1
|
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
|
2
|
Ning L, Zanella S, Tomov ML, Amoli MS, Jin L, Hwang B, Saadeh M, Chen H, Neelakantan S, Dasi LP, Avazmohammadi R, Mahmoudi M, Bauser‐Heaton HD, Serpooshan V. Targeted Rapamycin Delivery via Magnetic Nanoparticles to Address Stenosis in a 3D Bioprinted in Vitro Model of Pulmonary Veins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400476. [PMID: 38696618 PMCID: PMC11234432 DOI: 10.1002/advs.202400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/09/2024] [Indexed: 05/04/2024]
Abstract
Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.
Collapse
Affiliation(s)
- Liqun Ning
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of Mechanical EngineeringCleveland State UniversityClevelandOH44115USA
| | - Stefano Zanella
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Martin L. Tomov
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Mehdi Salar Amoli
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Linqi Jin
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Boeun Hwang
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Maher Saadeh
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Huang Chen
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Sunder Neelakantan
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lakshmi Prasad Dasi
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Reza Avazmohammadi
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
- J. Mike Walker ’66 Department of Mechanical EngineeringTexas A&M UniversityCollege StationTX77840USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LandingMI48824USA
| | - Holly D. Bauser‐Heaton
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
- Sibley Heart Center at Children's Healthcare of AtlantaAtlantaGA30322USA
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
| |
Collapse
|
3
|
Koo EH. Current state of endothelial cell therapy. Curr Opin Ophthalmol 2024; 35:304-308. [PMID: 38602486 DOI: 10.1097/icu.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Currently, there is heightened interest surrounding endothelial cell therapy for the treatment of corneal edema. The purpose of this review article is to describe and summarize the background information as well as the research surrounding the emerging treatment modalities for endothelial cell therapy. RECENT FINDINGS Marked advancements have been made in the translational research in this area, and increasing refinements have been demonstrated in the treatment protocols for cell therapy. Human clinical trials in this field are ongoing, specifically, in the area of injected human corneal endothelial cells (HCECs), with early results showing favorable safety and efficacy profiles. SUMMARY Efficient and effective delivery of HCECs to patients with corneal edema and dysfunction now appears feasible, and the results from ongoing human clinical trials are much anticipated. Adjunct therapeutics-in the form of pharmacological agents and/or surgical techniques, such as descemetorhexis-will likely continue to play an important role in defining the future of endothelial cell therapy.
Collapse
Affiliation(s)
- Ellen H Koo
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
4
|
Wang R, Wang Z, Tong L, Wang R, Yao S, Chen D, Hu H. Microfluidic Mechanoporation: Current Progress and Applications in Stem Cells. BIOSENSORS 2024; 14:256. [PMID: 38785730 PMCID: PMC11117831 DOI: 10.3390/bios14050256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Intracellular delivery, the process of transporting substances into cells, is crucial for various applications, such as drug delivery, gene therapy, cell imaging, and regenerative medicine. Among the different approaches of intracellular delivery, mechanoporation stands out by utilizing mechanical forces to create temporary pores on cell membranes, enabling the entry of substances into cells. This method is promising due to its minimal contamination and is especially vital for stem cells intended for clinical therapy. In this review, we explore various mechanoporation technologies, including microinjection, micro-nano needle arrays, cell squeezing through physical confinement, and cell squeezing using hydrodynamic forces. Additionally, we highlight recent research efforts utilizing mechanoporation for stem cell studies. Furthermore, we discuss the integration of mechanoporation techniques into microfluidic platforms for high-throughput intracellular delivery with enhanced transfection efficiency. This advancement holds potential in addressing the challenge of low transfection efficiency, benefiting both basic research and clinical applications of stem cells. Ultimately, the combination of microfluidics and mechanoporation presents new opportunities for creating comprehensive systems for stem cell processing.
Collapse
Affiliation(s)
- Rubing Wang
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Haining 314400, China;
| | - Ziqi Wang
- Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; (Z.W.); (L.T.)
| | - Lingling Tong
- Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; (Z.W.); (L.T.)
| | - Ruoming Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), International Campus, Zhejiang University, Haining 314400, China; (R.W.); (S.Y.)
| | - Shuo Yao
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), International Campus, Zhejiang University, Haining 314400, China; (R.W.); (S.Y.)
| | - Di Chen
- Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; (Z.W.); (L.T.)
- Center for Reproductive Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310003, China
- National Key Laboratory of Biobased Transportation Fuel Technology, Haining 314400, China
| | - Huan Hu
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Haining 314400, China;
| |
Collapse
|
5
|
Rowe-Rendleman C, Eveleth D, Goldberg JL, Jurkunas U, Okumura N, Dawson D, Sawant OB. Development of Anterior Segment Focused Biologic Therapies to Regenerate Corneal Tissue for the Treatment of Disease: Drug Development Experience. J Ocul Pharmacol Ther 2023; 39:551-562. [PMID: 37733302 DOI: 10.1089/jop.2023.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023] Open
Abstract
On February 24-27, 2021, the Association for Ocular Pharmacology and Therapeutics (AOPT) held its 15th biennial scientific meeting online. The meeting was organized by Dr. Sanjoy Bhattacharya of the University of Miami in conjunction with the board of trustees of the AOPT. The 3-day conference was attended by academic scientists, clinicians, and industry and regulatory professionals. The theme of the meeting was Restoring Vision through Regeneration and it was sponsored, in part, by the National Institutes of Health, Bright Focus, Regeneron, and Santen (USA). During the 3 days of the meeting, presentations from several sessions explored different aspects of regenerative medicine in ophthalmology, including optic nerve regeneration, drugs and devices in glaucoma, retinal neuroprotection and plasticity, visual perception, and degeneration of trabecular meshwork. This article summarizes the proceedings of the session on corneal regenerative medicine research and discusses emerging concepts in drug development for corneal epithelial and endothelial regeneration. Since the meeting in 2021, several of these concepts have advanced to clinical-stage therapies, but so far as of 2023, none has been approved by regional regulatory authorities in the United States. One form of corneal endothelial cell therapy has been approved in Japan and only for bullous keratopathy. Ongoing work is proceeding in the United States and other countries. Clinical Registration No: National Clinical Trials 04894110, 04812667; Japan Registry for Clinical Trials a031210199.
Collapse
Affiliation(s)
| | | | | | - Ula Jurkunas
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Onkar B Sawant
- Center for Vision and Eye Banking Research, Eversight, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Sasseville S, Karami S, Tchatchouang A, Charpentier P, Anney P, Gobert D, Proulx S. Biomaterials used for tissue engineering of barrier-forming cell monolayers in the eye. Front Bioeng Biotechnol 2023; 11:1269385. [PMID: 37840667 PMCID: PMC10569698 DOI: 10.3389/fbioe.2023.1269385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell monolayers that form a barrier between two structures play an important role for the maintenance of tissue functionality. In the anterior portion of the eye, the corneal endothelium forms a barrier that controls fluid exchange between the aqueous humor of the anterior chamber and the corneal stroma. This monolayer is central in the pathogenesis of Fuchs endothelial corneal dystrophy (FECD). FECD is a common corneal disease, in which corneal endothelial cells deposit extracellular matrix that increases the thickness of its basal membrane (Descemet's membrane), and forms excrescences (guttae). With time, there is a decrease in endothelial cell density that generates vision loss. Transplantation of a monolayer of healthy corneal endothelial cells on a Descemet membrane substitute could become an interesting alternative for the treatment of this pathology. In the back of the eye, the retinal pigment epithelium (RPE) forms the blood-retinal barrier, controlling fluid exchange between the choriocapillaris and the photoreceptors of the outer retina. In the retinal disease dry age-related macular degeneration (dry AMD), deposits (drusen) form between the RPE and its basal membrane (Bruch's membrane). These deposits hinder fluid exchange, resulting in progressive RPE cell death, which in turn generates photoreceptor cell death, and vision loss. Transplantation of a RPE monolayer on a Bruch's membrane/choroidal stromal substitute to replace the RPE before photoreceptor cell death could become a treatment alternative for this eye disease. This review will present the different biomaterials that are proposed for the engineering of a monolayer of corneal endothelium for the treatment of FECD, and a RPE monolayer for the treatment of dry AMD.
Collapse
Affiliation(s)
- Samantha Sasseville
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Samira Karami
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Ange Tchatchouang
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Pascale Charpentier
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Princia Anney
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Delphine Gobert
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre universitaire d’ophtalmologie (CUO), Hôpital du Saint-Sacrement, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Stéphanie Proulx
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
7
|
Park JH, Lee K, Park CY. Effect of Magnetic Microparticles on Cultivated Human Corneal Endothelial Cells. Transl Vis Sci Technol 2023; 12:14. [PMID: 36757343 PMCID: PMC9924430 DOI: 10.1167/tvst.12.2.14] [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] [Indexed: 02/10/2023] Open
Abstract
Purpose To investigate effects of magnetic microparticles on movement of magnet controlled human corneal endothelial cells (HCECs). Methods Immortalized HCEC line (B4G12) and primary culture of HCECs were exposed to two commercially available magnetic micro- or nanoparticles, SiMAG (average size 100 nm) and fluidMAG (average size <1000 nm). Cell viability assays and reactive oxygen species production assays were performed. Cellular structural changes, intracellular distribution of microparticles, and expression levels of proteins related to cellular survival were analyzed. Ex vivo human corneas were exposed to microparticles to further evaluate their effects. Magnetic particle-laden HCECs were cultured under the influence of a neodymium magnet. Results No significant decrease of viability was found in HCECs after exposure to both magnetic particles at concentrations up to 20 µg/mL for 48 hours. However, high concentrations (40 µg/mL and 80 µg/mL) of SiMAG and FluidMAG significantly decreased viability in immortalized HCECs, and only 80 µg/mL of SiMAG and FluidMAG decreased viability in primary HCECs after 48 hours of exposure. There was relative stability of viability at various concentrations of magnetic particles, despite a dose-dependent increase of reactive oxygen species, lactate dehydrogenase, and markers of apoptosis. Ex vivo human cornea study further revealed that exposure to 20 µg/mL of SiMAG and fluidMAG for 72 hours was tolerable. Endocytosed magnetic particles were mainly localized in the cytoplasm. The application of a magnetic field during cell culture successfully demonstrated that magnetic particle-loaded HCECs moved toward the magnet area and that the population density of HCECs was significantly increased. Conclusions We verified short-term effects of SiMAG and fluidMAG on HCECs and their ability to control movement of HCECs by an external magnetic field. Translational Relevance A technology of applying magnetic particles to a human corneal endothelial cell culture and controlling the movement of cells to a desired area using a magnetic field could be used to increase cell density during cell culture or improve the localization of corneal endothelial cells injected into the anterior chamber to the back of the cornea.
Collapse
Affiliation(s)
- Joo-Hee Park
- Department of Ophthalmology, Dongguk University, Ilsan Hospital, Goyang, South Korea
| | - Kangmin Lee
- Department of Ophthalmology, Dongguk University, Ilsan Hospital, Goyang, South Korea
| | - Choul Yong Park
- Department of Ophthalmology, Dongguk University, Ilsan Hospital, Goyang, South Korea
| |
Collapse
|
8
|
Bandeira F, Grottone GT, Covre JL, Cristovam PC, Loureiro RR, Pinheiro FI, Casaroli-Marano RP, Donato W, Gomes JÁP. A Framework for Human Corneal Endothelial Cell Culture and Preliminary Wound Model Experiments with a New Cell Tracking Approach. Int J Mol Sci 2023; 24:ijms24032982. [PMID: 36769303 PMCID: PMC9917640 DOI: 10.3390/ijms24032982] [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: 11/08/2022] [Revised: 01/21/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Cell injection therapy is emerging as an alternative to treat corneal endothelial dysfunction (CED) and to avoid corneal scarring due to bullous keratopathy. However, establishing a standardized culture procedure that provides appropriate cell yield while retaining functional features remains a challenge. Here, we describe a detailed framework obtained from in vitro culture of human corneal endothelial cells (HCECs) and comparative in vivo experimental models for CED treatment with a new cell tracking approach. Two digestion methods were compared regarding HCEC morphology and adhesion. The effect of Y-27632 (ROCKi) supplementation on final cell yield was also assessed. Cell adhesion efficacy with two cell delivery systems (superparamagnetic embedding and cell suspension) was evaluated in an ex vivo human cornea model and in an in vivo rabbit CED model. The injection of supplemented culture medium or balanced salt solution (BSS) was used for the positive and negative controls, respectively. HCEC isolation with collagenase resulted in better morphology and adhesion of cultured HCEC when compared to EDTA. Y-27632 supplementation resulted in a 2.6-fold increase in final cell yield compared to the control. Ex vivo and in vivo adhesion with both cell delivery systems was confirmed by cell tracker fluorescence detection. Corneal edema and opacity improved in both animal groups treated with cultured HCEC. The corneas in the control groups remained opaque. Both HCEC delivery systems seemed comparable as treatments for CED and for the prevention of corneal scarring.
Collapse
Affiliation(s)
- Francisco Bandeira
- Department of Ophthalmology, Federal University of São Paulo, São Paulo 04023-062, Brazil
- Medicine School, Barcelona University, 08007 Barcelona, Spain
- Correspondence: ; Tel.: +55-2197-2355-742
| | | | - Joyce Luciana Covre
- Department of Ophthalmology, Federal University of São Paulo, São Paulo 04023-062, Brazil
| | | | - Renata Ruoco Loureiro
- Department of Ophthalmology, Federal University of São Paulo, São Paulo 04023-062, Brazil
| | - Francisco Irochima Pinheiro
- Biotechnology Post-Graduate Program, Potiguar University, Natal 59082-902, Brazil
- Department of Surgery, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | | | - Waleska Donato
- Department of Ophthalmology, Federal University of São Paulo, São Paulo 04023-062, Brazil
| | | |
Collapse
|
9
|
Chew LA, Jun AS, Barnett BP. Corneal endothelial transplantation from bench to bedside: A review of animal models and their translational value for therapeutic development. Exp Eye Res 2022; 224:109241. [PMID: 36075460 PMCID: PMC10782848 DOI: 10.1016/j.exer.2022.109241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/10/2022] [Accepted: 08/27/2022] [Indexed: 11/23/2022]
Affiliation(s)
- Lindsey A Chew
- Duke University, School of Medicine, 40 Duke Medicine Circle, 124 Davison Building, Durham, NC, 27710, USA
| | - Albert S Jun
- Wilmer Eye Institute, 1800 Orleans St., Baltimore, MD, 21287, USA
| | - Brad P Barnett
- California LASIK & Eye, 1111 Exposition Blvd., Bldg. 200 Ste. 2000, Sacramento, CA, 95815, USA.
| |
Collapse
|
10
|
Delivery of Cells to the Cornea Using Synthetic Biomaterials. Cornea 2022; 41:1325-1336. [DOI: 10.1097/ico.0000000000003094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/24/2022] [Indexed: 11/26/2022]
|
11
|
Chakrabarty P, Gupta P, Illath K, Kar S, Nagai M, Tseng FG, Santra TS. Microfluidic mechanoporation for cellular delivery and analysis. Mater Today Bio 2022; 13:100193. [PMID: 35005598 PMCID: PMC8718663 DOI: 10.1016/j.mtbio.2021.100193] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Highly efficient intracellular delivery strategies are essential for developing therapeutic, diagnostic, biological, and various biomedical applications. The recent advancement of micro/nanotechnology has focused numerous researches towards developing microfluidic device-based strategies due to the associated high throughput delivery, cost-effectiveness, robustness, and biocompatible nature. The delivery strategies can be carrier-mediated or membrane disruption-based, where membrane disruption methods find popularity due to reduced toxicity, enhanced delivery efficiency, and cell viability. Among all of the membrane disruption techniques, the mechanoporation strategies are advantageous because of no external energy source required for membrane deformation, thereby achieving high delivery efficiencies and increased cell viability into different cell types with negligible toxicity. The past two decades have consequently seen a tremendous boost in mechanoporation-based research for intracellular delivery and cellular analysis. This article provides a brief review of the most recent developments on microfluidic-based mechanoporation strategies such as microinjection, nanoneedle arrays, cell-squeezing, and hydroporation techniques with their working principle, device fabrication, cellular delivery, and analysis. Moreover, a brief discussion of the different mechanoporation strategies integrated with other delivery methods has also been provided. Finally, the advantages, limitations, and future prospects of this technique are discussed compared to other intracellular delivery techniques.
Collapse
Affiliation(s)
- Pulasta Chakrabarty
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Srabani Kar
- Department of Electrical Engineering, University of Cambridge, Cambridge, CB30FA, UK
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Aichi, Japan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
12
|
Hatamie S, Shih PJ, Chen BW, Shih HJ, Wang IJ, Young TH, Yao DJ. Effects of Electromagnets on Bovine Corneal Endothelial Cells Treated with Dendrimer Functionalized Magnetic Nanoparticles. Polymers (Basel) 2021; 13:3306. [PMID: 34641122 PMCID: PMC8512180 DOI: 10.3390/polym13193306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/17/2022] Open
Abstract
To improve bovine corneal endothelial cell (BCEC) migration, enhance cell energy, and facilitate symmetric cell distribution in corneal surfaces, an electromagnet device was fabricated. Twenty nanometer superparamagnetic iron oxide nanoparticles (SPIONs) functionalized with fourth-generation dendrimer macromolecules were synthesized, and their size and structure were evaluated using transmission electron microscopy (TEM), X-ray photoelectron spectroscopy (XPS), and Fourier transform infrared spectroscopy (FTIR). The results confirmed the configuration of the dendrimer on the SPION surfaces. In vitro biocompatibility was assessed using the 3-[4,5-dimethylthiazol-2yl]-2,5-diphenyl tetrazolium bromide assay. No significant toxicity was noted on BCECs within 24 h of incubation. In the cell migration assay, cells treated with dendrimer-coated SPIONs exhibited a relatively high wound healing rate under sample addition (1 μg/mL) under a magnetic field. Real-time PCR on BCECs treated with dendrimer-coated SPIONs revealed upregulation of specific genes, including AT1P1 and NCAM1, for BCECs-dendrimer-coated SPIONs under a magnetic field. The three-dimensional dispersion of BCECs containing dendrimer-coated SPIONs under a magnetic field was evaluated using COMSOL Multiphysics software. The results revealed the BCECs-SPION vortex pattern layers in the corneal surface corresponded to the electromagnet's displacement from the ocular surface. Magnetic resonance imaging (MRI) indicated that dendrimer-coated SPIONs can be used as a T2 contrast agent.
Collapse
Affiliation(s)
- Shadie Hatamie
- College of Medicine, National Taiwan University, Taipei 10048, Taiwan;
- Department of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan;
| | - Po-Jen Shih
- Department of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan;
| | - Bo-Wei Chen
- Institute of Nanoengineering and Microsystem, National Tsing Hua University, Hsinchu 30013, Taiwan; (B.-W.C.); (D.-J.Y.)
| | - Hua-Ju Shih
- Institute of Applied Mechanics, National Taiwan University, Taipei 10617, Taiwan;
| | - I-Jong Wang
- College of Medicine, National Taiwan University, Taipei 10048, Taiwan;
| | - Tai-Horng Young
- Department of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan;
| | - Da-Jeng Yao
- Institute of Nanoengineering and Microsystem, National Tsing Hua University, Hsinchu 30013, Taiwan; (B.-W.C.); (D.-J.Y.)
| |
Collapse
|
13
|
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: 25] [Impact Index Per Article: 8.3] [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
|
14
|
Amador C, Shah R, Ghiam S, Kramerov AA, Ljubimov AV. Gene therapy in the anterior eye segment. Curr Gene Ther 2021; 22:104-131. [PMID: 33902406 DOI: 10.2174/1566523221666210423084233] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/14/2021] [Accepted: 04/04/2021] [Indexed: 11/22/2022]
Abstract
This review provides comprehensive information about the advances in gene therapy in the anterior segment of the eye including cornea, conjunctiva, lacrimal gland, and trabecular meshwork. We discuss gene delivery systems including viral and non-viral vectors as well as gene editing techniques, mainly CRISPR-Cas9, and epigenetic treatments including antisense and siRNA therapeutics. We also provide a detailed analysis of various anterior segment diseases where gene therapy has been tested with corresponding outcomes. Disease conditions include corneal and conjunctival fibrosis and scarring, corneal epithelial wound healing, corneal graft survival, corneal neovascularization, genetic corneal dystrophies, herpetic keratitis, glaucoma, dry eye disease, and other ocular surface diseases. Although most of the analyzed results on the use and validity of gene therapy at the ocular surface have been obtained in vitro or using animal models, we also discuss the available human studies. Gene therapy approaches are currently considered very promising as emerging future treatments of various diseases, and this field is rapidly expanding.
Collapse
Affiliation(s)
- Cynthia Amador
- Eye Program, Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ruchi Shah
- Eye Program, Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Sean Ghiam
- Sackler School of Medicine, New York State/American Program of Tel Aviv University, Tel Aviv, Israel
| | - Andrei A Kramerov
- Eye Program, Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Alexander V Ljubimov
- Eye Program, Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
15
|
Chen F, Bian M, Nahmou M, Myung D, Goldberg JL. Fusogenic liposome-enhanced cytosolic delivery of magnetic nanoparticles. RSC Adv 2021; 11:35796-35805. [PMID: 35492766 PMCID: PMC9043121 DOI: 10.1039/d1ra03094a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022] Open
Abstract
Fusogenic liposomes facilitate MNPs passage into the cytosol and enable direct contact between MNPs and organelles other than endosomes.
Collapse
Affiliation(s)
- Fang Chen
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute at Stanford University, Palo Alto, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Minjuan Bian
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute at Stanford University, Palo Alto, CA, 94304, USA
| | - Michael Nahmou
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute at Stanford University, Palo Alto, CA, 94304, USA
| | - David Myung
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute at Stanford University, Palo Alto, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Jeffrey L. Goldberg
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute at Stanford University, Palo Alto, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| |
Collapse
|
16
|
Khalili M, Asadi M, Kahroba H, Soleyman MR, Andre H, Alizadeh E. Corneal endothelium tissue engineering: An evolution of signaling molecules, cells, and scaffolds toward 3D bioprinting and cell sheets. J Cell Physiol 2020; 236:3275-3303. [PMID: 33090510 DOI: 10.1002/jcp.30085] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/31/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
Cornea is an avascular and transparent tissue that focuses light on retina. Cornea is supported by the corneal-endothelial layer through regulation of hydration homeostasis. Restoring vision in patients afflicted with corneal endothelium dysfunction-mediated blindness most often requires corneal transplantation (CT), which faces considerable constrictions due to donor limitations. An emerging alternative to CT is corneal endothelium tissue engineering (CETE), which involves utilizing scaffold-based methods and scaffold-free strategies. The innovative scaffold-free method is cell sheet engineering, which typically generates cell layers surrounded by an intact extracellular matrix, exhibiting tunable release from the stimuli-responsive surface. In some studies, scaffold-based or scaffold-free technologies have been reported to achieve promising outcomes. However, yet some issues exist in translating CETE from bench to clinical practice. In this review, we compare different corneal endothelium regeneration methods and elaborate on the application of multiple cell types (stem cells, corneal endothelial cells, and endothelial precursors), signaling molecules (growth factors, cytokines, chemical compounds, and small RNAs), and natural and synthetic scaffolds for CETE. Furthermore, we discuss the importance of three-dimensional bioprinting strategies and simulation of Descemet's membrane by biomimetic topography. Finally, we dissected the recent advances, applications, and prospects of cell sheet engineering for CETE.
Collapse
Affiliation(s)
- Mostafa Khalili
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Asadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Houman Kahroba
- Biomedicine Institute, and Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Soleyman
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Helder Andre
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Effat Alizadeh
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Okumura N, Koizumi N. Review and perspective of tissue engineering therapy for the treatment of corneal endothelial decompensation. EXPERT REVIEW OF OPHTHALMOLOGY 2020. [DOI: 10.1080/17469899.2020.1811088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
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
|
18
|
Price MO, Mehta JS, Jurkunas UV, Price FW. Corneal endothelial dysfunction: Evolving understanding and treatment options. Prog Retin Eye Res 2020; 82:100904. [PMID: 32977001 DOI: 10.1016/j.preteyeres.2020.100904] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022]
Abstract
The cornea is exquisitely designed to protect the eye while transmitting and focusing incoming light. Precise control of corneal hydration by the endothelial cell layer that lines the inner surface of the cornea is required for optimal transparency, and endothelial dysfunction or damage can result in corneal edema and visual impairment. Advances in corneal transplantation now allow selective replacement of dysfunctional corneal endothelium, providing rapid visual rehabilitation. A series of technique improvements have minimized complications and various adaptations allow use even in eyes with complicated anatomy. While selective endothelial keratoplasty sets a very high standard for safety and efficacy, a shortage of donor corneas in many parts of the world restricts access, prompting a search for alternatives. Clinical trials are underway to evaluate the potential for self-recovery after removal of dysfunctional central endothelium in patients with healthy peripheral endothelium. Various approaches to using cultured human corneal endothelial cells are also in clinical trials; these aim to multiply cells from a single donor cornea for use in potentially hundreds of patients. Pre-clinical studies are underway with induced pluripotent stem cells, endothelial stem cell regeneration, gene therapy, anti-sense oligonucleotides, and various biologic/pharmacologic approaches designed to treat, prevent, or retard corneal endothelial dysfunction. The availability of more therapeutic options will hopefully expand access around the world while also allowing treatment to be more precisely tailored to each individual patient.
Collapse
Affiliation(s)
- Marianne O Price
- Cornea Research Foundation of America, 9002 N. Meridian St., Suite 212, Indianapolis, IN, USA.
| | - Jodhbir S Mehta
- Singapore National Eye Centre, 11 Third Hospital Ave #08-00, 168751, Singapore
| | - Ula V Jurkunas
- Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA, USA
| | - Francis W Price
- Price Vision Group, 9002 N. Meridian St., Suite 100, Indianapolis, IN, USA
| |
Collapse
|
19
|
Kono Y, Gogatsubo S, Ohba T, Fujita T. Enhanced macrophage delivery to the colon using magnetic lipoplexes with a magnetic field. Drug Deliv 2020; 26:935-943. [PMID: 31530198 PMCID: PMC6758636 DOI: 10.1080/10717544.2019.1662515] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Magnetically guided cell delivery systems would be valuable to achieve effective macrophage-based cell therapy for colonic inflammatory diseases. In the current study, we developed a method for the efficient and simultaneous introduction of superparamagnetic iron oxide nanoparticles (SPIONs) and plasmid DNA (pDNA) into RAW264 murine macrophage-like cells using SPION-incorporated cationic liposome/pDNA complexes (magnetic lipoplexes). SPIONs and pDNA were introduced for magnetization and functionalization of the macrophages, respectively. We also evaluated the adhesive properties of magnetized RAW264 cells using magnetic lipoplexes in the murine colon under a magnetic field. Significant cellular association and gene expression without cytotoxicity were observed when magnetic cationic liposomes and pDNA were mixed at a weight ratio of 10:1, and SPION concentration and magnetic field exposure time was 0.1 mg/mL and 10 min, respectively. We also observed that cytokine production in magnetized RAW264 cells was similar to that in non-treated RAW264 cells, whereas nitric oxide production was significantly increased in magnetized RAW264 cells. Furthermore, magnetized RAW264 cells highly adhered to a Caco-2 cell monolayer and colon in mice, under a magnetic field. These results suggest that this magnetic cell delivery system can improve the colonic delivery of macrophages and its therapeutic efficacy against colonic inflammatory diseases.
Collapse
Affiliation(s)
- Yusuke Kono
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University , Kusatsu , Japan.,Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan
| | - Serika Gogatsubo
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan
| | - Takeshi Ohba
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan
| | - Takuya Fujita
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University , Kusatsu , Japan.,Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University , Kusatsu , Japan.,Research Center for Drug Discovery and Development, Ritsumeikan University , Kusatsu , Japan
| |
Collapse
|
20
|
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
|
21
|
Xia X, Atkins M, Dalal R, Kuzmenko O, Chang KC, Sun CB, Benatti CA, Rak DJ, Nahmou M, Kunzevitzky NJ, Goldberg JL. Magnetic Human Corneal Endothelial Cell Transplant: Delivery, Retention, and Short-Term Efficacy. Invest Ophthalmol Vis Sci 2019; 60:2438-2448. [PMID: 31158276 PMCID: PMC6546151 DOI: 10.1167/iovs.18-26001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose Corneal endothelial dysfunction leads to corneal edema, pain, and vision loss. Adequate animal models are needed to study the safety and efficacy of novel cell therapies as an alternative to corneal transplantation. Methods Primary human corneal endothelial cells (HCECs) were isolated from cadaveric donor corneas, expanded in vitro, transduced to express green fluorescent protein (GFP), loaded with superparamagnetic nanoparticles, and injected into the anterior chamber of adult rabbits immediately after endothelial cell or Descemet's membrane stripping. The same volume of balanced salt solution plus (BSS+) was injected in control eyes. We compared different models for inducing corneal edema in rabbits, and examined the ability of transplanted HCECs to reduce corneal edema over time by measuring central corneal thickness and tracking corneal clarity. GFP-positive donor cells were tracked in vivo using optical coherence tomography (OCT) fluorescence angiography module, and the transplanted cells were confirmed by human nuclei immunostaining. Results Magnetic HCECs integrated onto the recipient corneas with intact Descemet's membrane, and donor identity was confirmed by GFP expression and immunostaining for human nuclei marker. Donor HCECs formed a monolayer on the posterior corneal surface and expressed HCEC functional markers of tight junction formation. No GFP-positive cells were observed in the trabecular meshwork or on the iris, and intraocular pressure remained stable through the length of the study. Conclusions Our results demonstrate magnetic cell-based therapy efficiently delivers HCECs to restore corneal transparency without detectable toxicity or adverse effect on intraocular pressure. Magnetic delivery of HCECs may enhance corneal function and should be explored further for human therapies.
Collapse
Affiliation(s)
- Xin Xia
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - Melissa Atkins
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - Roopa Dalal
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - Olga Kuzmenko
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - Kun-Che Chang
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - Catalina B Sun
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - C Andres Benatti
- Shiley Eye Center, University of California, San Diego, La Jolla, California, United States
| | - Dillon J Rak
- Shiley Eye Center, University of California, San Diego, La Jolla, California, United States
| | - Michael Nahmou
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - Noelia J Kunzevitzky
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States.,Shiley Eye Center, University of California, San Diego, La Jolla, California, United States.,Emmecell, Menlo Park, California, United States
| | - Jeffrey L Goldberg
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States.,Shiley Eye Center, University of California, San Diego, La Jolla, California, United States
| |
Collapse
|
22
|
Chen F, Zhao ER, Hableel G, Hu T, Kim T, Li J, Gonzalez-Pech NI, Cheng DJ, Lemaster JE, Xie Y, Grassian VH, Sen GL, Jokerst JV. Increasing the Efficacy of Stem Cell Therapy via Triple-Function Inorganic Nanoparticles. ACS NANO 2019; 13:6605-6617. [PMID: 31188564 PMCID: PMC8106804 DOI: 10.1021/acsnano.9b00653] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Stem cell therapy in heart disease is challenged by mis-injection, poor survival, and low cell retention. Here, we describe a biocompatible multifunctional silica-iron oxide nanoparticle to help solve these issues. The nanoparticles were made via an in situ growth of Fe3O4 nanoparticles on both the external surfaces and pore walls of mesocellular foam silica nanoparticles. In contrast to previous work, this approach builds a magnetic moiety inside the pores of a porous silica structure. These materials serve three roles: drug delivery, magnetic manipulation, and imaging. The addition of Fe3O4 to the silica nanoparticles increased their colloidal stability, T2-based magnetic resonance imaging contrast, and superparamagnetism. We then used the hybrid materials as a sustained release vehicle of insulin-like growth factor-a pro-survival agent that can increase cell viability. In vivo rodent studies show that labeling stem cells with this nanoparticle increased the efficacy of stem cell therapy in a ligation/reperfusion model. The nanoparticle-labeled cells increase the mean left ventricular ejection fraction by 11 and 21% and the global longitudinal strain by 24 and 34% on days 30 and 60, respectively. In summary, this multifunctional nanomedicine improves stem cell survival via the sustained release of pro-survival agents.
Collapse
Affiliation(s)
- Fang Chen
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Eric Ruike Zhao
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Ghanim Hableel
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Tao Hu
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
| | - Taeho Kim
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Biomedical Engineering, Institute of Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
| | - Jingting Li
- Department of Dermatology and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Natalia Isabel Gonzalez-Pech
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - David J. Cheng
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jeanne E. Lemaster
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yijun Xie
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Vicki H. Grassian
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Scripps Institution of Oceanography, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - George L. Sen
- Department of Dermatology and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jesse V. Jokerst
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Radiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Corresponding Author. Tel.: +1 (858) 246-0896
| |
Collapse
|
23
|
Bartakova A, Kuzmenko O, Alvarez-Delfin K, Kunzevitzky NJ, Goldberg JL. A Cell Culture Approach to Optimized Human Corneal Endothelial Cell Function. Invest Ophthalmol Vis Sci 2018; 59:1617-1629. [PMID: 29625488 PMCID: PMC5869002 DOI: 10.1167/iovs.17-23637] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Cell-based therapies to replace corneal endothelium depend on culture methods to optimize human corneal endothelial cell (HCEC) function and minimize endothelial-mesenchymal transition (EnMT). Here we explore contribution of low-mitogenic media on stabilization of phenotypes in vitro that mimic those of HCECs in vivo. Methods HCECs were isolated from cadaveric donor corneas and expanded in vitro, comparing continuous presence of exogenous growth factors (“proliferative media”) to media without those factors (“stabilizing media”). Identity based on canonical morphology and expression of surface marker CD56, and function based on formation of tight junction barriers measured by trans-endothelial electrical resistance assays (TEER) were assessed. Results Primary HCECs cultured in proliferative media underwent EnMT after three to four passages, becoming increasingly fibroblastic. Stabilizing the cells before each passage by switching them to a media low in mitogenic growth factors and serum preserved canonical morphology and yielded a higher number of cells. HCECs cultured in stabilizing media increased both expression of the identity marker CD56 and also tight junction monolayer integrity compared to cells cultured without stabilization. Conclusions HCECs isolated from donor corneas and expanded in vitro with a low-mitogenic media stabilizing step before each passage demonstrate more canonical structural and functional features and defer EnMT, increasing the number of passages and total canonical cell yield. This approach may facilitate development of HCEC-based cell therapies.
Collapse
Affiliation(s)
- Alena Bartakova
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Olga Kuzmenko
- Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States
| | - Karen Alvarez-Delfin
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Florida, United States
| | - Noelia J Kunzevitzky
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States.,Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States.,Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Florida, United States.,Emmecell, Bridgewater, Connecticut, United States
| | - Jeffrey L Goldberg
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States.,Byers Eye Institute and Spencer Center for Vision Research, Department of Ophthalmology, Stanford University, Palo Alto, California, United States.,Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Florida, United States
| |
Collapse
|
24
|
Jia Y, Li W, Duan H, Li Z, Zhou Q, Shi W. Mini-Sheet Injection for Cultured Corneal Endothelial Transplantation. Tissue Eng Part C Methods 2018; 24:474-479. [DOI: 10.1089/ten.tec.2018.0077] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Yanni Jia
- Department of Medicine, Qingdao University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Wenjing Li
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Haoyun Duan
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Zongyi Li
- Department of Medicine, Qingdao University, Qingdao, China
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Qingjun Zhou
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Weiyun Shi
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| |
Collapse
|
25
|
[Novel concepts for treatment of the corneal endothelium with nanoparticles]. Ophthalmologe 2018; 115:190-194. [PMID: 29294166 DOI: 10.1007/s00347-017-0640-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND The number of corneal transplantations has increased in Germany in recent years. One reason is an earlier time point of surgery due to less invasive posterior lamellar grafting techniques. To date, penetrating and lamellar corneal transplantations are the only established therapeutic options to treat corneal diseases resulting from endothelial cell pathologies. AIM This review article provides an insight into nanoparticle-related translational strategies to improve or to avoid corneal transplantation. RESULTS Nanoparticle-based strategies for optimization of the corneal endothelium have different fields of application: 1. during donor cornea cultivation in culture medium, 2. for single cell injection therapies and 3. to treat the patients' own endothelium in an effort to avoid transplantation. CONCLUSION Several translational concepts exist to improve or to avoid grafting of a donor cornea. The coming decade will provide established alternatives to conventional corneal transplantation.
Collapse
|
26
|
Sun P, Shen L, Zhang C, Du L, Wu X. Promoting the expansion and function of human corneal endothelial cells with an orbital adipose-derived stem cell-conditioned medium. Stem Cell Res Ther 2017; 8:287. [PMID: 29262856 PMCID: PMC5738836 DOI: 10.1186/s13287-017-0737-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/16/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Corneal endothelial dysfunction causes severe impairment of vision. The only solution is corneal transplantation. However, this treatment is hampered by a worldwide shortage of donor corneas. New therapies may replace the conventional donor corneal transplantation alongside the developments in regenerative medicine and tissue engineering, but sufficient functional corneal endothelial cells (CECs) are essential. The aim of this study was to promote the expansion and function of human corneal endothelial cells (HCECs) in vitro and in vivo. METHODS The phenotypes of human orbital adipose-derived stem cells (OASCs) were detected by flow cytometry and immunofluorescence. HCECs were isolated and cultured using a conditioned medium obtained from OASCs (OASC-CM) in vitro. Related cell markers of HCECs were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR), Western blot, and immunofluorescence. The cell counting kit-8 (CCK-8) assay and the wound healing assay were performed to evaluate the proliferation ability of the cells. The cultured HCECs were then transplanted into rabbit and monkey corneal endothelial dysfunction models by cell injection. RESULTS CD29, CD105, CD49e, CD166, and vimentin were highly expressed in cultured human OASCs. The CEC-relative markers zonula occludens-1 (ZO-1), Na+/K+ ATPase, N-cadherin, Col8a2, and SLC4A4 were expressed in HCECs cultured by OASC-CM. The HCECs were able to maintain polygonal cell morphology and good proliferative capacity. In animal experiments, corneal transparency was achieved after the injection of HCECs, which demonstrated the good repair capacity of the cells. CONCLUSIONS The proliferation abilities of the cells were significantly enhanced, and related functional markers were strongly positive, while HCEC morphology was maintained using OASC-CM. HCECs obtained some stem cell-like properties. This preclinical study confirmed the therapeutic ability of the HCECs in vivo. Our findings demonstrated that cultured HCECs with OASC-CM might be a promising source for research and clinical treatment.
Collapse
Affiliation(s)
- Peng Sun
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Lin Shen
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Canwei Zhang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Liqun Du
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xinyi Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
27
|
Umbilical cord stem cells in the treatment of corneal disease. Surv Ophthalmol 2017; 62:803-815. [DOI: 10.1016/j.survophthal.2017.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
|
28
|
Bartakova A, Alvarez-Delfin K, Weisman AD, Salero E, Raffa GA, Merkhofer RM, Kunzevitzky NJ, Goldberg JL. Novel Identity and Functional Markers for Human Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2017; 57:2749-62. [PMID: 27196322 PMCID: PMC4884060 DOI: 10.1167/iovs.15-18826] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Human corneal endothelial cell (HCEC) density decreases with age, surgical complications, or disease, leading to vision impairment. Such endothelial dysfunction is an indication for corneal transplantation, although there is a worldwide shortage of transplant-grade tissue. To overcome the current poor donor availability, here we isolate, expand, and characterize HCECs in vitro as a step toward cell therapy. Methods Human corneal endothelial cells were isolated from cadaveric corneas and expanded in vitro. Cell identity was evaluated based on morphology and immunocytochemistry, and gene expression analysis and flow cytometry were used to identify novel HCEC-specific markers. The functional ability of HCEC to form barriers was assessed by transendothelial electrical resistance (TEER) assays. Results Cultured HCECs demonstrated canonical morphology for up to four passages and later underwent endothelial-to-mesenchymal transition (EnMT). Quality of donor tissue influenced cell measures in culture including proliferation rate. Cultured HCECs expressed identity markers, and microarray analysis revealed novel endothelial-specific markers that were validated by flow cytometry. Finally, canonical HCECs expressed higher levels of CD56, which correlated with higher TEER than fibroblastic HCECs. Conclusions In vitro expansion of HCECs from cadaveric donor corneas yields functional cells identifiable by morphology and a panel of novel markers. Markers described correlated with function in culture, suggesting a basis for cell therapy for corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Alena Bartakova
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Karen Alvarez-Delfin
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Alejandra D Weisman
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Enrique Salero
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Gabriella A Raffa
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Richard M Merkhofer
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Noelia J Kunzevitzky
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States 2Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States 3Emmecell, K
| | - Jeffrey L Goldberg
- Shiley Eye Institute, University of California San Diego, La Jolla, California, United States 2Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States 4Byers Eye I
| |
Collapse
|
29
|
|
30
|
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: 4.3] [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
|
31
|
Czugala M, Mykhaylyk O, Böhler P, Onderka J, Stork B, Wesselborg S, Kruse FE, Plank C, Singer BB, Fuchsluger TA. Efficient and safe gene delivery to human corneal endothelium using magnetic nanoparticles. Nanomedicine (Lond) 2016; 11:1787-800. [DOI: 10.2217/nnm-2016-0144] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: To develop a safe and efficient method for targeted, anti-apoptotic gene therapy of corneal endothelial cells (CECs). Materials & methods: Magnetofection (MF), a combination of lipofection with magnetic nanoparticles (MNPs; PEI-Mag2, SO-Mag5, PalD1-Mag1), was tested in human CECs and in explanted human corneas. Effects on cell viability and function were investigated. Immunocompatibility was assessed in human peripheral blood mononuclear cells. Results: Silica iron-oxide MNPs (SO-Mag5) combined with X-tremeGENE-HP achieved high transfection efficiency in human CECs and explanted human corneas, without altering cell viability or function. Magnetofection caused no immunomodulatory effects in human peripheral blood mononuclear cells. Magnetofection with anti-apoptotic P35 gene effectively blocked apoptosis in CECs. Conclusion: Magnetofection is a promising tool for gene therapy of corneal endothelial cells with potential for targeted on-site delivery.
Collapse
Affiliation(s)
- Marta Czugala
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Institute of Anatomy, University Duisburg-Essen, Essen, Germany
| | - Olga Mykhaylyk
- Institute of Immunology & Experimental Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Philip Böhler
- Institute of Molecular Medicine I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jasmine Onderka
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Björn Stork
- Institute of Molecular Medicine I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Wesselborg
- Institute of Molecular Medicine I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Friedrich E Kruse
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Plank
- Institute of Immunology & Experimental Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Thomas A Fuchsluger
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
32
|
Soh YQ, Peh GSL, Mehta JS. Translational issues for human corneal endothelial tissue engineering. J Tissue Eng Regen Med 2016; 11:2425-2442. [DOI: 10.1002/term.2131] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/19/2015] [Accepted: 12/10/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Yu Qiang Soh
- Tissue Engineering and Stem Cell Group; Singapore Eye Research Institute; Singapore
- Singapore National Eye Centre; Singapore
| | - Gary S. L. Peh
- Tissue Engineering and Stem Cell Group; Singapore Eye Research Institute; Singapore
- Ophthalmology Academic Clinical Programme; Duke-NUS Graduate Medical School; Singapore
| | - Jodhbir S. Mehta
- Tissue Engineering and Stem Cell Group; Singapore Eye Research Institute; Singapore
- Singapore National Eye Centre; Singapore
- Ophthalmology Academic Clinical Programme; Duke-NUS Graduate Medical School; Singapore
- Department of Clinical Sciences; Duke-NUS Graduate Medical School; Singapore
| |
Collapse
|
33
|
Mehra NK, Cai D, Kuo L, Hein T, Palakurthi S. Safety and toxicity of nanomaterials for ocular drug delivery applications. Nanotoxicology 2016; 10:836-60. [PMID: 27027670 DOI: 10.3109/17435390.2016.1153165] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multifunctional nanomaterials are rapidly emerging for ophthalmic delivery of therapeutics to facilitate safe and effective targeting with improved patient compliance. Because of their extremely high area to volume ratio, nanomaterials often have physicochemical properties that are different from those of their larger counterparts. There exists a complex relationship between the physicochemical properties (composition, size, shape, charge, roughness, and porosity) of the nanomaterials and their interaction with the biological system. The eye is a very sensitive accessible organ and is subjected to intended and unintended exposure to nanomaterials. Currently, various ophthalmic formulations are available in the market, while some are underway in preclinical and clinical phases. However, the data on safety, efficacy, and toxicology of these advanced nanomaterials for ocular drug delivery are sparse. Focus of the present review is to provide a comprehensive report on the safety, biocompatibility and toxicities of nanomaterials in the eye.
Collapse
Affiliation(s)
- Neelesh K Mehra
- a Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , Kingsville , TX , USA
| | - Defu Cai
- a Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , Kingsville , TX , USA
| | - Lih Kuo
- b Department of Medical Physiology, College of Medicine , Texas A&M Health Science Center , Temple , TX , USA ;,c Department of Surgery and Scott & White Eye Institute, College of Medicine , Texas A&M Health Science Center , Temple , TX , USA
| | - Travis Hein
- c Department of Surgery and Scott & White Eye Institute, College of Medicine , Texas A&M Health Science Center , Temple , TX , USA
| | - Srinath Palakurthi
- a Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , Kingsville , TX , USA
| |
Collapse
|
34
|
McCabe KL, Kunzevitzky NJ, Chiswell BP, Xia X, Goldberg JL, Lanza R. Efficient Generation of Human Embryonic Stem Cell-Derived Corneal Endothelial Cells by Directed Differentiation. PLoS One 2015; 10:e0145266. [PMID: 26689688 PMCID: PMC4686926 DOI: 10.1371/journal.pone.0145266] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/30/2015] [Indexed: 12/13/2022] Open
Abstract
Aim To generate human embryonic stem cell derived corneal endothelial cells (hESC-CECs) for transplantation in patients with corneal endothelial dystrophies. Materials and Methods Feeder-free hESC-CECs were generated by a directed differentiation protocol. hESC-CECs were characterized by morphology, expression of corneal endothelial markers, and microarray analysis of gene expression. Results hESC-CECs were nearly identical morphologically to primary human corneal endothelial cells, expressed Zona Occludens 1 (ZO-1) and Na+/K+ATPaseα1 (ATPA1) on the apical surface in monolayer culture, and produced the key proteins of Descemet’s membrane, Collagen VIIIα1 and VIIIα2 (COL8A1 and 8A2). Quantitative PCR analysis revealed expression of all corneal endothelial pump transcripts. hESC-CECs were 96% similar to primary human adult CECs by microarray analysis. Conclusion hESC-CECs are morphologically similar, express corneal endothelial cell markers and express a nearly identical complement of genes compared to human adult corneal endothelial cells. hESC-CECs may be a suitable alternative to donor-derived corneal endothelium.
Collapse
Affiliation(s)
- Kathryn L. McCabe
- Ocata Therapeutics, Marlborough, MA, 01752, United States of America
| | - Noelia J. Kunzevitzky
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, 33136, United States of America
- Emmecell, Key Biscayne, FL, 33149, United States of America
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, United States of America
| | - Brian P. Chiswell
- Ocata Therapeutics, Marlborough, MA, 01752, United States of America
| | - Xin Xia
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, United States of America
| | - Jeffrey L. Goldberg
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, 33136, United States of America
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, United States of America
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, 94303, United States of America
| | - Robert Lanza
- Ocata Therapeutics, Marlborough, MA, 01752, United States of America
- * E-mail:
| |
Collapse
|
35
|
Navaratnam J, Utheim TP, Rajasekhar VK, Shahdadfar A. Substrates for Expansion of Corneal Endothelial Cells towards Bioengineering of Human Corneal Endothelium. J Funct Biomater 2015; 6:917-45. [PMID: 26378588 PMCID: PMC4598685 DOI: 10.3390/jfb6030917] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/31/2015] [Accepted: 09/02/2015] [Indexed: 12/13/2022] Open
Abstract
Corneal endothelium is a single layer of specialized cells that lines the posterior surface of cornea and maintains corneal hydration and corneal transparency essential for vision. Currently, transplantation is the only therapeutic option for diseases affecting the corneal endothelium. Transplantation of corneal endothelium, called endothelial keratoplasty, is widely used for corneal endothelial diseases. However, corneal transplantation is limited by global donor shortage. Therefore, there is a need to overcome the deficiency of sufficient donor corneal tissue. New approaches are being explored to engineer corneal tissues such that sufficient amount of corneal endothelium becomes available to offset the present shortage of functional cornea. Although human corneal endothelial cells have limited proliferative capacity in vivo, several laboratories have been successful in in vitro expansion of human corneal endothelial cells. Here we provide a comprehensive analysis of different substrates employed for in vitro cultivation of human corneal endothelial cells. Advances and emerging challenges with ex vivo cultured corneal endothelial layer for the ultimate goal of therapeutic replacement of dysfunctional corneal endothelium in humans with functional corneal endothelium are also presented.
Collapse
Affiliation(s)
- Jesintha Navaratnam
- Department of Ophthalmology, Oslo University Hospital, Postbox 4950 Nydalen, Oslo 0424, Norway.
| | - Tor P Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Postbox 4950 Nydalen, Oslo 0424, Norway.
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Postbox 1052, Blindern, Oslo 0316, Norway.
| | - Vinagolu K Rajasekhar
- Memorial Sloan Kettering Cancer Center, Rockefeller Research Building, Room 1163, 430 East 67th Street/1275 York Avenue, New York, NY 10065, USA.
| | - Aboulghassem Shahdadfar
- Department of Ophthalmology, Oslo University Hospital, Postbox 4950 Nydalen, Oslo 0424, Norway.
| |
Collapse
|