1002/term.2633] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 11/07/2017] [Accepted: 12/11/2017] [Indexed: 12/13/2022]
Abstract
The idea of transplanting a sheet of laboratory-grown corneal endothelium dates back to 1978; however, the ideal scaffold is still lacking. We hypothesized that human crystalline lens capsules (LCs) could qualify as a scaffold and aimed to characterize the properties of this material for endothelial tissue engineering. LCs were isolated from donor eyes, stored at -80 °C, and decellularized with water and trypsin-EDTA. The decellularization was investigated by nuclear staining and counting and the capsule thickness was determined by optical coherence tomography and compared with Descemet's membrane (DM). Transparency was examined by spectrometry, and collagenase degradation was performed to evaluate its resistance to degradation. Cell-scaffold interaction was assessed by measuring focal adhesions surface area on LC and plastic. Finally, primary corneal endothelial cells were grown on LCs to validate the phenotype. Trypsin-EDTA decellularized most effectively, removing 99% of cells. The mean LC thickness was 35.76 ± 0.43 μm, whereas DM measured 25.93 ± 0.26 μm (p < .0001). Light transmission was 90% for both LC and DM. On a collagenase challenge, LC and amniotic membrane were digested after 13 hr, whereas DM was digested after 17 hr. The surface area of focal adhesions for cells grown on coated LCs was at least double that compared with other conditions, whereas tight junctions, ion pumps, and hexagonal morphology were well maintained when endothelial cells were cultured on LCs. In conclusion, LCs demonstrate excellent scaffolding properties for tissue engineering and sustain the cell phenotype and can be considered a suitable substrate for ocular tissue engineering or as a template for future scaffolds.
Collapse
Affiliation(s)
- Bert Van den Bogerd
- Ophthalmology, Visual Optics and Visual Rehabilitation, Translational Neurosciences, Faculty of MedicineUniversity of AntwerpWilrijkBelgium
| | - Sorcha Ní Dhubhghaill
- Ophthalmology, Visual Optics and Visual Rehabilitation, Translational Neurosciences, Faculty of MedicineUniversity of AntwerpWilrijkBelgium
- Department of OphthalmologyAntwerp University HospitalEdegemBelgium
| | - Nadia Zakaria
- Ophthalmology, Visual Optics and Visual Rehabilitation, Translational Neurosciences, Faculty of MedicineUniversity of AntwerpWilrijkBelgium
- Department of OphthalmologyAntwerp University HospitalEdegemBelgium
- Centre for Cell Therapy and Regenerative MedicineAntwerp University HospitalEdegemBelgium
| |
Collapse
|
40
|
Zakharevich M, Kattan JM, Chen JL, Lin BR, Cervantes AE, Chung DD, Frausto RF, Aldave AJ. Elucidating the molecular basis of PPCD: Effects of decreased ZEB1 expression on corneal endothelial cell function. Mol Vis 2017; 23:740-752. [PMID: 29046608 PMCID: PMC5644665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/12/2017] [Indexed: 11/01/2022] Open
Abstract
PURPOSE To investigate the functional role that the zinc e-box binding homeobox 1 (ZEB1) gene, which underlies the genetic basis of posterior polymorphous corneal dystrophy 3 (PPCD3), plays in corneal endothelial cell proliferation, apoptosis, migration, and barrier function. METHODS A human corneal endothelial cell line (HCEnC-21T) was transfected with siRNA targeting ZEB1 mRNA. Cell proliferation, apoptosis, migration, and barrier assays were performed: Cell proliferation was assessed with cell counting using a hemocytometer; cell apoptosis, induced by either ultraviolet C (UVC) radiation or doxorubicin treatment, was quantified by measuring cleaved caspase 3 (cCASP3) protein levels; and cell migration and barrier function were monitored with electric cell-substrate impedance sensing (ECIS). RESULTS ZEB1 knockdown in HCEnC-21T cells transfected with siRNA targeting ZEB1 did not result in a significant difference in cell proliferation when compared with control. Although knockdown of ZEB1 in HCEnC-21T cells sensitized the cells to UV-induced apoptosis, ZEB1 knockdown did not alter the cells' susceptibility to doxorubicin-induced apoptosis, as measured with cCASP3 protein levels, compared with controls. Similarly, no difference was observed in cell migration following ZEB1 knockdown. However, cell barrier function increased significantly following ZEB1 knockdown. CONCLUSIONS The corneal endothelium in PPCD3 is characterized by morphologic, anatomic, and molecular features that are more consistent with an epithelial-like rather than an endothelial-like phenotype. Although these characteristics have been well documented, we demonstrate for the first time that susceptibility to UV-induced apoptosis and cell barrier function are significantly altered in the setting of reduced ZEB1. The significance of an altered cellular response to apoptotic stimuli and increased cell barrier function in the pathobiology of PPCD remains to be fully elucidated.
Collapse
|
41
|
Chung DD, Frausto RF, Lin BR, Hanser EM, Cohen Z, Aldave AJ. Transcriptomic Profiling of Posterior Polymorphous Corneal Dystrophy. Invest Ophthalmol Vis Sci 2017; 58:3202-3214. [PMID: 28654985 PMCID: PMC5488878 DOI: 10.1167/iovs.17-21423] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose To investigate the molecular basis of posterior polymorphous corneal dystrophy (PPCD) by examining the PPCD transcriptome and the effect of decreased ZEB1 expression on corneal endothelial cell (CEnC) gene expression. Methods Next-generation RNA sequencing (RNA-seq) analyses of corneal endothelium from two PPCD-affected individuals (one with PPCD3 and one of unknown genetic cause) compared with two age-matched controls, and primary human CEnC (pHCEnC) transfected with siRNA-mediated ZEB1 knockdown. The expression of selected differentially expressed genes was validated by quantitative polymerase chain reaction (qPCR) and/or assessed by in situ hybridization in the corneal endothelium of four independent cases of PPCD (one with PPCD3 and three of unknown genetic cause). Results Expression of 16% and 46% of the 104 protein-coding genes specific to ex vivo corneal endothelium was lost in the endothelium of two individuals with PPCD. Thirty-two genes associated with ZEB1 and 3 genes (BMP4, CCND1, ZEB1) associated with OVOL2 were differentially expressed in the same direction in both individuals with PPCD. Immunohistochemistry staining and RNA-seq analyses demonstrated variable expression of type IV collagens in PPCD corneas. Decreasing ZEB1 expression in pHCEnC altered expression of 711 protein-coding genes, many of which are associated with canonical pathways regulating various cellular processes. Conclusions Identification of the altered transcriptome in PPCD and in a cell-based model of PPCD provided insight into the molecular alterations characterizing PPCD. Further study of the differentially expressed genes associated with ZEB1 and OVOL2 is expected to identify candidate genes for individuals with PPCD and without a ZEB1 or OVOL2 mutation.
Collapse
Affiliation(s)
- Doug D Chung
- Stein Eye Institute, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | - Ricardo F Frausto
- Stein Eye Institute, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | - Benjamin R Lin
- Stein Eye Institute, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | - Evelyn M Hanser
- Stein Eye Institute, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | - Zack Cohen
- Stein Eye Institute, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | - Anthony J Aldave
- Stein Eye Institute, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| |
Collapse
|
42
|
Nagymihály R, Veréb Z, Albert R, Sidney L, Dua H, Hopkinson A, Petrovski G. Cultivation and characterisation of the surface markers and carbohydrate profile of human corneal endothelial cells. Clin Exp Ophthalmol 2017; 45:509-519. [PMID: 28032398 DOI: 10.1111/ceo.12903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 11/16/2016] [Accepted: 12/08/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND The study aims to characterise human corneal endothelial cell (HCEnC) cultures generated by the peel-and-digest method based on their surface protein/carbohydrate expression pattern. METHODS Quantitative polymerase chain reaction was used to compare expression of vimentin, CD90, Cytokeratin-19, ZO-1 and Claudin 14 in cultured HCEnC and cell line B4G12 versus stromal cells. Fluorescence-activated cell sorting was used to assess surface protein distribution of cultured and uncultured HCEnC. Distribution of surface proteins/carbohydrates was visualised by immunofluorescent and lectin staining. RESULTS Human corneal endothelial cell and B4G12 showed lower expression level for vimentin, CD90, Cytokeratin-19 compared with stromal cells; while ZO-1 was expressed in endothelial cells, Claudin 14 was detected in B4G12 only. Fluorescence-activated cell sorting analyses revealed CD166, CD47, CD44, CD54, CD73, CD90, CD105, CD106, CD112, CD146 and CD325 to be present, with CD34 to be absent from cultured HCEnC. Freshly isolated, non-cultivated HCEnCs were CD90, CD73, CD146 and CD325 positive. Carbohydrates were detected by lectins LCA, PHA E, PHA L, PSA, sWGA, Con A, RCA 120 and WGA, but cultured HCEnC showed negative for GSL I, SBA, DBA, PNA and UEA I. CONCLUSION Cultures established by the peel-and-digest method are probably not prone to stromal contamination, but the cells are likely to undergo endothelial-to mesenchymal transition as suggested by apparent morphological changes.
Collapse
Affiliation(s)
- Richárd Nagymihály
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Zoltán Veréb
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Réka Albert
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Laura Sidney
- Academic Department of Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Harminder Dua
- Academic Department of Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Andrew Hopkinson
- Academic Department of Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Goran Petrovski
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Center for Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
43
|
Rizwan M, Peh GS, Adnan K, Naso SL, Mendez AR, Mehta JS, Yim EKF. In Vitro Topographical Model of Fuchs Dystrophy for Evaluation of Corneal Endothelial Cell Monolayer Formation. Adv Healthc Mater 2016; 5:2896-2910. [PMID: 27701826 DOI: 10.1002/adhm.201600848] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Indexed: 12/13/2022]
Abstract
A common indication for corneal transplantation, which is the most transplanted tissue, is a dysfunctional corneal endothelium due to Fuchs' endothelial dystrophy (FED). FED is diagnosed by the presence of in vivo pathological microtopography on the Descemet membrane, which is called corneal guttata. Minimally invasive corneal endothelial cell regenerative procedures such as endothelial cell injection therapy and Rho kinase inhibitor pharmacotherapy have been proposed as alternatives to conventional corneal transplantation for FED patients. However, the effect of guttata on monolayer reformation following such therapies is unknown and there is no equivalent in vitro or animal model to study monolayer reformation. Using a synthetic guttata FED disease model, the formation of the monolayer is investigated to evaluate the efficacy of both therapies. Results obtained suggest that guttata dimensions, density, and spacing greatly affect the fate of corneal endothelial cells in terms of migratory behavior and monolayer reformation. Densely packed synthetic guttata mimicking late-stage FED hinders monolayer reformation, while synthetic guttata of lower height and density show improved monolayer formation. These results suggest that severity of the FED, as determined by height and density of existing guttata, can potentially attenuate corneal endothelial monolayer formation of corneal cell injection therapy and pharmacotherapy.
Collapse
Affiliation(s)
- Muhammad Rizwan
- Department of Biomedical Engineering; National University of Singapore; Singapore 117583
- Institute of Materials Research and Engineering; Agency for Science; Technology and Research (A*STAR); Singapore 138634
| | - Gary S. Peh
- Tissue Engineering and Stem Cell Group; Singapore Eye Research Institute; Singapore 169856
- Duke-NUS Graduate Medical School; Singapore 169857
| | - Khadijah Adnan
- Tissue Engineering and Stem Cell Group; Singapore Eye Research Institute; Singapore 169856
| | - Sacha L. Naso
- Tissue Engineering and Stem Cell Group; Singapore Eye Research Institute; Singapore 169856
| | - Alon R. Mendez
- Department of Biomedical Engineering; National University of Singapore; Singapore 117583
| | - Jodhbir S. Mehta
- Duke-NUS Graduate Medical School; Singapore 169857
- Singapore National Eye Centre; Singapore 168751
| | - Evelyn K. F. Yim
- Department of Biomedical Engineering; National University of Singapore; Singapore 117583
- Department of Surgery; National University of Singapore, Singapore; Mechanobiology Institute; National University of Singapore; Singapore 117411
- Department of Chemical Engineering; University of Waterloo; Waterloo Ontario Canada N2L 3G1
| |
Collapse
|
44
|
Kao L, Azimov R, Shao XM, Frausto RF, Abuladze N, Newman D, Aldave AJ, Kurtz I. Multifunctional ion transport properties of human SLC4A11: comparison of the SLC4A11-B and SLC4A11-C variants. Am J Physiol Cell Physiol 2016; 311:C820-C830. [PMID: 27581649 DOI: 10.1152/ajpcell.00233.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Congenital hereditary endothelial dystrophy (CHED), Harboyan syndrome (CHED with progressive sensorineural deafness), and potentially a subset of individuals with late-onset Fuchs' endothelial corneal dystrophy are caused by mutations in the SLC4A11 gene that results in corneal endothelial cell abnormalities. Originally classified as a borate transporter, the function of SLC4A11 as a transport protein remains poorly understood. Elucidating the transport function(s) of SLC4A11 is needed to better understand how its loss results in the aforementioned posterior corneal dystrophic disease processes. Quantitative PCR experiments demonstrated that, of the three known human NH2-terminal variants, SLC4A11-C is the major transcript expressed in human corneal endothelium. We studied the expression pattern of the three variants in mammalian HEK-293 cells and demonstrated that the SLC4A11-B and SLC4A11-C variants are plasma membrane proteins, whereas SLC4A11-A is localized intracellularly. SLC4A11-B and SLC4A11-C were shown to be multifunctional ion transporters capable of transporting H+ equivalents in both a Na+-independent and Na+-coupled mode. In both transport modes, SLC4A11-C H+ flux was significantly greater than SLC4A11-B. In the presence of ammonia, SLC4A11-B and SLC4A11-C generated inward currents that were comparable in magnitude. Chimera SLC4A11-C-NH2-terminus-SLC4A11-B experiments demonstrated that the SLC4A11-C NH2-terminus functions as an autoactivating domain, enhancing Na+-independent and Na+-coupled H+ flux without significantly affecting the electrogenic NH3-H(n)+ cotransport mode. All three modes of transport were significantly impaired in the presence of the CHED causing p.R109H (SLC4A11-C numbering) mutation. These complex ion transport properties need to be addressed in the context of corneal endothelial disease processes caused by mutations in SLC4A11.
Collapse
Affiliation(s)
- Liyo Kao
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Rustam Azimov
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Xuesi M Shao
- Department of Neurobiology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ricardo F Frausto
- Stein Eye Institute, and.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Natalia Abuladze
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Debra Newman
- Division of Nephrology.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Anthony J Aldave
- Stein Eye Institute, and.,David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ira Kurtz
- Division of Nephrology, .,Brain Research Institute.,David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
45
|
Chung DWD, Frausto RF, Chiu S, Lin BR, Aldave AJ. Investigating the Molecular Basis of PPCD3: Characterization of ZEB1 Regulation of COL4A3 Expression. Invest Ophthalmol Vis Sci 2016; 57:4136-43. [PMID: 27537263 PMCID: PMC4991021 DOI: 10.1167/iovs.16-19533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/06/2016] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To investigate the role of the zinc finger e-box binding homeobox 1 (ZEB1) transcription factor in posterior polymorphous corneal dystrophy 3 by demonstrating its ability to regulate type IV collagen gene transcription via binding to putative E2 box motifs. METHODS Putative E2 box motifs were identified by in silico analysis within the promoter region of collagen, type IV, alpha3 (COL4A3) and collagen, type IV, alpha4 (COL4A4). To test the ability of ZEB1 to bind to each identified E2 box, electrophoretic mobility shift assays were performed by incubating ZEB1-enriched nuclear extracts with DIG-labeled probes containing one of each of the identified E2 box motifs. Dual-luciferase reporter assays were performed to test the effects of ZEB1 on the luciferase activity of COL4A3 and cadherin 1 (CDH1) promoter constructs, and to determine the effect of a ZEB1 truncating mutation on CDH1 promoter activity. RESULTS ZEB1 exhibited binding to six of the nine COL4A3 E2 box probes, whereas no binding was observed for either of the two COL4A4 E2 box probes. ZEB1 overexpression resulted in reduced activity of the COL4A3 promoter construct containing all identified E2 box motifs, whereas a truncating ZEB1 mutation led to the loss of ZEB1-dependent repression of the CDH1 promoter. CONCLUSIONS COL4A3 gene expression is negatively regulated by ZEB1 binding to E2 box motifs in the COL4A3 promoter region. Therefore, the altered expression of type IV collagens, particularly COL4A3, in the corneal endothelium in individuals with PPCD3 is likely due to reduced transcriptional repression in the setting of a single functional ZEB1 allele.
Collapse
Affiliation(s)
- Duk-Won D. Chung
- Stein Eye Institute David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Ricardo F. Frausto
- Stein Eye Institute David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Stephan Chiu
- Stein Eye Institute David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Benjamin R. Lin
- Stein Eye Institute David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Anthony J. Aldave
- Stein Eye Institute David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| |
Collapse
|
46
|
Le DJ, Chung DWD, Frausto RF, Kim MJ, Aldave AJ. Identification of Potentially Pathogenic Variants in the Posterior Polymorphous Corneal Dystrophy 1 Locus. PLoS One 2016; 11:e0158467. [PMID: 27355326 PMCID: PMC4927100 DOI: 10.1371/journal.pone.0158467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 06/16/2016] [Indexed: 12/13/2022] Open
Abstract
Posterior polymorphous corneal dystrophy 1 (PPCD1) is a genetic disorder that affects corneal endothelial cell function and leads to loss of visual acuity. PPCD1 has been linked to a locus on chromosome 20 in multiple families; however, Sanger sequencing of protein-coding genes in the consensus region failed to identify any causative missense mutations. In this study, custom capture probes were utilized for targeted next-generation sequencing of the linked region in a previously reported family with PPCD1. Variants were detected through two bioinformatics pipelines and filtered according to multiple criteria. Additionally, a high-resolution microarray was used to detect copy number variations. No non-synonymous variants in the protein-coding region of annotated genes were identified. However, 12 single nucleotide variants in 10 genes, and 9 indels in 7 genes met the filtering criteria and were considered candidate variants for PPCD1. Eleven single nucleotide variants were confirmed by Sanger sequencing, including 2 synonymous variants and 9 non-coding variants, in 9 genes. One microdeletion was detected in an intron of OVOL2 by microarray but was subsequently not identified by PCR. Using a comprehensive next-generation sequencing approach, a total of 16 genes containing single nucleotide variants or indels that segregated with the affected phenotype in an affected family previously mapped to the PPCD1 locus were identified. Screening of these candidate genes in other families previously mapped to the PPCD1 locus will likely result in the identification of the genetic basis of PPCD1.
Collapse
Affiliation(s)
- Derek J. Le
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Duk-Won D. Chung
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Ricardo F. Frausto
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Michelle J. Kim
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Anthony J. Aldave
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|