1
|
Kamachi Kobashigawa K, Franchi de Barros Sobrinho AA, Espírito Santo Silva P, Siqueira Vasconcelos CR, Cardoso Cristovam P, Pereira Gomes JÁ, Laus JL, Aldrovani Rodrigues M. Enhancing Ex Vivo Limbal Epithelial Cell Expansion on Amniotic Membrane: A Comparative Study of Monolayer (2D) Versus Sandwich (3D) Culture Configurations. Cornea 2024:00003226-990000000-00734. [PMID: 39509280 DOI: 10.1097/ico.0000000000003753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE This study compared two-dimensional (monolayer) and three-dimensional (sandwich) systems for expanding ex vivo limbal epithelial cells on amniotic membrane and evaluated the outcomes after transplantation into rabbits with experimentally induced limbal stem cell deficiency. METHODS Evaluations included markers for progenitor cells, proliferation, apoptosis, and clinical monitoring for up to 63 days. In the monolayer culture, epithelial cells derived from limbal explants were expanded on amniotic membrane as the substrate. In the sandwich culture, the cells were cultured between 2 layers of amniotic membrane. Evaluations included markers for progenitor cells, proliferation, and apoptosis, along with clinical monitoring for up to 63 days. RESULTS Sandwich cultures demonstrated increased cellular proliferation and fewer progenitor cells compared with monolayer cultures. In treating limbal stem cell deficiency, the group receiving transplantation from sandwich cultures exhibited reduced neovascularization and decreased corneal ulceration compared with those treated with monolayer cultures, with similar clinical outcomes in corneal opacity. The configuration of the culture system did not affect the presence of apoptotic cells. Corneas treated with sandwich cultures showed a higher presence of progenitor cells compared with the monolayer group, suggesting a potential long-term viability advantage for these transplants. CONCLUSIONS In conclusion, although the sandwich culture system enhanced cellular proliferation, it also resulted in a decrease in progenitor cells within the cultures. Nevertheless, both systems demonstrated comparable therapeutic efficacy in treating limbal stem cell deficiency, with the sandwich approach potentially offering long-term benefits because of the increased presence of progenitor cells in the transplanted cornea.
Collapse
Affiliation(s)
- Karina Kamachi Kobashigawa
- Department of Veterinary Clinic and Surgery, São Paulo State University (UNESP), Jaboticabal, SP, Brazil
| | | | | | | | - Priscila Cardoso Cristovam
- Advanced Center of Ocular Surface (CASO), Department of Ophthalmology and Visual Science, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - José Álvaro Pereira Gomes
- Advanced Center of Ocular Surface (CASO), Department of Ophthalmology and Visual Science, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - José Luiz Laus
- Department of Veterinary Clinic and Surgery, São Paulo State University (UNESP), Jaboticabal, SP, Brazil
| | | |
Collapse
|
2
|
Wang F, Xu Y, Zhou Q, Xie L. Biomolecule-based hydrogels as delivery systems for limbal stem cell transplantation: A review. Int J Biol Macromol 2024; 280:135778. [PMID: 39304050 DOI: 10.1016/j.ijbiomac.2024.135778] [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: 03/25/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Limbal stem cell deficiency (LSCD) is a complex disease of the cornea resulting from dysfunction and/or loss of limbal stem cells (LSCs) and their niche. Most patients with LSCD cannot be treated by conventional corneal transplants because the donor tissue lacks the LSCs necessary for corneal epithelial regeneration. Successful treatment of LSCD depends on effective stem cell transplantation to the ocular surface for replenishment of the LSC reservoir. Thus, stem cell therapies employing carrier substrates for LSCs have been widely explored. Hydrogel biomaterials have many favorable characteristics, including hydrophilicity, flexibility, cytocompatibility, and optical properties suitable for the transplantation of LSCs. Therefore, due to these properties, along with the necessary signals for stem cell proliferation and differentiation, hydrogels are ideal carrier substrates for LSCD treatment. This review summarizes the use of different medical-type hydrogels in LSC transplantation from 2001 to 2024. First, a brief background of LSCD is provided. Then, studies that employed various hydrogel scaffolds as LSC carriers are highlighted to provide a multimodal strategic reference for LSCD treatment. Finally, an analysis of prospective future developments and challenges in the field of hydrogels as LSC carriers for treating LSCD is presented.
Collapse
Affiliation(s)
- Fuyan Wang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China
| | - Yuehe Xu
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China.
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China.
| |
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
|
Dimmock RL, Rotherham M, El Haj AJ, Yang Y. Fabrication and Characterisation of Hydrogels with Reversible Wrinkled Surfaces for Limbal Study and Reconstruction. Gels 2023; 9:915. [PMID: 37999005 PMCID: PMC10671082 DOI: 10.3390/gels9110915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
In the biomedical field, there is a demand for the development of novel approaches for the investigation of optical epithelial anatomical features with biomimetic materials. These materials are not only required to replicate structures but also enable dynamic modelling for disease states such as limbal stem cell deficiency and ageing. In the present study, the effective generation of reversible wrinkled polydimethylsiloxane (PDMS) substrates was undertaken to mimic the undulating anatomy of the limbal epithelial stem cell niche. This undulating surface pattern was formed through a dual treatment with acid oxidation and plasma using an innovatively designed stretching frame. This system enabled the PDMS substrate to undergo deformation and relaxation, creating a reversible and tuneable wrinkle pattern with cell culture applications. The crypt-like pattern exhibited a width of 70-130 µm and a depth of 17-40 µm, resembling the topography of a limbal epithelial stem cell niche, which is characterised by an undulating anatomy. The cytocompatibility of the patterned substrate was markedly improved using a gelatin methacrylate polymer (GelMa) coating. It was also observed that these wrinkled PDMS surfaces were able to dictate cell growth patterns, showing alignment in motile cells and colony segregation in colony-forming cells when using human and porcine limbal cells, respectively.
Collapse
Affiliation(s)
- Ryan L. Dimmock
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK
| | - Michael Rotherham
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| | - Alicia J. El Haj
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK
| |
Collapse
|
5
|
The progress in techniques for culturing human limbal epithelial stem cells. Hum Cell 2023; 36:1-14. [PMID: 36181663 DOI: 10.1007/s13577-022-00794-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/11/2022] [Indexed: 01/07/2023]
Abstract
In vitro culture of human limbal epithelial stem cells (hLESCs) is crucial to cell therapy in the treatment of limbal stem cell deficiency, a potentially vision-threatening disease that is characterized by persistent corneal epithelial defects and corneal epithelium conjunctivalization. Traditionally, hLESCs are cultivated based on either limbal tissue explants or single-cell suspensions in culture media containing xenogenous components, such as fetal bovine serum and murine 3T3 feeder cells. Plastic culture dishes and human amniotic membranes are classical growth substrates used in conventional hLESC culture systems. The past few decades have witnessed considerable progress and innovations in hLESC culture techniques to ensure a higher level of biosafety and lower immunogenicity for further cell treatment, including complete removal of xenogenous components from culture media, the application of human-derived feeder cells, and the development of novel scaffolds. Three-dimensional artificial niches and three-dimensional culture techniques have also been established to simulate the real microenvironment of limbal crypts for better cell outgrowth and proliferation. All these progresses ensure that in vitro cultured hLESCs are more adaptable to translational stem cell therapy for limbal stem cell deficiency.
Collapse
|
6
|
Kalaimani L, Devarajan B, Namperumalsamy VP, Veerappan M, Daniels JT, Chidambaranathan GP. Hsa-miR-143-3p inhibits Wnt-β-catenin and MAPK signaling in human corneal epithelial stem cells. Sci Rep 2022; 12:11432. [PMID: 35794158 PMCID: PMC9259643 DOI: 10.1038/s41598-022-15263-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/21/2022] [Indexed: 11/09/2022] Open
Abstract
Our previous study demonstrated hsa-miR-143-3p as one of the highly expressed miRNAs in enriched corneal epithelial stem cells (CESCs). Hence this study aims to elucidate the regulatory role of hsa-miR-143-3p in the maintenance of stemness in CESCs. The target genes of hsa-miR-143-3p were predicted and subjected to pathway analysis to select the targets for functional studies. Primary cultured limbal epithelial cells were transfected with hsa-miR-143-3p mimic, inhibitor or scrambled sequence using Lipofectamine 3000. The transfected cells were analysed for (i) colony forming potential, (ii) expression of stem cell (SC) markers/ transcription factors (ABCG2, NANOG, OCT4, KLF4, ΔNp63), (iii) differentiation marker (Cx43), (iv) predicted five targets of hsa-miR-143-3p (DVL3, MAPK1, MAPK14, KRAS and KAT6A), (v) MAPK signaling regulators and (vi) Wnt-β-catenin signaling regulators by qPCR, immunofluorescence staining and/or Western blotting. High expression of hsa-miR-143-3p increased the colony forming potential (10.04 ± 1.35%, p < 0.001) with the ability to form holoclone-like colonies in comparison to control (3.33 ± 0.71%). The mimic treated cells had increased expression of SC markers but reduced expression of Cx43 and hsa-miR-143-3p targets involved in Wnt-β-catenin and MAPK signaling pathways. The expression of β-catenin, active β-catenin and ERK2 in hsa-miR-143-3p inhibitor transfected cells were higher than the control cells and the localized nuclear expression indicated the activation of Wnt and MAPK signaling. Thus, the probable association of hsa-miR-143-3p in the maintenance of CESCs through inhibition of Wnt and MAPK signaling pathways was thus indicated.
Collapse
Affiliation(s)
- Lavanya Kalaimani
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India.,Department of Biotechnology, Aravind Medical Research Foundation-Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India.,Institute of Ophthalmology, University College London, London, UK
| | - Bharanidharan Devarajan
- Department of Bioinformatics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | | | - Muthukkaruppan Veerappan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India
| | - Julie T Daniels
- Institute of Ophthalmology, University College London, London, UK
| | - Gowri Priya Chidambaranathan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India. .,Department of Biotechnology, Aravind Medical Research Foundation-Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
7
|
Hidalgo-Alvarez V, Dhowre HS, Kingston OA, Sheridan CM, Levis HJ. Biofabrication of Artificial Stem Cell Niches in the Anterior Ocular Segment. Bioengineering (Basel) 2021; 8:135. [PMID: 34677208 PMCID: PMC8533470 DOI: 10.3390/bioengineering8100135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
The anterior segment of the eye is a complex set of structures that collectively act to maintain the integrity of the globe and direct light towards the posteriorly located retina. The eye is exposed to numerous physical and environmental insults such as infection, UV radiation, physical or chemical injuries. Loss of transparency to the cornea or lens (cataract) and dysfunctional regulation of intra ocular pressure (glaucoma) are leading causes of worldwide blindness. Whilst traditional therapeutic approaches can improve vision, their effect often fails to control the multiple pathological events that lead to long-term vision loss. Regenerative medicine approaches in the eye have already had success with ocular stem cell therapy and ex vivo production of cornea and conjunctival tissue for transplant recovering patients' vision. However, advancements are required to increase the efficacy of these as well as develop other ocular cell therapies. One of the most important challenges that determines the success of regenerative approaches is the preservation of the stem cell properties during expansion culture in vitro. To achieve this, the environment must provide the physical, chemical and biological factors that ensure the maintenance of their undifferentiated state, as well as their proliferative capacity. This is likely to be accomplished by replicating the natural stem cell niche in vitro. Due to the complex nature of the cell microenvironment, the creation of such artificial niches requires the use of bioengineering techniques which can replicate the physico-chemical properties and the dynamic cell-extracellular matrix interactions that maintain the stem cell phenotype. This review discusses the progress made in the replication of stem cell niches from the anterior ocular segment by using bioengineering approaches and their therapeutic implications.
Collapse
Affiliation(s)
- Veronica Hidalgo-Alvarez
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Hala S. Dhowre
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Olivia A. Kingston
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Carl M. Sheridan
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Hannah J. Levis
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| |
Collapse
|
8
|
Abdul-Al M, Kyeremeh GK, Saeinasab M, Heidari Keshel S, Sefat F. Stem Cell Niche Microenvironment: Review. Bioengineering (Basel) 2021; 8:bioengineering8080108. [PMID: 34436111 PMCID: PMC8389324 DOI: 10.3390/bioengineering8080108] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
The cornea comprises a pool of self-regenerating epithelial cells that are crucial to preserving clarity and visibility. Limbal epithelial stem cells (LESCs), which live in a specialized stem cell niche (SCN), are crucial for the survival of the human corneal epithelium. They live at the bottom of the limbal crypts, in a physically enclosed microenvironment with a number of neighboring niche cells. Scientists also simplified features of these diverse microenvironments for more analysis in situ by designing and recreating features of different SCNs. Recent methods for regenerating the corneal epithelium after serious trauma, including burns and allergic assaults, focus mainly on regenerating the LESCs. Mesenchymal stem cells, which can transform into self-renewing and skeletal tissues, hold immense interest for tissue engineering and innovative medicinal exploration. This review summarizes all types of LESCs, identity and location of the human epithelial stem cells (HESCs), reconstruction of LSCN and artificial stem cells for self-renewal.
Collapse
Affiliation(s)
- Mohamed Abdul-Al
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD71DP, UK; (M.A.-A.); (G.K.K.)
| | - George Kumi Kyeremeh
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD71DP, UK; (M.A.-A.); (G.K.K.)
| | - Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 91779 48974, Iran;
| | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839 69411, Iran;
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD71DP, UK; (M.A.-A.); (G.K.K.)
- Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford BD71DP, UK
- Correspondence:
| |
Collapse
|
9
|
Ramos-Rodriguez DH, MacNeil S, Claeyssens F, Ortega Asencio I. Fabrication of Topographically Controlled Electrospun Scaffolds to Mimic the Stem Cell Microenvironment in the Dermal-Epidermal Junction. ACS Biomater Sci Eng 2021; 7:2803-2813. [PMID: 33905240 DOI: 10.1021/acsbiomaterials.0c01775] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of microfabrication techniques for the development of innovative constructs for tissue regeneration is a growing area of research. This area comprises both manufacturing and biological approaches for the development of smart materials aiming to control and direct cell behavior to enhance tissue healing. Many groups have focused their efforts on introducing complexity within these innovative constructs via the inclusion of nano- and microtopographical cues mimicking physical and biological aspects of the native stem cell niche. Specifically, in the area of skin tissue engineering, seminal work has reported replicating the microenvironments located in the dermal-epithelial junction, which are known as rete ridges. The rete ridges are key for both stem cell control and the physiological performance of the skin. In this work, we have introduced complexity within electrospun membranes to mimic the morphology of the rete ridges in the skin. We designed and tested three different patterns, characterized them, and explored their performance in vitro, using 3D skin models. One of the studied patterns (pattern B) was shown to aid in the development of an in vitro rite-ridgelike skin model that resulted in the expression of relevant epithelial markers such as collagen IV and integrin β1. In summary, we have developed a new skin model including synthetic rete-ridgelike structures that replicate both morphology and function of the native dermal-epidermal junction and that offer new insights for the development of smart skin tissue engineering constructs.
Collapse
Affiliation(s)
- David H Ramos-Rodriguez
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, U.K
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, U.K
| | - Sheila MacNeil
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, U.K
| | - Frederik Claeyssens
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, U.K
| | - Ilida Ortega Asencio
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, U.K
| |
Collapse
|
10
|
Ramos-Rodriguez DH, MacNeil S, Claeyssens F, Asencio IO. The Use of Microfabrication Techniques for the Design and Manufacture of Artificial Stem Cell Microenvironments for Tissue Regeneration. Bioengineering (Basel) 2021; 8:50. [PMID: 33922428 PMCID: PMC8146165 DOI: 10.3390/bioengineering8050050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
The recapitulation of the stem cell microenvironment is an emerging area of research that has grown significantly in the last 10 to 15 years. Being able to understand the underlying mechanisms that relate stem cell behavior to the physical environment in which stem cells reside is currently a challenge that many groups are trying to unravel. Several approaches have attempted to mimic the biological components that constitute the native stem cell niche, however, this is a very intricate environment and, although promising advances have been made recently, it becomes clear that new strategies need to be explored to ensure a better understanding of the stem cell niche behavior. The second strand in stem cell niche research focuses on the use of manufacturing techniques to build simple but functional models; these models aim to mimic the physical features of the niche environment which have also been demonstrated to play a big role in directing cell responses. This second strand has involved a more engineering approach in which a wide set of microfabrication techniques have been explored in detail. This review aims to summarize the use of these microfabrication techniques and how they have approached the challenge of mimicking the native stem cell niche.
Collapse
Affiliation(s)
- David H. Ramos-Rodriguez
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK;
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK; (S.M.); (F.C.)
| | - Sheila MacNeil
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK; (S.M.); (F.C.)
| | - Frederik Claeyssens
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK; (S.M.); (F.C.)
| | - Ilida Ortega Asencio
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK;
| |
Collapse
|
11
|
Gumus K. On the Horizon: Biologics and Nutrients for Neurotrophic Keratitis. Eye Contact Lens 2021; 47:154-156. [PMID: 33156130 DOI: 10.1097/icl.0000000000000757] [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] [Accepted: 09/27/2020] [Indexed: 10/23/2022]
Abstract
ABSTRACT Neurotrophic keratitis (NK), a potentially sight-threatening corneal disease, still does not have a specific treatment. The reduction or complete loss of corneal sensation, the most important factor in its pathogenesis, is one of the most important factors that complicate the treatment of corneal wound healing. In addition, the visual outcome may be adversely affected because of aggressive stromal fibrosis in severe NK cases. Basically, the current management of NK aims to accelerate wound healing and prevent the progression. However, new therapeutic agents, particularly developed depending on cell type-specific healing mechanisms are required for better visual outcomes. In recent years, several studies have started to use new promising areas of translational research, including gene therapy, stem cell therapy, miRNA, and bioengineering. Evidence has emerged that future treatment strategies for NK will be designed by the results of these studies. In this review, it is aimed to summarize scientific data of new treatment modalities for NK.
Collapse
Affiliation(s)
- Koray Gumus
- Ophthalmology, Memorial Ankara Hospital, Eye Clinic, Ankara, Turkey
| |
Collapse
|
12
|
Robust Topographical Micro-Patterning of Nanofibrillar Collagen Gel by In Situ Photochemical Crosslinking-Assisted Collagen Embossing. NANOMATERIALS 2020; 10:nano10122574. [PMID: 33371525 PMCID: PMC7767481 DOI: 10.3390/nano10122574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022]
Abstract
The topographical micro-patterning of nanofibrillar collagen gels is promising for the fabrication of biofunctional constructs mimicking topographical cell microenvironments of in vivo extracellular matrices. Nevertheless, obtaining structurally robust collagen micro-patterns through this technique is still a challenging issue. Here, we report a novel in situ photochemical crosslinking-assisted collagen embossing (IPC-CE) process as an integrative fabrication technique based on collagen compression-based embossing and UV–riboflavin crosslinking. The IPC-CE process using a micro-patterned polydimethylsiloxane (PDMS) master mold enables the compaction of collagen nanofibrils into micro-cavities of the mold and the simultaneous occurrence of riboflavin-mediated photochemical reactions among the nanofibrils, resulting in a robust micro-patterned collagen construct. The micro-patterned collagen construct fabricated through the IPC-CE showed a remarkable mechanical resistivity against rehydration and manual handling, which could not be achieved through the conventional collagen compression-based embossing alone. Micro-patterns of various sizes (minimum feature size <10 μm) and shapes could be obtained by controlling the compressive pressure (115 kPa) and the UV dose (3.00 J/cm2) applied during the process. NIH 3T3 cell culture on the micro-patterned collagen construct finally demonstrated its practical applicability in biological applications, showing a notable effect of anisotropic topography on cells in comparison with the conventional construct.
Collapse
|
13
|
García-Posadas L, Diebold Y. Three-Dimensional Human Cell Culture Models to Study the Pathophysiology of the Anterior Eye. Pharmaceutics 2020; 12:E1215. [PMID: 33333869 PMCID: PMC7765302 DOI: 10.3390/pharmaceutics12121215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 02/07/2023] Open
Abstract
In recent decades, the establishment of complex three-dimensional (3D) models of tissues has allowed researchers to perform high-quality studies and to not only advance knowledge of the physiology of these tissues but also mimic pathological conditions to test novel therapeutic strategies. The main advantage of 3D models is that they recapitulate the spatial architecture of tissues and thereby provide more physiologically relevant information. The eye is an extremely complex organ that comprises a large variety of highly heterogeneous tissues that are divided into two asymmetrical portions: the anterior and posterior segments. The anterior segment consists of the cornea, conjunctiva, iris, ciliary body, sclera, aqueous humor, and the lens. Different diseases in these tissues can have devastating effects. To study these pathologies and develop new treatments, the use of cell culture models is instrumental, and the better the model, the more relevant the results. Thus, the development of sophisticated 3D models of ocular tissues is a significant challenge with enormous potential. In this review, we present a comprehensive overview of the latest advances in the development of 3D in vitro models of the anterior segment of the eye, with a special focus on those that use human primary cells.
Collapse
Affiliation(s)
- Laura García-Posadas
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain;
| | - Yolanda Diebold
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Nosrati H, Alizadeh Z, Nosrati A, Ashrafi-Dehkordi K, Banitalebi-Dehkordi M, Sanami S, Khodaei M. Stem cell-based therapeutic strategies for corneal epithelium regeneration. Tissue Cell 2020; 68:101470. [PMID: 33248403 DOI: 10.1016/j.tice.2020.101470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Any significant loss of vision or blindness caused by corneal damages is referred to as corneal blindness. Corneal blindness is the fourth most common cause of blindness worldwide, representing more than 5% of the total blind population. Currently, corneal transplantation is used to treat many corneal diseases. In some cases, implantation of artificial cornea (keratoprosthesis) is suggested after a patient has had a donor corneal transplant failure. The shortage of donors and the side effects of keratoprosthesis are limiting these approaches. Recently, researchers have been actively pursuing new approaches for corneal regeneration because of these limitations. Nowadays, tissue engineering of different corneal layers (epithelium, stroma, endothelium, or full thickness tissue) is a promising approach that has attracted a great deal of interest from researchers and focuses on regenerative strategies using different cell sources and biomaterials. Various sources of corneal and non-corneal stem cells have shown significant advantages for corneal epithelium regeneration applications. Pluripotent stem cells (embryonic stem cells and iPS cells), epithelial stem cells (derived from oral mucus, amniotic membrane, epidermis and hair follicle), mesenchymal stem cells (bone marrow, adipose-derived, amniotic membrane, placenta, umbilical cord), and neural crest origin stem cells (dental pulp stem cells) are the most promising sources in this regard. These cells could also be used in combination with natural or synthetic scaffolds to improve the efficacy of the therapeutic approach. As the ocular surface is exposed to external damage, the number of studies on regeneration of the corneal epithelium is rising. In this paper, we reviewed the stem cell-based strategies for corneal epithelium regeneration.
Collapse
Affiliation(s)
- Hamed Nosrati
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Zohreh Alizadeh
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Nosrati
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Korosh Ashrafi-Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mehdi Banitalebi-Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Sanami
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Khodaei
- Department of Materials Science and Engineering, Golpayegan University of Technology, Golpayegan, Iran
| |
Collapse
|
15
|
Alió Del Barrio JL, Arnalich-Montiel F, De Miguel MP, El Zarif M, Alió JL. Corneal stroma regeneration: Preclinical studies. Exp Eye Res 2020; 202:108314. [PMID: 33164825 DOI: 10.1016/j.exer.2020.108314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
Corneal grafting is one of the most common and successful forms of human tissue transplantation in the world, but the need for corneal grafting is growing and availability of human corneal donor tissue to fulfill this increasing demand is not assured worldwide. The stroma is responsible for many features of the cornea, including its strength, refractive power and transparency, so enormous efforts have been put into replicating the corneal stroma in the laboratory to find an alternative to classical corneal transplantation. Unfortunately this has not been yet accomplished due to the extreme difficulty in mimicking the highly complex ultrastructure of the corneal stroma, and none of the obtained substitutes that have been assayed has been able to replicate this complexity yet. In general, they can neither match the mechanical properties nor recreate the local nanoscale organization and thus the transparency and optical properties of a normal cornea. In this context, there is an increasing interest in cellular therapy of the corneal stroma using Induced Pluripotent Stem Cells (iPSCs) or mesenchymal stem cells (MSCs) from either ocular or extraocular sources, as they have proven to be capable of producing new collagen within the host stroma, modulate preexisting scars and enhance transparency by corneal stroma remodeling. Despite some early clinical data is already available, in the current article we will summary the available preclinical evidence about the topic corneal stroma regeneration. Both, in vitro and in vivo experiments in the animal model will be shown.
Collapse
Affiliation(s)
- Jorge L Alió Del Barrio
- Cornea, Cataract and Refractive Surgery Unit, Vissum (Miranza Group), Alicante, Spain; Division of Ophthalmology, Universidad Miguel Hernández, Alicante, Spain
| | - Francisco Arnalich-Montiel
- IRYCIS. Ophthalmology Department. Ramón y Cajal University Hospital, Madrid, Spain; Cornea Unit. Hospital Vissum Madrid (Miranza Group), Madrid, Spain
| | - María P De Miguel
- Cell Engineering Laboratory, IdiPAZ, La Paz Hospital Research Institute, Madrid, Spain
| | | | - Jorge L Alió
- Cornea, Cataract and Refractive Surgery Unit, Vissum (Miranza Group), Alicante, Spain; Division of Ophthalmology, Universidad Miguel Hernández, Alicante, Spain.
| |
Collapse
|
16
|
O'Callaghan AR, Dziasko MA, Sheth-Shah R, Lewis MP, Daniels JT. Oral Mucosa Tissue Equivalents for the Treatment of Limbal Stem Cell Deficiency. ACTA ACUST UNITED AC 2020; 4:e1900265. [PMID: 32515079 DOI: 10.1002/adbi.201900265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Cultured limbal and oral epithelial cells have been successfully used to treat patients with limbal stem cell deficiency (LSCD). The most common culture method for these cell therapies utilizes amniotic membrane as a cell support and/or murine 3T3s as feeder fibroblasts. The aim of this study is to refine the production of autologous oral mucosal cell therapy for the treatment of LSCD. Real architecture for 3D tissue (RAFT) is used as an alternative cell culture support. In addition, oral mucosal cells (epithelial and fibroblast) are used as autologous alternatives to donor human limbal epithelial cells (HLE) and murine 3T3s. The following tissue equivalents are produced and characterized: first, for patients with bilateral LSCD, an oral mucosa tissue equivalent consisting of human oral mucosal epithelial cells on RAFT supported by human oral mucosal fibroblasts (HOMF). Second, for patients with unilateral LSCD, HLE on RAFT supported by HOMF. For both tissue equivalent types, features of the cornea are observed including a multi-layered epithelium with small cells with a stem cell like phenotype in the basal layer and squamous cells in the top layers, and p63α and PAX6 expression. These tissue equivalents may therefore be useful in the treatment of LSCD.
Collapse
Affiliation(s)
- Anna R O'Callaghan
- Cells for Sight, UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Marc A Dziasko
- Cells for Sight, UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Radhika Sheth-Shah
- Cells for Sight, UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Mark P Lewis
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University, Leicestershire, LE11 3TU, UK
| | - Julie T Daniels
- Cells for Sight, UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| |
Collapse
|
17
|
Mobaraki M, Abbasi R, Omidian Vandchali S, Ghaffari M, Moztarzadeh F, Mozafari M. Corneal Repair and Regeneration: Current Concepts and Future Directions. Front Bioeng Biotechnol 2019; 7:135. [PMID: 31245365 PMCID: PMC6579817 DOI: 10.3389/fbioe.2019.00135] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
The cornea is a unique tissue and the most powerful focusing element of the eye, known as a window to the eye. Infectious or non-infectious diseases might cause severe visual impairments that need medical intervention to restore patients' vision. The most prominent characteristics of the cornea are its mechanical strength and transparency, which are indeed the most important criteria considerations when reconstructing the injured cornea. Corneal strength comes from about 200 collagen lamellae which criss-cross the cornea in different directions and comprise nearly 90% of the thickness of the cornea. Regarding corneal transparency, the specific characteristics of the cornea include its immune and angiogenic privilege besides its limbus zone. On the other hand, angiogenic privilege involves several active cascades in which anti-angiogenic factors are produced to compensate for the enhanced production of proangiogenic factors after wound healing. Limbus of the cornea forms a border between the corneal and conjunctival epithelium, and its limbal stem cells (LSCs) are essential in maintenance and repair of the adult cornea through its support of corneal epithelial tissue repair and regeneration. As a result, the main factors which threaten the corneal clarity are inflammatory reactions, neovascularization, and limbal deficiency. In fact, the influx of inflammatory cells causes scar formation and destruction of the limbus zone. Current studies about wound healing treatment focus on corneal characteristics such as the immune response, angiogenesis, and cell signaling. In this review, studied topics related to wound healing and new approaches in cornea regeneration, which are mostly related to the criteria mentioned above, will be discussed.
Collapse
Affiliation(s)
- Mohammadmahdi Mobaraki
- Biomaterials Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Reza Abbasi
- Biomaterials Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Sajjad Omidian Vandchali
- Biomaterials Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Maryam Ghaffari
- Biomaterials Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Fathollah Moztarzadeh
- Biomaterials Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Haagdorens M, Cėpla V, Melsbach E, Koivusalo L, Skottman H, Griffith M, Valiokas R, Zakaria N, Pintelon I, Tassignon MJ. In Vitro Cultivation of Limbal Epithelial Stem Cells on Surface-Modified Crosslinked Collagen Scaffolds. Stem Cells Int 2019; 2019:7867613. [PMID: 31065280 PMCID: PMC6466865 DOI: 10.1155/2019/7867613] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To investigate the efficacy of recombinant human collagen type I (RHC I) and collagen-like peptide (CLP) hydrogels as alternative carrier substrates for the cultivation of limbal epithelial stem cells (LESC) under xeno-free culture conditions. METHODS Human LESC were cultivated on seven different collagen-derived hydrogels: (1) unmodified RHC I, (2) fibronectin-patterned RHC I, (3) carbodiimide-crosslinked CLP (CLP-12 EDC), (4) DMTMM- (4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methyl-morpholinium-) crosslinked CLP (CLP-12), (5) fibronectin-patterned CLP-12, (6) "3D limbal niche-mimicking" CLP-12, and (7) DMTMM-crosslinked CLP made from higher CLP concentration solution. Cell proliferation, cell morphology, and expression of LESC markers were analyzed. All data were compared to cultures on human amniotic membrane (HAM). RESULTS Human LESC were successfully cultivated on six out of seven hydrogel formulations, with primary cell cultures on CLP-12 EDC being deemed unsuccessful since the area of outgrowth did not meet quality standards (i.e., inconsistence in outgrowth and confluence) after 14 days of culture. Upon confluence, primary LESC showed high expression of the stem cell marker ΔNp63, proliferation marker cytokeratin (KRT) 14, adhesion markers integrin-β4 and E-cadherin, and LESC-specific extracellular matrix proteins laminin-α1, and collagen type IV. Cells showed low expression of differentiation markers KRT3 and desmoglein 3 (DSG3). Significantly higher gene expression of KRT3 was observed for cells cultured on CLP hydrogels compared to RHC I and HAM. Surface patterning of hydrogels influenced the pattern of proliferation but had no significant effect on the phenotype or genotype of cultures. Overall, the performance of RHC I and DMTMM-crosslinked CLP hydrogels was equivalent to that of HAM. CONCLUSION RHC I and DMTMM-crosslinked CLP hydrogels, irrespective of surface modification, support successful cultivation of primary human LESC using a xeno-free cultivation protocol. The regenerated epithelium maintained similar characteristics to HAM-based cultures.
Collapse
Affiliation(s)
- Michel Haagdorens
- Faculty of Medicine and Health Sciences, Department of Ophthalmology, Visual Optics and Visual Rehabilitation, University of Antwerp, Campus Drie Eiken, T building, T4-Ophthalmology, Universiteitsplein 1, 2610 Antwerp, Belgium
- Department of Ophthalmology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Antwerp, Belgium
| | - Vytautas Cėpla
- Department of Nanoengineering, Center for Physical Sciences and Technology, Savanorių 231, 02300 Vilnius, Lithuania
- Ferentis UAB, Savanorių 235, 02300 Vilnius, Lithuania
| | - Eline Melsbach
- Department of Ophthalmology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, CCRG-Oogheelkunde, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Laura Koivusalo
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33014, Finland
| | - Heli Skottman
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33014, Finland
| | - May Griffith
- Maisonneuve-Rosemont Hospital Research Centre and Department of Ophthalmology, University of Montreal, Montreal, QC, Canada H1T 4B3
| | - Ramūnas Valiokas
- Department of Nanoengineering, Center for Physical Sciences and Technology, Savanorių 231, 02300 Vilnius, Lithuania
- Ferentis UAB, Savanorių 235, 02300 Vilnius, Lithuania
| | - Nadia Zakaria
- Faculty of Medicine and Health Sciences, Department of Ophthalmology, Visual Optics and Visual Rehabilitation, University of Antwerp, Campus Drie Eiken, T building, T4-Ophthalmology, Universiteitsplein 1, 2610 Antwerp, Belgium
- Department of Ophthalmology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, CCRG-Oogheelkunde, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Antwerp University, Campus Drie Eiken, T building, T1-Veterinary Sciences, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Marie-José Tassignon
- Faculty of Medicine and Health Sciences, Department of Ophthalmology, Visual Optics and Visual Rehabilitation, University of Antwerp, Campus Drie Eiken, T building, T4-Ophthalmology, Universiteitsplein 1, 2610 Antwerp, Belgium
- Department of Ophthalmology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Antwerp, Belgium
| |
Collapse
|
19
|
Williams R, Lace R, Kennedy S, Doherty K, Levis H. Biomaterials for Regenerative Medicine Approaches for the Anterior Segment of the Eye. Adv Healthc Mater 2018; 7:e1701328. [PMID: 29388397 DOI: 10.1002/adhm.201701328] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/22/2017] [Indexed: 12/13/2022]
Abstract
The role of biomaterials in tissue engineering and regenerative medicine strategies to treat vision loss associated with damage to tissues in the anterior segment of the eye has been studied for several years. This has mostly involved replacement and support for the cornea and conjunctiva. These are complex tissues with specific functional requirements for different parts of the tissue. Amniotic membrane (AM) is used in clinical practice to transplant autologous or allogenic cells to the corneal surface. Fibrin gels have also progressed to clinical use under specific conditions. Alternatives to AM such as collagen gels, other natural materials, for example keratin and silks, and synthetic polymers have received considerable attention in laboratory and animal studies. This experience is building a body of evidence to demonstrate the potential of tissue engineering and regenerative medicine in corneal and conjunctival reconstruction and can also lead to other applications in the anterior segment of the eye, for example, the trabecular meshwork. There is a real clinical need for new procedures to overcome vision loss but there are also opportunities for developments in ocular applications to lead to biomaterials innovations for use in other clinical areas.
Collapse
Affiliation(s)
- Rachel Williams
- Department of Eye and Vision Science, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Rebecca Lace
- Department of Eye and Vision Science, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Stephnie Kennedy
- Department of Eye and Vision Science, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Kyle Doherty
- Department of Eye and Vision Science, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Hannah Levis
- Department of Eye and Vision Science, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| |
Collapse
|
20
|
Chermnykh E, Kalabusheva E, Vorotelyak E. Extracellular Matrix as a Regulator of Epidermal Stem Cell Fate. Int J Mol Sci 2018; 19:ijms19041003. [PMID: 29584689 PMCID: PMC5979429 DOI: 10.3390/ijms19041003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 12/17/2022] Open
Abstract
Epidermal stem cells reside within the specific anatomic location, called niche, which is a microenvironment that interacts with stem cells to regulate their fate. Regulation of many important processes, including maintenance of stem cell quiescence, self-renewal, and homeostasis, as well as the regulation of division and differentiation, are common functions of the stem cell niche. As it was shown in multiple studies, extracellular matrix (ECM) contributes a lot to stem cell niches in various tissues, including that of skin. In epidermis, ECM is represented, primarily, by a highly specialized ECM structure, basement membrane (BM), which separates the epidermal and dermal compartments. Epidermal stem cells contact with BM, but when they lose the contact and migrate to the overlying layers, they undergo terminal differentiation. When considering all of these factors, ECM is of fundamental importance in regulating epidermal stem cells maintenance, proper mobilization, and differentiation. Here, we summarize the remarkable progress that has recently been made in the research of ECM role in regulating epidermal stem cell fate, paying special attention to the hair follicle stem cell niche. We show that the destruction of ECM components impairs epidermal stem cell morphogenesis and homeostasis. A deep understanding of ECM molecular structure as well as the development of in vitro system for stem cell maintaining by ECM proteins may bring us to developing new approaches for regenerative medicine.
Collapse
Affiliation(s)
- Elina Chermnykh
- Koltzov Institute of Developmental Biology Russian Academy of Sciences, Moscow 119334, Russia.
- Department of Regenerative Medicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
| | - Ekaterina Kalabusheva
- Koltzov Institute of Developmental Biology Russian Academy of Sciences, Moscow 119334, Russia.
- Department of Regenerative Medicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
| | - Ekaterina Vorotelyak
- Koltzov Institute of Developmental Biology Russian Academy of Sciences, Moscow 119334, Russia.
- Department of Regenerative Medicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119991, Russia.
| |
Collapse
|
21
|
Prina E, Mistry P, Sidney LE, Yang J, Wildman RD, Bertolin M, Breda C, Ferrari B, Barbaro V, Hopkinson A, Dua HS, Ferrari S, Rose FRAJ. 3D Microfabricated Scaffolds and Microfluidic Devices for Ocular Surface Replacement: a Review. Stem Cell Rev Rep 2018; 13:430-441. [PMID: 28573367 DOI: 10.1007/s12015-017-9740-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, there has been increased research interest in generating corneal substitutes, either for use in the clinic or as in vitro corneal models. The advancement of 3D microfabrication technologies has allowed the reconstruction of the native microarchitecture that controls epithelial cell adhesion, migration and differentiation. In addition, such technology has allowed the inclusion of a dynamic fluid flow that better mimics the physiology of the native cornea. We review the latest innovative products in development in this field, from 3D microfabricated hydrogels to microfluidic devices.
Collapse
Affiliation(s)
- Elisabetta Prina
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Pritesh Mistry
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Laura E Sidney
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Jing Yang
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Ricky D Wildman
- Faculty of Engineering, University of Nottingham, Nottingham, UK
| | - Marina Bertolin
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Claudia Breda
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Barbara Ferrari
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Vanessa Barbaro
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Andrew Hopkinson
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Harminder S Dua
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Stefano Ferrari
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy.
| | - Felicity R A J Rose
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
22
|
Paterson TE, Beal SN, Santocildes-Romero ME, Sidambe AT, Hatton PV, Asencio IO. Selective laser melting-enabled electrospinning: Introducing complexity within electrospun membranes. Proc Inst Mech Eng H 2018. [PMID: 28639518 DOI: 10.1177/0954411917690182] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Additive manufacturing technologies enable the creation of very precise and well-defined structures that can mimic hierarchical features of natural tissues. In this article, we describe the development of a manufacturing technology platform to produce innovative biodegradable membranes that are enhanced with controlled microenvironments produced via a combination of selective laser melting techniques and conventional electrospinning. This work underpins the manufacture of a new generation of biomaterial devices that have significant potential for use as both basic research tools and components of therapeutic implants. The membranes were successfully manufactured and a total of three microenvironment designs (niches) were chosen for thorough characterisation. Scanning electron microscopy analysis demonstrated differences in fibre diameters within different areas of the niche structures as well as differences in fibre density. We also showed the potential of using the microfabricated membranes for supporting mesenchymal stromal cell culture and proliferation. We demonstrated that mesenchymal stromal cells grow and populate the membranes penetrating within the niche-like structures. These findings demonstrate the creation of a very versatile tool that can be used in a variety of tissue regeneration applications including bone healing.
Collapse
Affiliation(s)
- Thomas E Paterson
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | - Selina N Beal
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | - Martin E Santocildes-Romero
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | - Alfred T Sidambe
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | - Paul V Hatton
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | - Ilida Ortega Asencio
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| |
Collapse
|
23
|
Generation and characterisation of decellularised human corneal limbus. Graefes Arch Clin Exp Ophthalmol 2018; 256:547-557. [PMID: 29392398 DOI: 10.1007/s00417-018-3904-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/29/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Limbal epithelial stem cells (LESC) reside in a niche in the corneo-scleral transition zone. Deficiency leads to pain, corneal opacity, and eventually blindness. LESC transplantation of ex-vivo expanded human LESC on a carrier such as human amniotic membrane is a current treatment option. We evaluated decellularised human limbus (DHL) as a potential carrier matrix for the transplantation of LESC. METHODS Human corneas were obtained from the local eye bank. The limbal tissue was decellularised by sodium desoxychelate and DNase solution and sterilised by γ-irradiation. Native limbus- and DHL-surface structures were assessed by scanning electron microscopy and collagen ultrastructure using transmission electron microscopy. Presence and preservation of limbal basement membrane proteins in native limbus and DHL were analysed immunohistochemically. Absence of DNA after decellularisation was assessed by Feulgen staining and DNA quantification. Presence of immune cells was explored by CD45 staining, and potential cytotoxicity was tested using a cell viability assay. RESULTS In the DHL, the DNA content was reduced from 1.5 ± 0.3 μg/mg to 0.15 ± 0.01 μg/mg; the three-dimensional structure and the arrangement of the collagen fibrils were preserved. Main basement membrane proteins such as collagen IV, laminin, and fibronectin were still present after decellularisation and γ-irradiation. CD45-expressing cells were evident neither in the native limbus nor in the DHL. DHL did not convey cytotoxicity. CONCLUSIONS The extracellular matrix (ECM) of the limbus provides a tissue specific morphology and three-dimensionality consisting of particular ECM proteins. It therefore represents a substantial component of the stem cell niche. The DHL provides a specific limbal niche surrounding, and might serve as an easily producible carrier matrix for LESC transplantation.
Collapse
|
24
|
Nguyen KN, Bobba S, Richardson A, Park M, Watson SL, Wakefield D, Di Girolamo N. Native and synthetic scaffolds for limbal epithelial stem cell transplantation. Acta Biomater 2018; 65:21-35. [PMID: 29107055 DOI: 10.1016/j.actbio.2017.10.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/22/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022]
Abstract
UNLABELLED Limbal stem cell deficiency (LSCD) is a complex blinding disease of the cornea, which cannot be treated with conventional corneal transplants. Instead, a stem cell (SC) graft is required to replenish the limbal epithelial stem cell (LESC) reservoir, which is ultimately responsible for regenerating the corneal epithelium. Current therapies utilize limbal tissue biopsies that harbor LESCs as well as tissue culture expanded cells. Typically, this tissue is placed on a scaffold that supports the formation of corneal epithelial cell sheets, which are then transferred to diseased eyes. A wide range of biological and synthetic materials have been identified as carrier substrates for LESC, some of which have been used in the clinic, including amniotic membrane, fibrin, and silicon hydrogel contact lenses, each with their own advantages and limitations. This review will provide a brief background of LSCD, focusing on bio-scaffolds that have been utilized in limbal stem cell transplantation (LSCT) and materials that are being developed as potentially novel therapeutics for patients with this disease. STATEMENT OF SIGNIFICANCE The outcome of patients with corneal blindness that receive stem cell grafts to restore eye health and correct vision varies considerably and may be due to the different biological and synthetic scaffolds used to deliver these cells to the ocular surface. This review will highlight the positive attributes and limitations of the myriad of carriers developed for clinical use as well as those that are being trialled in pre-clinical models. The overall focus is on developing a standardized therapy for patients, however due to the multiple causes of corneal blindness, a personal regenerative medicine approach may be the best option.
Collapse
Affiliation(s)
- Kim N Nguyen
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Samantha Bobba
- Prince of Wales Hospital Clinical School, Sydney, Australia
| | | | - Mijeong Park
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | - Denis Wakefield
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Nick Di Girolamo
- School of Medical Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
25
|
Eberwein P, Reinhard T. [New biomaterials and alternative stem cell sources for the reconstruction of the limbal stem cell niche]. Ophthalmologe 2017; 114:318-326. [PMID: 28378048 DOI: 10.1007/s00347-017-0463-5] [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: 11/26/2022]
Abstract
Reconstruction of the limbal stem cell niche in patients with limbal stem cell insufficiency remains one of the most challenging tasks in the treatment of ocular surface diseases. Ex vivo expansion of limbal stem cells still has potential for optimization despite positive reports in centers worldwide. New biomaterials as well as alternative cell sources for the reconstruction of the limbal stem cell niche have been published in recent years. The aim of this review is to provide insight into new biomaterials and cell sources which may find their way into clinical routine in the upcoming decades.
Collapse
Affiliation(s)
- P Eberwein
- Klinik für Augenheilkunde, Uniklinikum Freiburg, Killianstr. 5, 79106, Freiburg, Deutschland.
| | - T Reinhard
- Klinik für Augenheilkunde, Uniklinikum Freiburg, Killianstr. 5, 79106, Freiburg, Deutschland
| |
Collapse
|
26
|
Ghezzi CE, Marelli B, Omenetto FG, Funderburgh JL, Kaplan DL. 3D Functional Corneal Stromal Tissue Equivalent Based on Corneal Stromal Stem Cells and Multi-Layered Silk Film Architecture. PLoS One 2017; 12:e0169504. [PMID: 28099503 PMCID: PMC5242458 DOI: 10.1371/journal.pone.0169504] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/19/2016] [Indexed: 12/13/2022] Open
Abstract
The worldwide need for human cornea equivalents continues to grow. Few clinical options are limited to allogenic and synthetic material replacements. We hypothesized that tissue engineered human cornea systems based on mechanically robust, patterned, porous, thin, optically clear silk protein films, in combination with human corneal stromal stem cells (hCSSCs), would generate 3D functional corneal stroma tissue equivalents, in comparison to previously developed 2D approaches. Silk film contact guidance was used to control the alignment and distribution of hCSSCs on RGD-treated single porous silk films, which were then stacked in an orthogonally, multi-layered architecture and cultured for 9 weeks. These systems were compared similar systems generated with human corneal fibroblasts (hCFs). Both cell types were viable and preferentially aligned along the biomaterial patterns for up to 9 weeks in culture. H&E histological sections showed that the systems seeded with the hCSSCs displayed ECM production throughout the entire thickness of the constructs. In addition, the ECM proteins tested positive for keratocyte-specific tissue markers, including keratan sulfate, lumican, and keratocan. The quantification of hCSSC gene expression of keratocyte-tissue markers, including keratocan, lumican, human aldehyde dehydrogenase 3A1 (ALDH3A1), prostaglandin D2 synthase (PTDGS), and pyruvate dehydrogenase kinase, isozyme 4 (PDK4), within the 3D tissue systems demonstrated upregulation when compared to 2D single silk films and to the systems generated with the hCFs. Furthermore, the production of ECM from the hCSSC seeded systems and subsequent remodeling of the initial matrix significantly improved cohesiveness and mechanical performance of the constructs, while maintaining transparency after 9 weeks.
Collapse
Affiliation(s)
- Chiara E. Ghezzi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Benedetto Marelli
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Fiorenzo G. Omenetto
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - James L. Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| |
Collapse
|
27
|
In Vitro Study of the Deturgescence Ability of Cultivated Human Corneal Endothelial Cells. Cornea 2016; 35:669-72. [DOI: 10.1097/ico.0000000000000792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
28
|
Shaharuddin B, Osei-Bempong C, Ahmad S, Rooney P, Ali S, Oldershaw R, Meeson A. Human limbal mesenchymal stem cells express ABCB5 and can grow on amniotic membrane. Regen Med 2016; 11:273-86. [DOI: 10.2217/rme-2016-0009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: To isolate and characterize limbal mesenchymal stem cells (LMSCs) from human corneoscleral rings. Materials & methods: Cells were isolated from corneoscleral rings and cultured in a mesenchymal stem cell (MSC)-selective media and examined for differentiation, phenotyping and characterization. Results: LMSCs were capable of trilineage differentiation, adhered to tissue culture plastic, expressed HLA class I and cell surface antigens associated with human MSC while having no/low expression of HLA class II and negative hematopoietic lineage markers. They were capable for CXCL12-mediated cellular migration. LMSCs adhered, proliferated on amniotic membrane and expressed the common putative limbal stem cell markers. Conclusion: Limbal-derived MSC exhibited plasticity, could maintain limbal markers expression and demonstrated viable growth on amniotic membrane.
Collapse
Affiliation(s)
- Bakiah Shaharuddin
- Institute of Genetic Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon-Tyne, NE1 3BZ, UK
- Advanced Medical & Dental Institute, Universiti Sains Malaysia, 13200 Pulau Pinang, Malaysia
| | - Charles Osei-Bempong
- Institute of Genetic Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon-Tyne, NE1 3BZ, UK
| | - Sajjad Ahmad
- St Paul's Eye Unit, Royal Liverpool University Hospital, Prescot Street, Liverpool, L7 8XP, UK
- Department of Eye & Vision Sciences, Institute of Ageing & Chronic Disease, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, L69 3GA, UK
| | - Paul Rooney
- Tissue Development Laboratory, NHS Blood & Transplant, Estuary Banks, Liverpool, L24 8RB, UK
| | - Simi Ali
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon-Tyne, NE2 4HH, UK
| | - Rachel Oldershaw
- Department of Musculoskeletal Biology Group I, Institute of Ageing & Chronic Disease, Faculty of Health & Life Sciences, University of Liverpool, Leahurst Campus, Chester High Road, Neston, Cheshire, CH64 7TE, UK
| | - Annette Meeson
- Institute of Genetic Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon-Tyne, NE1 3BZ, UK
| |
Collapse
|
29
|
Preservation of human limbal epithelial progenitor cells on carbodiimide cross-linked amniotic membrane via integrin-linked kinase-mediated Wnt activation. Acta Biomater 2016; 31:144-155. [PMID: 26612415 DOI: 10.1016/j.actbio.2015.11.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022]
Abstract
The Wnt pathway is a major signaling pathway that regulates corneal epithelial stem cells. However, little is known about how the ultrastructure of the limbal epithelial basement membrane (EBM) affects Wnt activity. Due to its enhanced matrix stability, the cross-linked amniotic membrane (AM) has gained increasing interest in the field of regenerative medicine. For the first time, we used EDC/NHS cross-linked denuded AM (CLDAM) as a simulated EBM substrate to investigate this mechanism. Human limbal epithelial (HLE) cells were cultured on dishes (HLE/dish), denuded AM (HLE/DAM) or CLDAM (HLE/CLDAM). Compared with HLE/dish or HLE/DAM cultures, HLE/CLDAM cultures showed greater BrdU retention and colony formation efficiency and expressed higher levels of p63, ABCG2, integrin β1, and integrin-linked kinase (ILK). Nuclear β-catenin and TCF-4 levels were higher in HLE/CLDAM cultures compared with HLE cells cultured on collagen IV, laminin, Matrigel, or DAM. Silencing of ILK in HLE/CLDAM cultures resulted in decreased levels of nuclear β-catenin, TCF-4 and deltaNp63α, whereas cytokeratin 12 expression increased. Over-expression of ILK in HLE/dish cultures had the opposite effects. Accordingly, we proposed that the CLDAM matrix, with its higher rigidity and rougher ultrastructure, better preserved HLE progenitor cells in vitro, possibly by activating integrin β1/ILK, which indirectly activated Wnt/β-catenin and subsequently deltaNp63α. Crosstalk between the integrin β1/ILK and Wnt/β-catenin pathways appears to play a crucial role in limbal progenitor cell survival on EBM. STATEMENT OF SIGNIFICANCE We demonstrated the superior capability of carbodiimide cross-linked denuded amniotic membrane (CLDAM) than natural DAM to preserve limbo-corneal epithelial progenitor cells in vitro, then we used CLDAM as a simulated epithelial basement membrane (EBM) to study how EBM maintains limbal epithelial stem cells (LESCs). We found that integrin-linked kinase (ILK) is an important mediator that transfers survival signals detected by integrin β1 to the Wnt/β-catenin pathway, which in turn up-regulates deltaNp63α, a master gene that regulates LESC function. The rougher surface of the limbal EBM suggests that the surface complexity of the LESC niche may be important in regulating LESC function, which is triggered by the recognition of topographic cues by integrin β1, followed by activation of the ILK/Wnt/β-catenin/p63 cascade.
Collapse
|
30
|
Establishment of a novel in vitro model of stratified epithelial wound healing with barrier function. Sci Rep 2016; 6:19395. [PMID: 26759072 PMCID: PMC4725353 DOI: 10.1038/srep19395] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/17/2015] [Indexed: 12/26/2022] Open
Abstract
The repair of wounds through collective movement of epithelial cells is a fundamental process in multicellular organisms. In stratified epithelia such as the cornea and skin, healing occurs in three steps that include a latent, migratory, and reconstruction phases. Several simple and inexpensive assays have been developed to study the biology of cell migration in vitro. However, these assays are mostly based on monolayer systems that fail to reproduce the differentiation processes associated to multilayered systems. Here, we describe a straightforward in vitro wound assay to evaluate the healing and restoration of barrier function in stratified human corneal epithelial cells. In this assay, circular punch injuries lead to the collective migration of the epithelium as coherent sheets. The closure of the wound was associated with the restoration of the transcellular barrier and the re-establishment of apical intercellular junctions. Altogether, this new model of wound healing provides an important research tool to study the mechanisms leading to barrier function in stratified epithelia and may facilitate the development of future therapeutic applications.
Collapse
|
31
|
Levis HJ, Daniels JT. Recreating the Human Limbal Epithelial Stem Cell Niche with Bioengineered Limbal Crypts. Curr Eye Res 2016; 41:1153-60. [PMID: 26727236 DOI: 10.3109/02713683.2015.1095932] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Creation of an in vitro model incorporating specific features that characterize a particular stem niche would allow researchers to study stem cell behavior in a more physiological environment. MATERIALS AND METHODS We have developed a tissue engineering process (RAFT) that rapidly and reliably creates bioengineered limbal crypts (BLCs) in the surface of collagen-based tissue equivalents (TEs). These BLCs mimic the three-dimensional topography of the limbal crypts (LCs), located in the limbal region of the human cornea, which are home to a population of limbal epithelial stem cells (LESCs). RESULTS Human limbal epithelial (hLE) cells occupying our BLCs expressed putative LESC markers such as ΔNp63α and Bmi1 and produced basement membrane proteins such as laminin β1 and laminin γ3; expression patterns are very similar to those seen in native LCs. Human limbal stromal cells elongate and align along the edge of native LCs and in our RAFT TEs, human limbal fibroblasts (hLFs) also appeared to exhibit this alignment and elongation behavior in response to the BLC topography. CONCLUSIONS We have demonstrated that we can maintain an immature population of hLE cells and aligned stromal cells in our BLCs to mimic some elements of the complexity of the human LESC niche.
Collapse
Affiliation(s)
- Hannah J Levis
- a Department of Ocular Biology and Therapeutics , UCL Institute of Ophthalmology , London , UK
| | - Julie T Daniels
- a Department of Ocular Biology and Therapeutics , UCL Institute of Ophthalmology , London , UK
| |
Collapse
|
32
|
Grieve K, Ghoubay D, Georgeon C, Thouvenin O, Bouheraoua N, Paques M, Borderie V. Three-dimensional structure of the mammalian limbal stem cell niche. Exp Eye Res 2015; 140:75-84. [DOI: 10.1016/j.exer.2015.08.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/24/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022]
|
33
|
Kavanagh JN, Waring EJ, Prise KM. Radiation responses of stem cells: targeted and non-targeted effects. RADIATION PROTECTION DOSIMETRY 2015; 166:110-117. [PMID: 25877536 DOI: 10.1093/rpd/ncv161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Stem cells are fundamental to the development of any tissue or organism via their ability to self-renew, which is aided by their unlimited proliferative capacity and their ability to produce fully differentiated offspring, often from multiple lineages. Stems cells are long lived and have the potential to accumulate mutations, including in response to radiation exposure. It is thought that stem cells have the potential to be induced into a cancer stem cell phenotype and that these may play an important role in resistance to radiotherapy. For radiation-induced carcinogenesis, the role of targeted and non-targeted effects is unclear with tissue or origin being important. Studies of genomic instability and bystander responses have shown consistent effects in haematopoietic models. Several models of radiation have predicted that stem cells play an important role in tumour initiation and that bystander responses could play a role in proliferation and self-renewal.
Collapse
Affiliation(s)
- J N Kavanagh
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - E J Waring
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - K M Prise
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| |
Collapse
|
34
|
Moving epithelia: Tracking the fate of mammalian limbal epithelial stem cells. Prog Retin Eye Res 2015; 48:203-25. [DOI: 10.1016/j.preteyeres.2015.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/10/2015] [Accepted: 04/16/2015] [Indexed: 12/13/2022]
|
35
|
Massie I, Kureshi AK, Schrader S, Shortt AJ, Daniels JT. Optimization of optical and mechanical properties of real architecture for 3-dimensional tissue equivalents: Towards treatment of limbal epithelial stem cell deficiency. Acta Biomater 2015; 24:241-250. [PMID: 26092352 PMCID: PMC4550494 DOI: 10.1016/j.actbio.2015.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 06/05/2015] [Accepted: 06/09/2015] [Indexed: 12/13/2022]
Abstract
Limbal epithelial stem cell (LESC) deficiency can cause blindness. Transplantation of cultured human limbal epithelial cells (hLE) on human amniotic membrane (HAM) can restore vision but clinical graft manufacture can be unreliable. We have developed a reliable and robust tissue equivalent (TE) alternative to HAM, Real Architecture for 3D Tissue (RAFT). Here, we aimed to optimize the optical and mechanical properties of RAFT TE for treatment of LESC deficiency in clinical application. The RAFT TE protocol is tunable; varying collagen concentration and volume produces differing RAFT TEs. These were compared with HAM samples taken from locations proximal and distal to the placental disc. Outcomes assessed were transparency, thickness, light transmission, tensile strength, ease of handling, degradation rates and suitability as substrate for hLE culture. Proximal HAM samples were thicker and stronger with poorer optical properties than distal HAM samples. RAFT TEs produced using higher amounts of collagen were thicker and stronger with poorer optical properties than those produced using lower amounts of collagen. The ‘optimal’ RAFT TE was thin, transparent but still handleable and was produced using 0.6 ml of 3 mg/ml collagen. Degradation rates of the ‘optimal’ RAFT TE and HAM were similar. hLE achieved confluency on ‘optimal’ RAFT TEs at comparable rates to HAM and cells expressed high levels of putative stem cell marker p63α. These findings support the use of RAFT TE for hLE transplantation towards treatment of LESC deficiency.
Collapse
|
36
|
Dziasko MA, Tuft SJ, Daniels JT. Limbal melanocytes support limbal epithelial stem cells in 2D and 3D microenvironments. Exp Eye Res 2015; 138:70-9. [PMID: 26142953 DOI: 10.1016/j.exer.2015.06.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 12/13/2022]
Abstract
Human limbal epithelial stem cells (LESCs) are essential for the maintenance of the corneal epithelium of the ocular surface. LESCs are located within limbal crypts between the palisades of Vogt in the limbus; the interface between the peripheral cornea and conjunctiva. The limbal crypts have been proposed as a LESC niche owing to their support of epithelial cells, which can form holoclone colonies in vitro. Closely associated with the limbal crypts is a concentrated population of melanocytes. The anatomical location and close proximity to putative LESC suggests that melanocytes might play a role in maintenance of these stem cells in the niche. The aim of this study was to assess the ability of human limbal melanocytes (hLM) to support the expansion of human limbal epithelial cells (LECs) in vitro as an indicator of functional cell-cell interaction. After observing that hLM co-localize with clusters of compact epithelial cells in the native limbal crypts, hLM were isolated from crypt-rich cadaveric limbal biopsies and used as feeders for the culture of LECs. Interestingly, LECs grown on mitotically active hLM were able to generate large epithelial colonies that contained small and compact cells with morphological stem cell characteristics. Immunocytochemistry revealed that LECs expanded on hLM were positive for the expression of the putative stem cell markers CK15, Bmi-1 and p63α and negative for the marker of terminal cell differentiation CK3. LECs and hLM were finally co-cultured on RAFT (real architecture for 3D tissue) collagen tissue equivalents. In 3D co-cultures, hLM promoted multi-layering of the epithelial sheet in which basal cells were maintained in an undifferentiated state. Taken together, these observations suggest melanocytes could play an important role in the maintenance of LESCs in the native human limbal stem cell niche.
Collapse
Affiliation(s)
- Marc A Dziasko
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom.
| | - Stephen J Tuft
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom.
| | - Julie T Daniels
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom.
| |
Collapse
|
37
|
Jia L, Ghezzi CE, Kaplan DL. Optimization of silk films as substrate for functional corneal epithelium growth. J Biomed Mater Res B Appl Biomater 2015; 104:431-41. [PMID: 25891207 DOI: 10.1002/jbm.b.33408] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/05/2015] [Accepted: 02/19/2015] [Indexed: 11/10/2022]
Abstract
The corneal epithelium is the first cellular barrier to protect the cornea. Thus, functional tissue engineering of the corneal epithelium is a strategy for clinical transplantation. In this study, the optimization of silk films (SFs) as substrates for functional human corneal epithelium growth was investigated with primary human corneal epithelial cells on SFs, poly-D-lysine (PDL) coated SFs, arginine-glycine-aspartic acid (RGD) modified SFs and PDL blended SFs. PDL coated SFs significantly promoted cell adhesion at early phases in comparison to the other study groups, while PDL blended SF significantly promoted cell migration in a "wound healing" model. All film modifications promoted cell proliferation and viability, and a multi-layered epithelium was achieved in 4 weeks of culture. The epithelia formed were tightly apposed and maintained an intact barrier function against rose bengal dye penetration. The results suggested that a differentiated human corneal epithelium can be established with primary corneal epithelial cells on SFs in vitro, by optimizing SF composition with PDL.
Collapse
Affiliation(s)
- Liang Jia
- Department of Biomedical Engineering, Tufts University, Medford, Massachuttes, 02155.,Department of Ophthalmology, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, Tufts University, Medford, Massachuttes, 02155
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachuttes, 02155
| |
Collapse
|
38
|
Choi HR, Byun SY, Kwon SH, Park KC. Niche interactions in epidermal stem cells. World J Stem Cells 2015; 7:495-501. [PMID: 25815134 PMCID: PMC4369506 DOI: 10.4252/wjsc.v7.i2.495] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/22/2014] [Accepted: 11/03/2014] [Indexed: 02/06/2023] Open
Abstract
Within the epidermis and dermis of the skin, cells secrete and are surrounded by the extracellular matrix (ECM), which provides structural and biochemical support. The ECM of the epidermis is the basement membrane, and collagen and other dermal components constitute the ECM of the dermis. There is significant variation in the composition of the ECM of the epidermis and dermis, which can affect “cell to cell” and “cell to ECM” interactions. These interactions, in turn, can influence biological responses, aging, and wound healing; abnormal ECM signaling likely contributes to skin diseases. Thus, strategies for manipulating cell-ECM interactions are critical for treating wounds and a variety of skin diseases. Many of these strategies focus on epidermal stem cells, which reside in a unique niche in which the ECM is the most important component; interactions between the ECM and epidermal stem cells play a major role in regulating stem cell fate. As they constitute a major portion of the ECM, it is likely that integrins and type IV collagens are important in stem cell regulation and maintenance. In this review, we highlight recent research-including our previous work-exploring the role that the ECM and its associated components play in shaping the epidermal stem cell niche.
Collapse
|
39
|
Kyle DJT, Oikonomou A, Hill E, Bayat A. Development and functional evaluation of biomimetic silicone surfaces with hierarchical micro/nano-topographical features demonstrates favourable in vitro foreign body response of breast-derived fibroblasts. Biomaterials 2015; 52:88-102. [PMID: 25818416 DOI: 10.1016/j.biomaterials.2015.02.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 01/25/2015] [Accepted: 02/01/2015] [Indexed: 12/19/2022]
Abstract
Reproducing extracellular matrix topographical cues, such as those present within acellular dermal matrix (ADM), in synthetic implant surfaces, may augment cellular responses, independent of surface chemistry. This could lead to enhanced implant integration and performance while reducing complications. In this work, the hierarchical micro and nanoscale features of ADM were accurately and reproducibly replicated in polydimethylsiloxane (PDMS), using an innovative maskless 3D grayscale fabrication process not previously reported. Human breast derived fibroblasts (n=5) were cultured on PDMS surfaces and compared to commercially available smooth and textured silicone implant surfaces, for up to one week. Cell attachment, proliferation and cytotoxicity, in addition to immunofluorescence staining, SEM imaging, qRT-PCR and cytokine array were performed. ADM PDMS surfaces promoted cell adhesion, proliferation and survival (p=<0.05), in addition to increased focal contact formation and spread fibroblast morphology when compared to commercially available implant surfaces. PCNA, vinculin and collagen 1 were up-regulated in fibroblasts on biomimetic surfaces while IL8, TNFα, TGFβ1 and HSP60 were down-regulated (p=<0.05). A reduced inflammatory cytokine response was also observed (p=<0.05). This study represents a novel approach to the development of functionalised biomimetic prosthetic implant surfaces which were demonstrated to significantly attenuate the acute in vitro foreign body reaction to silicone.
Collapse
Affiliation(s)
- Daniel J T Kyle
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK; School of Computer Science, Centre for Mesoscience and Nanotechnology, The University of Manchester, Manchester, UK
| | - Antonios Oikonomou
- School of Computer Science, Centre for Mesoscience and Nanotechnology, The University of Manchester, Manchester, UK
| | - Ernie Hill
- School of Computer Science, Centre for Mesoscience and Nanotechnology, The University of Manchester, Manchester, UK
| | - Ardeshir Bayat
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK.
| |
Collapse
|
40
|
Eberwein P, Reinhard T. Concise Reviews: The Role of Biomechanics in the Limbal Stem Cell Niche: New Insights for Our Understanding of This Structure. Stem Cells 2015; 33:916-24. [DOI: 10.1002/stem.1886] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/16/2014] [Indexed: 12/13/2022]
|
41
|
Ghezzi CE, Rnjak-Kovacina J, Kaplan DL. Corneal tissue engineering: recent advances and future perspectives. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:278-87. [PMID: 25434371 DOI: 10.1089/ten.teb.2014.0397] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To address the growing need for corneal transplants two main approaches are being pursued: allogenic and synthetic materials. Allogenic tissue from human donors is currently the preferred choice; however, there is a worldwide shortage in donated corneal tissue. In addition, tissue rejection often limits the long-term success of this approach. Alternatively, synthetic homologs to donor corneal grafts are primarily considered temporary replacements until suitable donor tissue becomes available, as they result in a high incidence of graft failure. Tissue engineered cornea analogs would provide effective cornea tissue substitutes and alternatives to address the need to reduce animal testing of commercial products. Recent progress toward these needs is reviewed here, along with future perspectives.
Collapse
Affiliation(s)
- Chiara E Ghezzi
- 1Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Jelena Rnjak-Kovacina
- 1Department of Biomedical Engineering, Tufts University, Medford, Massachusetts.,2Graduate School of Biomedical Engineering, UNSW Australia, Sydney, Australia
| | - David L Kaplan
- 1Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
42
|
Tissue Engineering the Cornea: The Evolution of RAFT. J Funct Biomater 2015; 6:50-65. [PMID: 25809689 PMCID: PMC4384100 DOI: 10.3390/jfb6010050] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/13/2015] [Indexed: 12/13/2022] Open
Abstract
Corneal blindness affects over 10 million people worldwide and current treatment strategies often involve replacement of the defective layer with healthy tissue. Due to a worldwide donor cornea shortage and the absence of suitable biological scaffolds, recent research has focused on the development of tissue engineering techniques to create alternative therapies. This review will detail how we have refined the simple engineering technique of plastic compression of collagen to a process we now call Real Architecture for 3D Tissues (RAFT). The RAFT production process has been standardised, and steps have been taken to consider Good Manufacturing Practice compliance. The evolution of this process has allowed us to create biomimetic epithelial and endothelial tissue equivalents suitable for transplantation and ideal for studying cell-cell interactions in vitro.
Collapse
|
43
|
Science and Art of Cell-Based Ocular Surface Regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 319:45-106. [DOI: 10.1016/bs.ircmb.2015.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Massie I, Dziasko M, Kureshi A, Levis HJ, Morgan L, Neale M, Sheth R, Tovell VE, Vernon AJ, Funderburgh JL, Daniels JT. Advanced imaging and tissue engineering of the human limbal epithelial stem cell niche. Methods Mol Biol 2015; 1235:179-202. [PMID: 25388395 DOI: 10.1007/978-1-4939-1785-3_15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The limbal epithelial stem cell niche provides a unique, physically protective environment in which limbal epithelial stem cells reside in close proximity with accessory cell types and their secreted factors. The use of advanced imaging techniques is described to visualize the niche in three dimensions in native human corneal tissue. In addition, a protocol is provided for the isolation and culture of three different cell types, including human limbal epithelial stem cells from the limbal niche of human donor tissue. Finally, the process of incorporating these cells within plastic compressed collagen constructs to form a tissue-engineered corneal limbus is described and how immunohistochemical techniques may be applied to characterize cell phenotype therein.
Collapse
Affiliation(s)
- Isobel Massie
- Department of Ocular Biology and Therapeutics, Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhang C, Wen J, Yan J, Kao Y, Ni Z, Cui X, Wang H. In situ growth induction of the corneal stroma cells using uniaxially aligned composite fibrous scaffolds. RSC Adv 2015. [DOI: 10.1039/c4ra16609d] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An electrospun fibrous scaffold was fabricated and used in the in situ remediation of rabbits’ corneal stromata.
Collapse
Affiliation(s)
- Cong Zhang
- College of Chemistry
- Jilin University
- Changchun 130012
- P. R. China
| | - Jihong Wen
- Department of Jilin University
- First Clinical Hospital of Bethune Medical
- Changchun 130012
- P. R. China
| | - Jing Yan
- Department of Chemistry and Biochemistry Nanocenter
- University of South Carolina
- Columbia
- USA
| | - Yanbing Kao
- Clinic College of Medicine
- Jilin University
- Changchun 130012
- P. R. China
| | - Zhiqiang Ni
- Department of Tumor Biological Therapy Jilin Province People’s Hospital
- Changchun 130012
- P. R. China
| | - Xuejun Cui
- College of Chemistry
- Jilin University
- Changchun 130012
- P. R. China
| | - Hongyan Wang
- College of Chemistry
- Jilin University
- Changchun 130012
- P. R. China
| |
Collapse
|
46
|
Massie I, Dale SB, Daniels JT. Limbal Fibroblasts Maintain Normal Phenotype in 3D RAFT Tissue Equivalents Suggesting Potential for Safe Clinical Use in Treatment of Ocular Surface Failure. Tissue Eng Part C Methods 2014; 21:576-84. [PMID: 25380529 DOI: 10.1089/ten.tec.2014.0458] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Limbal epithelial stem cell deficiency can cause blindness, but transplantation of these cells on a carrier such as human amniotic membrane can restore vision. Unfortunately, clinical graft manufacture using amnion can be inconsistent. Therefore, we have developed an alternative substrate, Real Architecture for 3D Tissue (RAFT), which supports human limbal epithelial cells (hLE) expansion. Epithelial organization is improved when human limbal fibroblasts (hLF) are incorporated into RAFT tissue equivalent (TE). However, hLF have the potential to transdifferentiate into a pro-scarring cell type, which would be incompatible with therapeutic transplantation. The aim of this work was to assess the scarring phenotype of hLF in RAFT TEs in hLE+ and hLE- RAFT TEs and in nonairlifted and airlifted RAFT TEs. Diseased fibroblasts (dFib) isolated from the fibrotic conjunctivae of ocular mucous membrane pemphigoid (Oc-MMP) patients were used as a pro-scarring positive control against which hLF were compared using surrogate scarring parameters: matrix metalloproteinase (MMP) activity, de novo collagen synthesis, α-smooth muscle actin (α-SMA) expression, and transforming growth factor-β (TGF-β) secretion. Normal hLF and dFib maintained different phenotypes in RAFT TE. MMP-2 and -9 activity, de novo collagen synthesis, and α-SMA expression were all increased in dFib cf. normal hLF RAFT TEs, although TGF-β1 secretion did not differ between normal hLF and dFib RAFT TEs. Normal hLF do not progress toward a scarring-like phenotype during culture in RAFT TEs and, therefore, may be safe to include in therapeutic RAFT TE, where they can support hLE, although in vivo work is required to confirm this. dFib RAFT TEs (used in this study as a positive control) may be useful toward the development of an ex vivo disease model of Oc-MMP.
Collapse
Affiliation(s)
- Isobel Massie
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Sarah B Dale
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Julie T Daniels
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
47
|
Lai JY, Wang PR, Luo LJ, Chen ST. Stabilization of collagen nanofibers with L-lysine improves the ability of carbodiimide cross-linked amniotic membranes to preserve limbal epithelial progenitor cells. Int J Nanomedicine 2014; 9:5117-30. [PMID: 25395849 PMCID: PMC4227622 DOI: 10.2147/ijn.s69689] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To overcome the drawbacks associated with limited cross-linking efficiency of carbodiimide modified amniotic membrane, this study investigated the use of l-lysine as an additional amino acid bridge to enhance the stability of a nanofibrous tissue matrix for a limbal epithelial cell culture platform. Results of ninhydrin assays and zeta potential measurements showed that the amount of positively charged amino acid residues incorporated into the tissue collagen chains is highly correlated with the l-lysine-pretreated concentration. The cross-linked structure and hydrophilicity of amniotic membrane scaffolding materials affected by the lysine molecular bridging effects were determined. With an increase in the l-lysine-pretreated concentration from 1 to 30 mM, the cross-linking density was significantly increased and water content was markedly decreased. The variations in resistance to thermal denaturation and enzymatic degradation were in accordance with the number of cross-links per unit mass of amniotic membrane, indicating l-lysine-modulated stabilization of collagen molecules. It was also noteworthy that the carbodiimide cross-linked tissue samples prepared using a relatively high l-lysine-pretreated concentration (ie, 30 mM) appeared to have decreased light transmittance and biocompatibility, probably due to the influence of a large nanofiber size and a high charge density. The rise in stemness gene and protein expression levels was dependent on improved cross-link formation, suggesting the crucial role of amino acid bridges in constructing suitable scaffolds to preserve limbal progenitor cells. It is concluded that mild to moderate pretreatment conditions (ie, 3–10 mM l-lysine) can provide a useful strategy to assist in the development of carbodiimide cross-linked amniotic membrane as a stable stem cell niche for corneal epithelial tissue engineering.
Collapse
Affiliation(s)
- Jui-Yang Lai
- Institute of Biochemical and Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan, Republic of China ; Biomedical Engineering Research Center, Chang Gung University, Taoyuan, Taiwan, Republic of China ; Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Pei-Ran Wang
- Institute of Biochemical and Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Li-Jyuan Luo
- Institute of Biochemical and Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Si-Tan Chen
- Institute of Biochemical and Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan, Republic of China
| |
Collapse
|
48
|
Massie I, Levis HJ, Daniels JT. Response of human limbal epithelial cells to wounding on 3D RAFT tissue equivalents: effect of airlifting and human limbal fibroblasts. Exp Eye Res 2014; 127:196-205. [PMID: 25108221 DOI: 10.1016/j.exer.2014.07.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/25/2014] [Accepted: 07/29/2014] [Indexed: 12/13/2022]
Abstract
Limbal epithelial stem cell deficiency can cause blindness but may be treated by human limbal epithelial cell (hLE) transplantation, normally on human amniotic membrane. Clinical outcomes using amnion can be unreliable and so we have developed an alternative tissue equivalent (TE), RAFT (Real Architecture for 3D Tissue), which supports hLE expansion, and stratification when airlifted. Human limbal fibroblasts (hLF) may be incorporated into RAFT TEs, where they support overlying hLE and improve phenotype. However, the impact of neither airlifting nor hLF on hLE function has been investigated. hLE on RAFT TEs (±hLF and airlifting) were wounded using heptanol and re-epithelialisation (fluorescein diacetate staining), and percentage putative stem cell marker p63α and proliferative marker Ki67 expression (wholemount immunohistochemistry), measured. Airlifted, hLF- RAFT TEs were unable to close the wound and p63α expression was 7 ± 0.2% after wounding. Conversely, non-airlifted, hLF- RAFT TEs closed the wound within 9 days and p63α expression was higher at 22 ± 5% (p < 0.01). hLE on both hLF- and hLF+ RAFT TEs (non-airlifted) closed the wound and p63α expression was 26 ± 8% and 36 ± 3% respectively (ns). Ki67 expression by hLE increased from 1.3 ± 0.5% before wounding to 7.89 ± 2.53% post-wounding for hLF- RAFT TEs (p < 0.01), and 0.8 ± 0.08% to 17.68 ± 10.88% for hLF+ RAFT TEs (p < 0.05), suggesting that re-epithelialisation was a result of proliferation. These data suggest that neither airlifting nor hLF are necessarily required to maintain a functional epithelium on RAFT TEs, thus simplifying and shortening the production process. This is important when working towards clinical application of regenerative medicine products.
Collapse
Affiliation(s)
- Isobel Massie
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Hannah J Levis
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Julie T Daniels
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
49
|
Tan NS, Alekseeva T, Brown RA. Roofed grooves: rapid layer engineering of perfusion channels in collagen tissue models. J Biomater Appl 2014; 29:605-16. [PMID: 24934499 PMCID: PMC4230962 DOI: 10.1177/0885328214538865] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Surface patterning (micro-moulding) of dense, biomimetic collagen is a simple tool to produce complex tissues using layer-by-layer assembly. The aim here was to channelise three-dimensional constructs for improved perfusion. Firstly, collagen fibril accumulation was measured by comparative image analysis to understand the mechanisms of structure formation in plastically compressed collagen during µ-moulding. This showed that shape (circular or rectangular) and dimensions of the template affected collagen distribution around moulded grooves and consequently their stability. In the second part, this was used for effective fabrication of multi-layered plastically compressed collagen constructs with internal channels by roofing the grooves with a second layer. Using rectangular templates of 25/50/100 µm widths and 75 µm depth, grooves were µ-moulded into the fluid-leaving surface of collagen layers with predictable width/depth fidelities. These grooves were then roofed by addition of a second plastically compressed collagen layer on top to produce µ-channels. Resulting µ-channels retained their dimensions and were stable over time in culture with fibroblasts and could be cell seeded with a lining layer by simple transfer of epithelial cells. The results of this study provide a valuable platform for rapid fabrication of complex collagen-based tissues in particular for provision of perfusing microchannels through the bulk material for improved core nutrient supply.
Collapse
Affiliation(s)
- Noah S Tan
- Tissue Repair & Engineering Centre, Institute of Orthopaedics, University College London, United Kingdom
| | - Tijna Alekseeva
- Tissue Repair & Engineering Centre, Institute of Orthopaedics, University College London, United Kingdom
| | - Robert A Brown
- Tissue Repair & Engineering Centre, Institute of Orthopaedics, University College London, United Kingdom
| |
Collapse
|
50
|
Rose JB, Pacelli S, Haj AJE, Dua HS, Hopkinson A, White LJ, Rose FRAJ. Gelatin-Based Materials in Ocular Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2014; 7:3106-3135. [PMID: 28788609 PMCID: PMC5453355 DOI: 10.3390/ma7043106] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/17/2014] [Accepted: 03/24/2014] [Indexed: 12/13/2022]
Abstract
Gelatin has been used for many years in pharmaceutical formulation, cell culture and tissue engineering on account of its excellent biocompatibility, ease of processing and availability at low cost. Over the last decade gelatin has been extensively evaluated for numerous ocular applications serving as cell-sheet carriers, bio-adhesives and bio-artificial grafts. These different applications naturally have diverse physical, chemical and biological requirements and this has prompted research into the modification of gelatin and its derivatives. The crosslinking of gelatin alone or in combination with natural or synthetic biopolymers has produced a variety of scaffolds that could be suitable for ocular applications. This review focuses on methods to crosslink gelatin-based materials and how the resulting materials have been applied in ocular tissue engineering. Critical discussion of recent innovations in tissue engineering and regenerative medicine will highlight future opportunities for gelatin-based materials in ophthalmology.
Collapse
Affiliation(s)
- James B Rose
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Settimio Pacelli
- Department of Drug Chemistry and Technologies, "Sapienza" University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Alicia J El Haj
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent ST4 7QB, UK.
| | - Harminder S Dua
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Andrew Hopkinson
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Lisa J White
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | | |
Collapse
|