1
|
Jo S, Kim YJ, Hwang T, Jang SY, Park SJ, Ye S, Jung Y, Yoo J. Injectable ultrathin porous membranes harnessing shape memory polymers for retinal tissue engineering. J Mater Chem B 2025; 13:3161-3172. [PMID: 39905826 DOI: 10.1039/d4tb02287d] [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: 02/06/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss, characterized by the progressive degeneration of retinal cells, particularly retinal pigment epithelial (RPE) cells. Conventional treatments primarily focus on slowing disease progression without providing a cure. Recent advances in tissue engineering and cell-based therapies offer promising avenues for regenerating retinal tissue and restoring vision. In this study, we developed ultrathin, nanoporous membrane scaffolds designed to mimic Bruch's membrane (BrM) for RPE cell transplantation using vapor-induced phase separation. These scaffolds, fabricated from a blend of poly(L-lactide-co-ε-caprolactone) (PLCL) and poly(lactic-co-glycolic acid) (PLGA), exhibited favorable topography, biocompatibility, and shape-memory properties. In vitro experiments confirmed that the nanoporous topography effectively supports the formation of RPE monolayers with intact tight junctions. Additionally, the shape-memory characteristic enables the membrane to self-expand at body temperature (37 °C), facilitating minimally invasive delivery via injection. ARPE-19 cell-attached nanothin membranes successfully demonstrated shape-recovery properties and were deliverable through a catheter in an ex vivo model. Our findings suggest that the developed scaffolds provide a promising approach for retinal tissue engineering and could significantly contribute to advanced treatments for AMD and other retinal degenerative diseases.
Collapse
Affiliation(s)
- SeongHoon Jo
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Yu-Jin Kim
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Taek Hwang
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
- Department of Metabiohealth, Sungkyun Convergence Institute, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Se Youn Jang
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - So-Jin Park
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
- School of Chemical Engineering & Materials Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Seongryeol Ye
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
- School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jin Yoo
- Biomaterials Research Center, Biomedical Research Division, Korea Instituten of Sicence and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| |
Collapse
|
2
|
Abbasi N, O'Neill H. Cytocompatibility of electrospun poly-L-lactic acid membranes for Bruch's membrane regeneration using human embryonic stem cell-derived retinal pigment epithelial cells. J Biomed Mater Res A 2024; 112:1902-1920. [PMID: 38726752 DOI: 10.1002/jbm.a.37736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/11/2024] [Accepted: 04/30/2024] [Indexed: 09/03/2024]
Abstract
Cell replacement therapy is under development for dry age-related macular degeneration (AMD). A thin membrane resembling the Bruch's membrane is required to form a cell-on-membrane construct with retinal pigment epithelial (RPE) cells. These cells have been differentiated from human embryonic stem cells (hESCs) in vitro. A carrier membrane is required for cell implantation, which is biocompatible for cell growth and has dimensions and physical properties resembling the Bruch's membrane. Here a nanofiber electrospun poly-L-lactic acid (PLLA) membrane is tested for capacity to support cell growth and maturation. The requirements for laminin coating of the membrane are identified here. A porous electrospun nanofibrous PLLA membrane of ∼50 nm fiber diameter was developed as a prototype support for functional RPE cells grown as a monolayer. The need for laminin coating applied to the membrane following treatment with poly-L-ornithine (PLO), was identified in terms of cell growth and survival. Test membranes were compared in terms of hydrophilicity after laminin coating, mechanical properties of surface roughness and Young's modulus, porosity and ability to promote the attachment and proliferation of hESC-RPE cells in culture for up to 8 weeks. Over this time, RPE cell proliferation, morphology, and marker and gene expression, were monitored. The functional capacity of cell monolayers was identified in terms of transepithelial electrical resistance (TEER), phagocytosis of cells, as well as expression of the cytokines, vascular endothelial growth factor (VEGF) and pigment epithelium-derived factor (PEDF). PLLA polymer fibers are naturally hydrophobic, so their hydrophilicity was improved by pretreatment with PLO for subsequent coating with the bioactive protein laminin. They were then assessed for amount of laminin adsorbed, contact angle and uniformity of coating using scanning electron microscopy (SEM). Pretreatment with 100% PLO gave the best result over 10% PLO treatment or no treatment prior to laminin adsorption with significantly greater surface stiffness and modulus. By 6 weeks after cell plating, the coated membranes could support a mature RPE monolayer showing a dense apical microvillus structure and pigmented 3D polygonal cell morphology. After 8 weeks, PLO (100%)-Lam coated membranes exhibited the highest cell number, cell proliferation, and RPE barrier function measured as TEER. RPE cells showed the higher levels of specific surface marker and gene expression. Microphthalmia-associated transcription factor expression was highly upregulated indicating maturation of cells. Functionality of cells was indicated by expression of VEGF and PEDF genes as well as phagocytic capacity. In conclusion, electrospun PLLA membranes coated with PLO-Lam have the physical and biological properties to support the distribution and migration of hESC-RPE cells throughout the whole structure. They represent a good membrane candidate for preparation of hESC-RPE cells as a monolayer for implantation into the subretinal space of AMD patients.
Collapse
Affiliation(s)
- Naghmeh Abbasi
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Helen O'Neill
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Queensland, Australia
| |
Collapse
|
3
|
Trousil J, Cabral JV, Voukali E, Nováčková J, Pop-Georgievski O, Vacík T, Studený P, Studenovska H, Jirsova K. Electrospun poly(l-lactide- co-dl-lactide) nanofibrous scaffold as substrate for ex vivo limbal epithelial cell cultivation. Heliyon 2024; 10:e30970. [PMID: 38803982 PMCID: PMC11128869 DOI: 10.1016/j.heliyon.2024.e30970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/13/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
Ultrathin electrospun poly (l-lactide-co-dl-lactide) nanofibrous membranes coated with fibronectin were explored as scaffolds for the ex vivo cultivation of limbal epithelial cells (LECs) for the treatment of limbal stem cell deficiency. The developed scaffolds were compared with the "gold-standard" fibrin gel. The resulting membranes composed of nanofibers possessed a very low thickness of 4 μm and allowed very good optical transparency in the wet state. The fibronectin-coated nanofibrous scaffolds demonstrated LEC expansion and successful cultivation similar to that on fibrin gel. Unlike the regular cobblestone epithelial cell morphology on the fibrin gel, the nanofibrous scaffold presented a mostly irregular epithelial morphology with a shift to a mesenchymal phenotype, as confirmed by the upregulation of profibroblastic genes: ACTA2 (p = 0.023), FBLN1 (p < 0.001), and THY1 (p < 0.001). Both culture conditions revealed comparable expression of stem cell markers, including KLF4, ΔNp63α and ABCG2, emphasizing the promise of polylactide-based nanofibrous membranes for further investigations.
Collapse
Affiliation(s)
- Jiří Trousil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Joao Victor Cabral
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Eleni Voukali
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jitka Nováčková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Vacík
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Studený
- Ophthalmology Department, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Hana Studenovska
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Katerina Jirsova
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
4
|
Dujardin C, Habeler W, Monville C, Letourneur D, Simon-Yarza T. Advances in the engineering of the outer blood-retina barrier: From in-vitro modelling to cellular therapy. Bioact Mater 2024; 31:151-177. [PMID: 37637086 PMCID: PMC10448242 DOI: 10.1016/j.bioactmat.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/13/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023] Open
Abstract
The outer blood-retina barrier (oBRB), crucial for the survival and the proper functioning of the overlying retinal layers, is disrupted in numerous diseases affecting the retina, leading to the loss of the photoreceptors and ultimately of vision. To study the oBRB and/or its degeneration, many in vitro oBRB models have been developed, notably to investigate potential therapeutic strategies against retinal diseases. Indeed, to this day, most of these pathologies are untreatable, especially once the first signs of degeneration are observed. To cure those patients, a current strategy is to cultivate in vitro a mature oBRB epithelium on a custom membrane that is further implanted to replace the damaged native tissue. After a description of the oBRB and the related diseases, this review presents an overview of the oBRB models, from the simplest to the most complex. Then, we propose a discussion over the used cell types, for their relevance to study or treat the oBRB. Models designed for in vitro applications are then examined, by paying particular attention to the design evolution in the last years, the development of pathological models and the benefits of co-culture models, including both the retinal pigment epithelium and the choroid. Lastly, this review focuses on the models developed for in vivo implantation, with special emphasis on the choice of the material, its processing and its characterization, before discussing the reported pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Chloé Dujardin
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| | - Walter Habeler
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100, Corbeil-Essonnes, France
- U861, I-Stem, AFM, Université Paris-Saclay, Université D’Evry, 91100, Corbeil-Essonnes, France
- CECS, Centre D’étude des Cellules Souches, 91100, Corbeil-Essonnes, France
| | - Christelle Monville
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100, Corbeil-Essonnes, France
- U861, I-Stem, AFM, Université Paris-Saclay, Université D’Evry, 91100, Corbeil-Essonnes, France
| | - Didier Letourneur
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| |
Collapse
|
5
|
Molins B, Mesquida M, Adan A. Bioengineering approaches for modelling retinal pathologies of the outer blood-retinal barrier. Prog Retin Eye Res 2022:101097. [PMID: 35840488 DOI: 10.1016/j.preteyeres.2022.101097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022]
Abstract
Alterations of the junctional complex of the outer blood-retinal barrier (oBRB), which is integrated by the close interaction of the retinal pigment epithelium, the Bruch's membrane, and the choriocapillaris, contribute to the loss of neuronal signalling and subsequent vision impairment in several retinal inflammatory disorders such as age-related macular degeneration and diabetic retinopathy. Reductionist approaches into the mechanisms that underlie such diseases have been hindered by the absence of adequate in vitro models using human cells to provide the 3D dynamic architecture that enables expression of the in vivo phenotype of the oBRB. Conventional in vitro cell models are based on 2D monolayer cellular cultures, unable to properly recapitulate the complexity of living systems. The main drawbacks of conventional oBRB models also emerge from the cell sourcing, the lack of an appropriate Bruch's membrane analogue, and the lack of choroidal microvasculature with flow. In the last years, the advent of organ-on-a-chip, bioengineering, and stem cell technologies is providing more advanced 3D models with flow, multicellularity, and external control over microenvironmental properties. By incorporating additional biological complexity, organ-on-a-chip devices can mirror physiologically relevant properties of the native tissue while offering additional set ups to model and study disease. In this review we first examine the current understanding of oBRB biology as a functional unit, highlighting the coordinated contribution of the different components to barrier function in health and disease. Then we describe recent advances in the use of pluripotent stem cells-derived retinal cells, Bruch's membrane analogues, and co-culture techniques to recapitulate the oBRB. We finally discuss current advances and challenges of oBRB-on-a-chip technologies for disease modelling.
Collapse
Affiliation(s)
- Blanca Molins
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain.
| | - Marina Mesquida
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Alfredo Adan
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Instituto Clínic de Oftalmología, Hospital Clínic Barcelona, C/ Sabino de Arana 1, 08028, Barcelona, Spain
| |
Collapse
|
6
|
Gullapalli VK, Zarbin MA. New Prospects for Retinal Pigment Epithelium Transplantation. Asia Pac J Ophthalmol (Phila) 2022; 11:302-313. [PMID: 36041145 DOI: 10.1097/apo.0000000000000521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Retinal pigment epithelium (RPE) transplants rescue photoreceptors in selected animal models of retinal degenerative disease. Early clinical studies of RPE transplants as treatment for age-related macular degeneration (AMD) included autologous and allogeneic transplants of RPE suspensions and RPE sheets for atrophic and neovascular complications of AMD. Subsequent studies explored autologous RPE-Bruch membrane-choroid transplants in patients with neovascular AMD with occasional marked visual benefit, which establishes a rationale for RPE transplants in late-stage AMD. More recent work has involved transplantation of autologous and allogeneic stem cell-derived RPE for patients with AMD and those with Stargardt disease. These early-stage clinical trials have employed RPE suspensions and RPE monolayers on biocompatible scaffolds. Safety has been well documented, but evidence of efficacy is variable. Current research involves development of better scaffolds, improved modulation of immune surveillance, and modification of the extracellular milieu to improve RPE survival and integration with host retina.
Collapse
Affiliation(s)
| | - Marco A Zarbin
- Iinstitute of Ophthalmology and visual Science, Rutgers-New Jersey Medical School, Rutgers University, Newark, NJ, US
| |
Collapse
|
7
|
Tichotová L, Studenovska H, Petrovski G, Popelka Š, Nemesh Y, Sedláčková M, Drutovič S, Rohiwal S, Jendelová P, Erceg S, Brymová A, Artero‐Castro A, Lytvynchuk L, Straňák Z, Ellederová Z, Motlík J, Ardan T. Advantages of nanofibrous membranes for culturing of primary RPE cells compared to commercial scaffolds. Acta Ophthalmol 2021; 100:e1172-e1185. [PMID: 34687141 DOI: 10.1111/aos.15034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Dysfunction of the retinal pigment epithelium (RPE) causes numerous forms of retinal degeneration. RPE replacement is a modern option to save vision. We aimed to test the results of transplanting cultured RPEs on biocompatible membranes. METHODS We cultivated porcine primary RPE cells isolated from cadaver eyes from the slaughterhouse on two types of membranes: commercial polyester scaffolds Transwell (Corning Inc., Kenneburg, ME, USA) with 0.4 µm pore size and prepared Poly (L-lactide-co-DL-lactide) (PDLLA) nanofibrous membranes with an average pore size of 0.4 µm. RESULTS Five types of assays were used for the analysis: immunocytochemistry (ICC), phagocytosis assay, Western blotting, real-time qPCR (RT-qPCR) and electron microscopy. RT-qPCR demonstrated that RPEs cultured on nanofibrous membranes have higher expressions of BEST1 (bestrophin 1), RLBP1 (retinaldehyde-binding protein 1), RPE65 (retinal pigment epithelium-specific 65 kDa protein), PAX6 (transcription factor PAX6), SOX9 (transcription factor SOX9), DCT (dopachrome tautomerase) and MITF (microphthalmia-associated transcription factor). ICC of the RPEs cultured on nanofibrous membranes showed more intensive staining of markers such as BEST1, MCT1 (monocarboxylate transporter 1), Na+ /K+ ATPase, RPE65 and acetylated tubulin in comparison with commercial ones. Additionally, the absence of α-SMA proved the stability of the RPE polarization state and the absence of epithelial-to-mesenchymal transition. RPE possessed high phagocytic activity. Electron microscopy of both membranes confirmed a confluent layer of RPE cells and their genuine morphological structure, which was comparable to native RPEs. CONCLUSIONS Retinal pigment epitheliums cultured on polylactide nanofibrous membranes improved the final quality of the cell product by having better maturation and long-term survival of the RPE monolayer compared to those cultured on commercial polyester scaffolds. PDLLA-cultured RPEs are a plausible source for the replacement of non-functioning RPEs during cell therapy.
Collapse
Affiliation(s)
- Lucie Tichotová
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
- Department of Cell Biology Faculty of Science Charles University Prague Czech Republic
| | - Hana Studenovska
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic Prague Czech Republic
| | - Goran Petrovski
- Center for Eye Research Department of Ophthalmology Oslo University Hospital and Institute for Clinical Medicine University of Oslo Oslo Norway
| | - Štěpán Popelka
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic Prague Czech Republic
| | - Yaroslav Nemesh
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
- Department of Cell Biology Faculty of Science Charles University Prague Czech Republic
| | - Miroslava Sedláčková
- Department of Histology and Embryology Faculty of Medicine Masaryk University Brno Czech Republic
| | - Saskia Drutovič
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Sonali Rohiwal
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Pavla Jendelová
- Institute of Experimental Medicine Academy of Sciences of the Czech Republic Prague Czech Republic
| | - Slaven Erceg
- Institute of Experimental Medicine Academy of Sciences of the Czech Republic Prague Czech Republic
- Stem Cell Therapies in Neurodegenerative Diseases Lab Research Center ‘Principe Felipe’ Valencia Spain
| | - Anna Brymová
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
- Department of Cell Biology Faculty of Science Charles University Prague Czech Republic
| | - Ana Artero‐Castro
- Stem Cell Therapies in Neurodegenerative Diseases Lab Research Center ‘Principe Felipe’ Valencia Spain
| | - Lyubomyr Lytvynchuk
- Department of Ophthalmology Justus‐Liebig‐University Giessen University Hospital Giessen and Marburg Giessen Germany
| | - Zbyněk Straňák
- Ophthalmology Department of 3rd Faculty of Medicine Charles University and University Hospital Kralovske Vinohrady Prague Czech Republic
- Third Faculty of Medicine Charles University Prague Czech Republic
| | - Zdeňka Ellederová
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Jan Motlík
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Taras Ardan
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| |
Collapse
|
8
|
Khodamoradi M, Eskandari M, Keshvari H, Zarei R. An electro-conductive hybrid scaffold as an artificial Bruch's membrane. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112180. [PMID: 34082980 DOI: 10.1016/j.msec.2021.112180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 11/29/2022]
Abstract
Many research groups have investigated the various kinds of scaffolds to mimic the natural Bruch's membrane (BM) and support the retinal pigmented epithelial cells to form an organized cellular monolayer. While using prosthetic BM is identified as a promising treatment of age-related macular degeneration (AMD), a degenerative and progressive retinal disease, the effects of different signals such as electrical and morphological cues on the retinal pigmented epithelial (RPE) cells are still unknown. In this study, a laminated and conductive hydrogel/fiber composite scaffold by adding conductive polyaniline (PANi) to the scaffold's nanofibrous phase was prepared. This hybrid scaffold offers the closest morphology to the native structure of the human Bruch's membrane by imitating the inner and outer collagenous layer and induces the electrical signal to the scaffold to assess the electrical cue on behaviors of polarized retinal pigmented epithelial cells in the retina. The electrospun nanofibrous phase consisted of gelatin-Polyaniline in different ratios incorporated into the hydrogel precursor, a blend of gelatin and 4-armed PEG. We used a novel dual crosslinking process by incorporating the exposure of gamma irradiation and glutaraldehyde vapor treatment to construct the scaffold's hydrogel phase. The results showed the best composition was the sample which included the 40/60, Polyaniline/gelatin nanofiber sheets ratio because this scaffold revealed a 2.66 ± 0.33 MPa, Young's modulus and 1.84 ± 0.21 S/cm, electrochemical conductivity, which are close to the main features of native Bruch's membrane. In addition, this scaffold showed good biocompatibility by reaching 83.47% cell viability.
Collapse
Affiliation(s)
- Maedeh Khodamoradi
- Biomedical Engineering Faculty, Amirkabir University of Technology, Tehran, Iran
| | - Mahnaz Eskandari
- Biomedical Engineering Faculty, Amirkabir University of Technology, Tehran, Iran.
| | - Hamid Keshvari
- Biomedical Engineering Faculty, Amirkabir University of Technology, Tehran, Iran
| | - Reza Zarei
- Farabi Eye Hospital, Eye Research Center, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
9
|
The Evolution of Fabrication Methods in Human Retina Regeneration. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11094102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Optic nerve and retinal diseases such as age-related macular degeneration and inherited retinal dystrophies (IRDs) often cause permanent sight loss. Currently, a limited number of retinal diseases can be treated. Hence, new strategies are needed. Regenerative medicine and especially tissue engineering have recently emerged as promising alternatives to repair retinal degeneration and recover vision. Here, we provide an overview of retinal anatomy and diseases and a comprehensive review of retinal regeneration approaches. In the first part of the review, we present scaffold-free approaches such as gene therapy and cell sheet technology while in the second part, we focus on fabrication techniques to produce a retinal scaffold with a particular emphasis on recent trends and advances in fabrication techniques. To this end, the use of electrospinning, 3D bioprinting and lithography in retinal regeneration was explored.
Collapse
|
10
|
Kim J, Park JY, Kong JS, Lee H, Won JY, Cho DW. Development of 3D Printed Bruch's Membrane-Mimetic Substance for the Maturation of Retinal Pigment Epithelial Cells. Int J Mol Sci 2021; 22:ijms22031095. [PMID: 33499245 PMCID: PMC7865340 DOI: 10.3390/ijms22031095] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
Retinal pigment epithelium (RPE) is a monolayer of the pigmented cells that lies on the thin extracellular matrix called Bruch's membrane. This monolayer is the main component of the outer blood-retinal barrier (BRB), which plays a multifunctional role. Due to their crucial roles, the damage of this epithelium causes a wide range of diseases related to retinal degeneration including age-related macular degeneration, retinitis pigmentosa, and Stargardt disease. Unfortunately, there is presently no cure for these diseases. Clinically implantable RPE for humans is under development, and there is no practical examination platform for drug development. Here, we developed porcine Bruch's membrane-derived bioink (BM-ECM). Compared to conventional laminin, the RPE cells on BM-ECM showed enhanced functionality of RPE. Furthermore, we developed the Bruch's membrane-mimetic substrate (BMS) via the integration of BM-ECM and 3D printing technology, which revealed structure and extracellular matrix components similar to those of natural Bruch's membrane. The developed BMS facilitated the appropriate functions of RPE, including barrier and clearance functions, the secretion of anti-angiogenic growth factors, and enzyme formation for phototransduction. Moreover, it could be used as a basement frame for RPE transplantation. We established BMS using 3D printing technology to grow RPE cells with functions that could be used for an in vitro model and RPE transplantation.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (J.K.); (J.Y.P.); (H.L.)
| | - Ju Young Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (J.K.); (J.Y.P.); (H.L.)
| | - Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Hyungseok Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (J.K.); (J.Y.P.); (H.L.)
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon 24341, Korea
| | - Jae Yon Won
- Department of Ophthalmology and Visual Science, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03312, Korea
- Catholic Institute for Visual Science, College of Medicine, The Catholic University of Korea, Seoul 14662, Korea
- Correspondence: (J.Y.W.); (D.W.C.)
| | - Dong Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (J.K.); (J.Y.P.); (H.L.)
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
- Correspondence: (J.Y.W.); (D.W.C.)
| |
Collapse
|
11
|
Greferath U, Huynh M, Jobling AI, Vessey KA, Venables G, Surrao D, O'Neill HC, Limnios IJ, Fletcher EL. Dorsal-Ventral Differences in Retinal Structure in the Pigmented Royal College of Surgeons Model of Retinal Degeneration. Front Cell Neurosci 2021; 14:553708. [PMID: 33536874 PMCID: PMC7848141 DOI: 10.3389/fncel.2020.553708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 12/17/2020] [Indexed: 11/29/2022] Open
Abstract
Retinitis pigmentosa is a family of inherited retinal degenerations associated with gradual loss of photoreceptors, that ultimately leads to irreversible vision loss. The Royal College of Surgeon's (RCS) rat carries a recessive mutation affecting mer proto-oncogene tyrosine kinase (merTK), that models autosomal recessive disease. The aim of this study was to understand the glial, microglial, and photoreceptor changes that occur in different retinal locations with advancing disease. Pigmented RCS rats (RCS-p+/LAV) and age-matched isogenic control rdy (RCS-rdy +p+/LAV) rats aged postnatal day 18 to 6 months were evaluated for in vivo retinal structure and function using optical coherence tomography and electroretinography. Retinal tissues were assessed using high resolution immunohistochemistry to evaluate changes in photoreceptors, glia and microglia in the dorsal, and ventral retina. Photoreceptor dysfunction and death occurred from 1 month of age. There was a striking difference in loss of photoreceptors between the dorsal and ventral retina, with a greater number of photoreceptors surviving in the dorsal retina, despite being adjacent a layer of photoreceptor debris within the subretinal space. Loss of photoreceptors in the ventral retina was associated with fragmentation of the outer limiting membrane, extension of glial processes into the subretinal space that was accompanied by possible adhesion and migration of mononuclear phagocytes in the subretinal space. Overall, these findings highlight that breakdown of the outer limiting membrane could play an important role in exacerbating photoreceptor loss in the ventral retina. Our results also highlight the value of using the RCS rat to model sectorial retinitis pigmentosa, a disease known to predominantly effect the inferior retina.
Collapse
Affiliation(s)
- Una Greferath
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Mario Huynh
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew Ian Jobling
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Kirstan Anne Vessey
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Gene Venables
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Denver Surrao
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Helen Christine O'Neill
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Ioannis J Limnios
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Erica Lucy Fletcher
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
12
|
Bagewadi S, Parameswaran S, Krishnakumar S, Sethuraman S, Subramanian A. Tissue engineering approaches towards the regeneration of biomimetic scaffolds for age-related macular degeneration. J Mater Chem B 2021; 9:5935-5953. [PMID: 34254105 DOI: 10.1039/d1tb00976a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Age-related macular degeneration (AMD) is the third major cause of blindness in people aged above 60 years. It causes dysfunction of the retinal pigment epithelium (RPE) and leads to an irreversible loss of central vision. The present clinical treatment options are more palliative in controlling the progression of the disease and do not functionally restore the degenerated RPE monolayer and photoreceptors. Currently, the clinical transplantation of RPE cells has shown poor engraftment potential due to the absence of an intact Bruch's membrane in AMD patients, thereby the vision is unable to be restored completely. Although tissue engineering strategies target the development of Bruch's membrane-mimetic substrates, the challenge still lies in the development of an ultrathin, biologically and mechanically equivalent membrane to restore visual acuity. Further, existing limitations such as cellular aggregation, surgical complications including retinal tissue damage, tissue rejection, disease transmission, inferior mechanical strength, and the loss of vision over time demand the search for an ideal strategy to restore the functional RPE. Hence, this review aims to provide insights into various approaches, from conventional cell therapy to 3D bioprinting, and their unmet challenges in treating AMD by outlining the pathophysiology of AMD and the host tissue response with respect to injury, treatment and preclinical animal models.
Collapse
Affiliation(s)
- Shambhavi Bagewadi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India.
| | - Sowmya Parameswaran
- Radheshyam Kanoi Stem Cell Laboratory, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Vision Research Foundation, Chennai, India
| | - Subramanian Krishnakumar
- Radheshyam Kanoi Stem Cell Laboratory, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology Vision Research Foundation, Chennai, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India.
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India.
| |
Collapse
|
13
|
Ghareeb AE, Lako M, Steel DH. Coculture techniques for modeling retinal development and disease, and enabling regenerative medicine. Stem Cells Transl Med 2020; 9:1531-1548. [PMID: 32767661 PMCID: PMC7695644 DOI: 10.1002/sctm.20-0201] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/22/2020] [Accepted: 07/05/2020] [Indexed: 12/14/2022] Open
Abstract
Stem cell-derived retinal organoids offer the opportunity to cure retinal degeneration of wide-ranging etiology either through the study of in vitro models or the generation of tissue for transplantation. However, despite much work in animals and several human pilot studies, satisfactory therapies have not been developed. Two major challenges for retinal regenerative medicine are (a) physical cell-cell interactions, which are critical to graft function, are not formed and (b) the host environment does not provide suitable queues for development. Several strategies offer to improve the delivery, integration, maturation, and functionality of cell transplantation. These include minimally invasive delivery, biocompatible material vehicles, retinal cell sheets, and optogenetics. Optimizing several variables in animal models is practically difficult, limited by anatomical and disease pathology which is often different to humans, and faces regulatory and ethical challenges. High-throughput methods are needed to experimentally optimize these variables. Retinal organoids will be important to the success of these models. In their current state, they do not incorporate a representative retinal pigment epithelium (RPE)-photoreceptor interface nor vascular elements, which influence the neural retina phenotype directly and are known to be dysfunctional in common retinal diseases such as age-related macular degeneration. Advanced coculture techniques, which emulate the RPE-photoreceptor and RPE-Bruch's-choriocapillaris interactions, can incorporate disease-specific, human retinal organoids and overcome these drawbacks. Herein, we review retinal coculture models of the neural retina, RPE, and choriocapillaris. We delineate the scientific need for such systems in the study of retinal organogenesis, disease modeling, and the optimization of regenerative cell therapies for retinal degeneration.
Collapse
Affiliation(s)
- Ali E. Ghareeb
- Sunderland Eye Infirmary, South Tyneside and Sunderland NHS Foundation TrustSunderlandUK
- Biosciences Institute, Newcastle UniversityNewcastle‐upon‐TyneUK
| | - Majlinda Lako
- Biosciences Institute, Newcastle UniversityNewcastle‐upon‐TyneUK
| | - David H. Steel
- Sunderland Eye Infirmary, South Tyneside and Sunderland NHS Foundation TrustSunderlandUK
- Biosciences Institute, Newcastle UniversityNewcastle‐upon‐TyneUK
| |
Collapse
|
14
|
O'Neill HC, Limnios IJ, Barnett NL. Advancing a Stem Cell Therapy for Age-Related Macular Degeneration. Curr Stem Cell Res Ther 2020; 15:89-97. [PMID: 31854278 DOI: 10.2174/1574888x15666191218094020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/08/2019] [Accepted: 11/25/2019] [Indexed: 01/01/2023]
Abstract
The retinal pigment epithelium (RPE) is a multifunctional monolayer located at the back of the eye required for the survival and function of the light-sensing photoreceptors. In Age-related Macular Degeneration (AMD), the loss of RPE cells leads to photoreceptor death and permanent blindness. RPE cell transplantation aims to halt or reverse vision loss by preventing the death of photoreceptor cells and is considered one of the most viable applications of stem cell therapy in the field of regenerative medicine. Proof-of-concept of RPE cell transplantation for treating retinal degenerative disease, such as AMD, has long been established in animal models and humans using primary RPE cells, while recent research has focused on the transplantation of RPE cells derived from human pluripotent stem cells (hPSC). Early results from clinical trials indicate that transplantation of hPSC-derived RPE cells is safe and can improve vision in AMD patients. Current hPSC-RPE cell production protocols used in clinical trials are nevertheless inefficient. Treatment of large numbers of AMD patients using stem cellderived products may be dependent on the ability to generate functional cells from multiple hPSC lines using robust and clinically-compliant methods. Transplantation outcomes may be improved by delivering RPE cells on a thin porous membrane for better integration into the retina, and by manipulation of the outcome through control of immune rejection and inflammatory responses.
Collapse
Affiliation(s)
- Helen C O'Neill
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast 4229, Queensland, Australia
| | - Ioannis J Limnios
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast 4229, Queensland, Australia
| | - Nigel L Barnett
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast 4229, Queensland, Australia
| |
Collapse
|
15
|
Phelan MA, Kruczek K, Wilson JH, Brooks MJ, Drinnan CT, Regent F, Gerstenhaber JA, Swaroop A, Lelkes PI, Li T. Soy Protein Nanofiber Scaffolds for Uniform Maturation of Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium. Tissue Eng Part C Methods 2020; 26:433-446. [PMID: 32635833 DOI: 10.1089/ten.tec.2020.0072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Retinal pigment epithelium (RPE) differentiated from human induced pluripotent stem cells, called induced retinal pigment epithelium (iRPE), is being explored as a cell-based therapy for the treatment of retinal degenerative diseases, especially age-related macular degeneration. The success of RPE implantation is linked to the use of biomimetic scaffolds that simulate Bruch's membrane and promote RPE maturation and integration as a functional tissue. Due to difficulties associated with animal protein-derived scaffolds, including sterility and pro-inflammatory responses, current practices favor the use of synthetic polymers, such as polycaprolactone (PCL), for generating nanofibrous scaffolds. In this study, we tested the hypothesis that plant protein-derived fibrous scaffolds can provide favorable conditions permissive for the maturation of RPE tissue sheets in vitro. Our natural, soy protein-derived nanofibrous scaffolds exhibited a J-shaped stress-strain curve that more closely resembled the mechanical properties of native tissues than PCL with significantly higher hydrophilicity of the natural scaffolds, favoring in vivo implantation. We then demonstrate that iRPE sheets growing on these soy protein scaffolds are equivalent to iRPE monolayers cultured on synthetic PCL nanofibrous scaffolds. Immunohistochemistry demonstrated RPE-like morphology and functionality with appropriate localization of RPE markers RPE65, PMEL17, Ezrin, and ZO1 and with anticipated histotypic polarization of vascular endothelial growth factor and pigment epithelium-derived growth factor as indicated by enzyme-linked immunosorbent assay. Scanning electron microscopy revealed dense microvilli on the cell surface and homogeneous tight junctional contacts between the cells. Finally, comparative transcriptome analysis in conjunction with principal component analysis demonstrated that iRPE on nanofibrous scaffolds, either natural or synthetic, matured more consistently than on nonfibrous substrates. Taken together, our studies suggest that the maturation of cultured iRPE sheets for subsequent clinical applications might benefit from the use of nanofibrous scaffolds generated from natural proteins. Impact statement Induced retinal pigment epithelium (iRPE) from patient-derived induced pluripotent stem cells (iPSCs) may yield powerful treatments of retinal diseases, including age-related macular degeneration. Recent studies, including early human clinical trials, demonstrate the importance of selecting appropriate biomaterial scaffolds to support tissue-engineered iRPE sheets during implantation. Electrospun scaffolds show particular promise due to their similarity to the structure of the native Bruch's membrane. In this study, we describe the use of electroprocessed nanofibrous soy protein scaffolds to generate polarized sheets of human iPSC-derived iRPE sheets. Our evaluation, including RNA-seq transcriptomics, indicates that these scaffolds are viable alternatives to scaffolds electrospun from synthetic polymers.
Collapse
Affiliation(s)
- Michael A Phelan
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
- Integrated Laboratory for Cellular Tissue Engineering and Regenerative Medicine, Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - Kamil Kruczek
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John H Wilson
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew J Brooks
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Charles T Drinnan
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Florian Regent
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan A Gerstenhaber
- Integrated Laboratory for Cellular Tissue Engineering and Regenerative Medicine, Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter I Lelkes
- Integrated Laboratory for Cellular Tissue Engineering and Regenerative Medicine, Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - Tiansen Li
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
McCormick R, Pearce I, Kaye S, Haneef A. Optimisation of a Novel Bio-Substrate as a Treatment for Atrophic Age-Related Macular Degeneration. Front Bioeng Biotechnol 2020; 8:456. [PMID: 32500067 PMCID: PMC7243032 DOI: 10.3389/fbioe.2020.00456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/21/2020] [Indexed: 01/13/2023] Open
Abstract
Atrophic age-related macular degeneration (AMD) is the most common form of AMD accounting for 90% of patients. During atrophic AMD the waste/exchange pathway between the blood supply (choroid) and the retinal pigment epithelium (RPE) is compromised. This results in atrophy and death of the RPE cells and subsequently the photoreceptors leading to central blindness. Although the mechanisms behind AMD are unknown, the growth of fatty deposits known as drusen, have been shown to play a role in the disease. There is currently no treatment or cure for atrophic AMD. Much research focuses on developing a synthetic substrate in order to transplant healthy cells to the native Bruch’s membrane (BM), however, the diseased native BM and related structures still leave potential for transplanted cells to succumb to disease. In this proof-of-concept work we electrospun poly(ethylene terephthalate) (PET) to fabricate a nanofibrous cytocompatible synthetic BM. The apical surface of the membrane was cultured with ARPE-19 cells and the underside was decorated with poly(lactic acid-co-glycolic acid) (PLGA) or poly(glycolic acid) (PGA) degradable nanoparticles by electrospraying. The membrane exhibited hydrophilicity, high tensile strength and structurally resembled the native BM. ARPE-19 cells were able to form a monolayer on the surface of the membrane and no cell invasion into the membrane was seen. The presence of both PLGA and PGA nanoparticles increased ARPE-19 cell metabolism but had no effect on cell viability. There was a decrease in pH of ARPE-19 cell culture media 7 days following culturing with the PLGA nanoparticles but this change was eliminated by 2 weeks; PGA nanoparticles had no effect on cell culture media pH. The fluorescent dye FITC was encapsulated into nanoparticles and showed sustained release from PLGA nanoparticles for 2 weeks and PGA nanoparticles for 1 day. Future work will focus on encapsulating biologically active moieties to target drusen. This could allow this novel bioactive substrate to be a potential treatment for atrophic AMD that would function two-fold: deliver the required monolayer of healthy RPE cells to the macula on a synthetic BM and remove diseased structures within the retina, restoring the waste/exchange pathway and preventing vision loss.
Collapse
Affiliation(s)
- Rachel McCormick
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Ian Pearce
- St Paul's Eye Unit, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Stephen Kaye
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,St Paul's Eye Unit, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Atikah Haneef
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
17
|
Murphy AR, Truong YB, O'Brien CM, Glattauer V. Bio-inspired human in vitro outer retinal models: Bruch's membrane and its cellular interactions. Acta Biomater 2020; 104:1-16. [PMID: 31945506 DOI: 10.1016/j.actbio.2020.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/17/2022]
Abstract
Retinal degenerative disorders, such as age-related macular degeneration (AMD), are one of the leading causes of blindness worldwide, however, treatments to completely stop the progression of these debilitating conditions are non-existent. Researchers require sophisticated models that can accurately represent the native structure of human retinal tissue to study these disorders. Current in vitro models used to study the retina are limited in their ability to fully recapitulate the structure and function of the retina, Bruch's membrane and the underlying choroid. Recent developments in the field of induced pluripotent stem cell technology has demonstrated the capability of retinal pigment epithelial cells to recapitulate AMD-like pathology. However, such studies utilise unsophisticated, bio-inert membranes to act as Bruch's membrane and support iPSC-derived retinal cells. This review presents a concise summary of the properties and function of the Bruch's membrane-retinal pigment epithelium complex, the initial pathogenic site of AMD as well as the current status for materials and fabrication approaches used to generate in vitro models of this complex tissue. Finally, this review explores required advances in the field of in vitro retinal modelling. STATEMENT OF SIGNIFICANCE: Retinal degenerative disorders such as age-related macular degeneration are worldwide leading causes of blindness. Previous attempts to model the Bruch's membrane-retinal pigment epithelial complex, the initial pathogenic site of age-related macular degeneration, have lacked the sophistication to elucidate valuable insights into disease mechanisms. Here we provide a detailed account of the morphological, physical and chemical properties of Bruch's membrane which may aid the fabrication of more sophisticated and physiologically accurate in vitro models of the retina, as well as various fabrication techniques to recreate this structure. This review also further highlights some recent advances in some additional challenging aspects of retinal tissue modelling including integrated fluid flow and photoreceptor alignment.
Collapse
Affiliation(s)
- Ashley R Murphy
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia.
| | - Yen B Truong
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia
| | - Carmel M O'Brien
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and Innovation Precinct (STRIP), Monash University, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia
| | | |
Collapse
|
18
|
Sahle FF, Kim S, Niloy KK, Tahia F, Fili CV, Cooper E, Hamilton DJ, Lowe TL. Nanotechnology in regenerative ophthalmology. Adv Drug Deliv Rev 2019; 148:290-307. [PMID: 31707052 PMCID: PMC7474549 DOI: 10.1016/j.addr.2019.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/18/2022]
Abstract
In recent years, regenerative medicine is gaining momentum and is giving hopes for restoring function of diseased, damaged, and aged tissues and organs and nanotechnology is serving as a catalyst. In the ophthalmology field, various types of allogenic and autologous stem cells have been investigated to treat some ocular diseases due to age-related macular degeneration, glaucoma, retinitis pigmentosa, diabetic retinopathy, and corneal and lens traumas. Nanomaterials have been utilized directly as nanoscaffolds for these stem cells to promote their adhesion, proliferation and differentiation or indirectly as vectors for various genes, tissue growth factors, cytokines and immunosuppressants to facilitate cell reprogramming or ocular tissue regeneration. In this review, we reviewed various nanomaterials used for retina, cornea, and lens regenerations, and discussed the current status and future perspectives of nanotechnology in tracking cells in the eye and personalized regenerative ophthalmology. The purpose of this review is to provide comprehensive and timely insights on the emerging field of nanotechnology for ocular tissue engineering and regeneration.
Collapse
Affiliation(s)
- Fitsum Feleke Sahle
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Sangyoon Kim
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Kumar Kulldeep Niloy
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Faiza Tahia
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Cameron V Fili
- Department of Comparative Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Emily Cooper
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - David J Hamilton
- Department of Comparative Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Tao L Lowe
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA.
| |
Collapse
|