151
|
Hodgetts SI, Edel M, Harvey AR. The State of Play with iPSCs and Spinal Cord Injury Models. J Clin Med 2015; 4:193-203. [PMID: 26237027 PMCID: PMC4470248 DOI: 10.3390/jcm4010193] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/08/2014] [Indexed: 01/10/2023] Open
Abstract
The application of induced pluripotent stem cell (iPSC) technologies in cell based strategies, for the repair of the central nervous system (with particular focus on the spinal cord), is moving towards the potential use of clinical grade donor cells. The ability of iPSCs to generate donor neuronal, glial and astrocytic phenotypes for transplantation is highlighted here, and we review recent research using iPSCs in attempts to treat spinal cord injury in various animal models. Also discussed are issues relating to the production of clinical grade iPSCs, recent advances in transdifferentiation protocols for iPSC-derived donor cell populations, concerns about tumourogenicity, and whether iPSC technologies offer any advantages over previous donor cell candidates or tissues already in use as therapeutic tools in experimental spinal cord injury studies.
Collapse
Affiliation(s)
- Stuart I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia 6009, Australia.
| | - Michael Edel
- Control of Pluripotency Laboratory, Department of Physiological Sciences I, Faculty of Medicine, University of Barcelona, Hospital Clinic, Casanova 143, Barcelona 08036, Spain.
- Faculty of Medicine, The University of Sydney Medical School, Division of Pediatrics and Child Health, Westmead Children's Hospital, Sydney 2010, Australia.
- School of Anatomy, Physiology and Human Biology, and the Harry Perkins Institute for Medical Research (CCTRM), The University of Western Australia, Western Australia 6009, Australia.
| | - Alan R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia 6009, Australia.
| |
Collapse
|
152
|
iPS Cells for Modelling and Treatment of Retinal Diseases. J Clin Med 2014; 3:1511-41. [PMID: 26237613 PMCID: PMC4470196 DOI: 10.3390/jcm3041511] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/16/2014] [Accepted: 11/18/2014] [Indexed: 01/10/2023] Open
Abstract
For many decades, we have relied on immortalised retinal cell lines, histology of enucleated human eyes, animal models, clinical observation, genetic studies and human clinical trials to learn more about the pathogenesis of retinal diseases and explore treatment options. The recent availability of patient-specific induced pluripotent stem cells (iPSC) for deriving retinal lineages has added a powerful alternative tool for discovering new disease-causing mutations, studying genotype-phenotype relationships, performing therapeutics-toxicity screening and developing personalised cell therapy. This review article provides a clinical perspective on the current and potential benefits of iPSC for managing the most common blinding diseases of the eye: inherited retinal diseases and age-related macular degeneration.
Collapse
|
153
|
Santos-Ferreira T, Postel K, Stutzki H, Kurth T, Zeck G, Ader M. Daylight Vision Repair by Cell Transplantation. Stem Cells 2014; 33:79-90. [DOI: 10.1002/stem.1824] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 08/06/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Tiago Santos-Ferreira
- CRTD/DFG-Center for Regenerative Therapies Dresden; Technische Universität Dresden; Dresden Germany
| | - Kai Postel
- CRTD/DFG-Center for Regenerative Therapies Dresden; Technische Universität Dresden; Dresden Germany
| | - Henrike Stutzki
- Natural and Medical Sciences Institute at the University of Tübingen; Reutlingen Germany
- Graduate Training Centre of Neuroscience; Tübingen Germany
| | - Thomas Kurth
- CRTD/DFG-Center for Regenerative Therapies Dresden; Technische Universität Dresden; Dresden Germany
| | - Günther Zeck
- Natural and Medical Sciences Institute at the University of Tübingen; Reutlingen Germany
| | - Marius Ader
- CRTD/DFG-Center for Regenerative Therapies Dresden; Technische Universität Dresden; Dresden Germany
| |
Collapse
|
154
|
Heidari R, Soheili ZS, Samiei S, Ahmadieh H, Davari M, Nazemroaya F, Bagheri A, Deezagi A. Alginate as a Cell Culture Substrate for Growth and Differentiation of Human Retinal Pigment Epithelial Cells. Appl Biochem Biotechnol 2014; 175:2399-412. [DOI: 10.1007/s12010-014-1431-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 11/28/2014] [Indexed: 11/30/2022]
|
155
|
Park SS, Bauer G, Abedi M, Pontow S, Panorgias A, Jonnal R, Zawadzki RJ, Werner JS, Nolta J. Intravitreal autologous bone marrow CD34+ cell therapy for ischemic and degenerative retinal disorders: preliminary phase 1 clinical trial findings. Invest Ophthalmol Vis Sci 2014; 56:81-9. [PMID: 25491299 DOI: 10.1167/iovs.14-15415] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Because human bone marrow (BM) CD34+ stem cells home into damaged tissue and may play an important role in tissue repair, this pilot clinical trial explored the safety and feasibility of intravitreal autologous CD34+ BM cells as potential therapy for ischemic or degenerative retinal conditions. METHODS This prospective study enrolled six subjects (six eyes) with irreversible vision loss from retinal vascular occlusion, hereditary or nonexudative age-related macular degeneration, or retinitis pigmentosa. CD34+ cells were isolated under Good Manufacturing Practice conditions from the mononuclear cellular fraction of the BM aspirate using a CliniMACs magnetic cell sorter. After intravitreal CD34+ cell injection, serial ophthalmic examinations, microperimetry/perimetry, fluorescein angiography, electroretinography (ERG), optical coherence tomography (OCT), and adaptive optics OCT were performed during the 6-month follow-up. RESULTS A mean of 3.4 million (range, 1-7 million) CD34+ cells were isolated and injected per eye. The therapy was well tolerated with no intraocular inflammation or hyperproliferation. Best-corrected visual acuity and full-field ERG showed no worsening after 6 months. Clinical examination also showed no worsening during follow-up except among age-related macular degeneration subjects in whom mild progression of geographic atrophy was noted in both the study eye and contralateral eye at 6-month follow-up, concurrent with some possible decline on multifocal ERG and microperimetry. Cellular in vivo imaging using adaptive optics OCT showed changes suggestive of new cellular incorporation into the macula of the hereditary macular degeneration study eye. CONCLUSIONS Intravitreal autologous BM CD34+ cell therapy appears feasible and well tolerated in eyes with ischemic or degenerative retinal conditions and merits further exploration. (ClinicalTrials.gov number, NCT01736059.).
Collapse
Affiliation(s)
- Susanna S Park
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Gerhard Bauer
- Institute for Regenerative Cures, University of California-Davis School of Medicine, Sacramento, California, United States
| | - Mehrdad Abedi
- Division of Hematology and Oncology, University of California-Davis Cancer Center, Sacramento, California, United States
| | - Suzanne Pontow
- Institute for Regenerative Cures, University of California-Davis School of Medicine, Sacramento, California, United States
| | - Athanasios Panorgias
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Ravi Jonnal
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Robert J Zawadzki
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - John S Werner
- Department of Ophthalmology and Vision Science, University of California-Davis Eye Center, Sacramento, California, United States
| | - Jan Nolta
- Institute for Regenerative Cures, University of California-Davis School of Medicine, Sacramento, California, United States
| |
Collapse
|
156
|
Sahel JA, Marazova K, Audo I. Clinical characteristics and current therapies for inherited retinal degenerations. Cold Spring Harb Perspect Med 2014; 5:a017111. [PMID: 25324231 DOI: 10.1101/cshperspect.a017111] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Inherited retinal degenerations (IRDs) encompass a large group of clinically and genetically heterogeneous diseases that affect approximately 1 in 3000 people (>2 million people worldwide) (Bessant DA, Ali RR, Bhattacharya SS. 2001. Molecular genetics and prospects for therapy of the inherited retinal dystrophies. Curr Opin Genet Dev 11: 307-316.). IRDs may be inherited as Mendelian traits or through mitochondrial DNA, and may affect the entire retina (e.g., rod-cone dystrophy, also known as retinitis pigmentosa, cone dystrophy, cone-rod dystrophy, choroideremia, Usher syndrome, and Bardet-Bidel syndrome) or be restricted to the macula (e.g., Stargardt disease, Best disease, and Sorsby fundus dystrophy), ultimately leading to blindness. IRDs are a major cause of severe vision loss, with profound impact on patients and society. Although IRDs remain untreatable today, significant progress toward therapeutic strategies for IRDs has marked the past two decades. This progress has been based on better understanding of the pathophysiological pathways of these diseases and on technological advances.
Collapse
Affiliation(s)
- José-Alain Sahel
- Institut de la Vision, Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Paris, F-75012, France INSERM, U968, Paris, F-75012, France CNRS, UMR 7210, Paris, F-75012, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, DHU ViewMaintain, INSERM-DHOS CIC 1423, Paris, F-75012, France Fondation Ophtalmologique Adolphe de Rothschild, Paris, F-75019, France Académie des Sciences-Institut de France, Paris, F-75006, France Institute of Ophthalmology-University College London, London EC1V 9EL, United Kingdom
| | - Katia Marazova
- Institut de la Vision, Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Paris, F-75012, France INSERM, U968, Paris, F-75012, France CNRS, UMR 7210, Paris, F-75012, France
| | - Isabelle Audo
- Institut de la Vision, Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Paris, F-75012, France INSERM, U968, Paris, F-75012, France CNRS, UMR 7210, Paris, F-75012, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, DHU ViewMaintain, INSERM-DHOS CIC 1423, Paris, F-75012, France Institute of Ophthalmology-University College London, London EC1V 9EL, United Kingdom
| |
Collapse
|
157
|
Differentiation of mouse iPS cells into ameloblast-like cells in cultures using medium conditioned by epithelial cell rests of Malassez and gelatin-coated dishes. Med Mol Morphol 2014; 48:138-45. [PMID: 25319805 DOI: 10.1007/s00795-014-0088-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/24/2014] [Indexed: 01/29/2023]
Abstract
Induced pluripotent stem (iPS) cells are generated from adult cells and are potentially of great value in regenerative medicine. Recently, it was shown that iPS cells can differentiate into ameloblast-like cells in cultures using feeder cells. In the present study, we sought to induce differentiation of ameloblast-like cells from iPS cells under feeder-free conditions using medium conditioned by cultured epithelial cell rests of Malassez (ERM) cells and gelatin-coated dishes. Two culture conditions were compared: co-cultures of iPS cells and ERM cells; and, culture of iPS cells in ERM cell-conditioned medium. Differentiation of ameloblast-like cells in the cultures was assessed using real-time RT-PCR assays of expression of the marker genes keratin 14, amelogenin, and ameloblastin and by immunocytochemical staining for amelogenin. We found greater evidence of ameloblast-like cell differentiation in the cultures using the conditioned medium. In the latter, the level of amelogenin expression increased daily and was significantly higher than controls on the 7th, 10th, and 14th days. Expression of ameloblastin also increased daily and was significantly higher than controls on the 14th day. The present study demonstrates that mouse iPS cells can be induced to differentiate into ameloblast-like cells in feeder-free cell cultures using ERM cell-conditioned medium and gelatin-coated dishes.
Collapse
|
158
|
Hu Y, Liang J, Cui H, Wang X, Rong H, Shao B, Cui H. Wharton's jelly mesenchymal stem cells differentiate into retinal progenitor cells. Neural Regen Res 2014; 8:1783-92. [PMID: 25206475 PMCID: PMC4145957 DOI: 10.3969/j.issn.1673-5374.2013.19.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 05/05/2013] [Indexed: 01/09/2023] Open
Abstract
Human Wharton's jelly mesenchymal stem cells were isolated from fetal umbilical cord. Cells were cultured in serum-free neural stem cell-conditioned medium or neural stem cell-conditioned medium supplemented with Dkk-1, a Wnt/β catenin pathway antagonist, and LeftyA, a Nodal signaling pathway antagonist to induce differentiation into retinal progenitor cells. Inverted microscopy showed that after induction, the spindle-shaped or fibroblast-like Wharton's jelly mesenchymal stem cells changed into bulbous cells with numerous processes. Immunofluorescent cytochemical ing and reverse-transcription PCR showed positive expression of retinal progenitor cell markers, Pax6 and Rx, as well as weakly down-regulated nestin expression. These results demonstrate that Wharton's jelly mesenchymal stem cells are capable of differentiating into retinal progenitor cells in vitro.
Collapse
Affiliation(s)
- Ying Hu
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University, Harbin 200120, Heilongjiang Province, China ; Department of Ophthalmology, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Jun Liang
- Department of Histology and Embryology, Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| | - Hongping Cui
- Department of Ophthalmology, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Xinmei Wang
- Department of Ophthalmology, Fourth Affiliated Hospital, Harbin Medical University, Harbin 200120, Heilongjiang Province, China
| | - Hua Rong
- Department of Ophthalmology, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Bin Shao
- Department of Head-Neck and Breast Tumor, Mudanjiang Tumor Hospital, Mudanjiang 157009, Heilongjiang Province, China
| | - Hao Cui
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University, Harbin 200120, Heilongjiang Province, China
| |
Collapse
|
159
|
Fang IM, Yang CH, Chiou SH, Yang CM. Induced pluripotent stem cells without c-Myc ameliorate retinal oxidative damage via paracrine effects and reduced oxidative stress in rats. J Ocul Pharmacol Ther 2014; 30:757-70. [PMID: 25121987 DOI: 10.1089/jop.2014.0020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE To investigate the efficacy and mechanisms of non-c-Myc induced pluripotent stem cell (iPSC) transplantation in a rat model of retinal oxidative damage. METHODS Paraquat was intravitreously injected into Sprague-Dawley rats. After non-c-Myc iPSC transplantation, retinal function was evaluated by electroretinograms (ERGs). The generation of reactive oxygen species (ROS) was determined by lucigenin- and luminol-enhanced chemiluminescence. The expression of brain-derived neurotrophic factor, ciliary neurotrophic factor, basic fibroblast growth factor (bFGF), stromal cell-derived factor (SDF)-1α, and CXCR4 was measured by immunohistochemistry and ELISA. An in vitro study using SH-SY5Y cells was performed to verify the protective effects of SDF-1α. RESULTS Transplantation of non-c-Myc iPSCs effectively promoted the recovery of the b-wave ratio in ERGs and significantly ameliorated retinal damage. Non-c-Myc iPSC transplantation decreased ROS production and increased the activities of superoxide dismutase and catalase, thereby reducing retinal oxidative damage and apoptotic cells. Moreover, non-c-Myc iPSC transplantation resulted in significant upregulation of SDF-1α, followed by bFGF, accompanied by a significant improvement in the ERG. In vitro studies confirmed that treatment with SDF-1α significantly reduced apoptosis in a dose-dependent manner in SH-SY5Y cells. Most transplanted cells remained in the subretinal space, with spare cells expressing neurofilament M markers at day 28. Six months after transplantation, no tumor formation was seen in animals with non-c-Myc iPSC grafts. CONCLUSIONS We demonstrated the potential benefits of non-c-Myc iPSC transplantation for treating oxidative-damage-induced retinal diseases. SDF-1α and bFGF play important roles in facilitating the amelioration of retinal oxidative damage after non-c-Myc iPSC transplantation.
Collapse
Affiliation(s)
- I-Mo Fang
- 1 Department of Ophthalmology, Taipei City Hospital Zhongxiao Branch , Taipei City, Taiwan
| | | | | | | |
Collapse
|
160
|
Cuenca N, Fernández-Sánchez L, Campello L, Maneu V, De la Villa P, Lax P, Pinilla I. Cellular responses following retinal injuries and therapeutic approaches for neurodegenerative diseases. Prog Retin Eye Res 2014; 43:17-75. [PMID: 25038518 DOI: 10.1016/j.preteyeres.2014.07.001] [Citation(s) in RCA: 316] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/03/2014] [Accepted: 07/07/2014] [Indexed: 01/17/2023]
Abstract
Retinal neurodegenerative diseases like age-related macular degeneration, glaucoma, diabetic retinopathy and retinitis pigmentosa each have a different etiology and pathogenesis. However, at the cellular and molecular level, the response to retinal injury is similar in all of them, and results in morphological and functional impairment of retinal cells. This retinal degeneration may be triggered by gene defects, increased intraocular pressure, high levels of blood glucose, other types of stress or aging, but they all frequently induce a set of cell signals that lead to well-established and similar morphological and functional changes, including controlled cell death and retinal remodeling. Interestingly, an inflammatory response, oxidative stress and activation of apoptotic pathways are common features in all these diseases. Furthermore, it is important to note the relevant role of glial cells, including astrocytes, Müller cells and microglia, because their response to injury is decisive for maintaining the health of the retina or its degeneration. Several therapeutic approaches have been developed to preserve retinal function or restore eyesight in pathological conditions. In this context, neuroprotective compounds, gene therapy, cell transplantation or artificial devices should be applied at the appropriate stage of retinal degeneration to obtain successful results. This review provides an overview of the common and distinctive features of retinal neurodegenerative diseases, including the molecular, anatomical and functional changes caused by the cellular response to damage, in order to establish appropriate treatments for these pathologies.
Collapse
Affiliation(s)
- Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain; Multidisciplinary Institute for Environmental Studies "Ramon Margalef", University of Alicante, Alicante, Spain.
| | - Laura Fernández-Sánchez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Pedro De la Villa
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Isabel Pinilla
- Department of Ophthalmology, Lozano Blesa University Hospital, Aragon Institute of Health Sciences, Zaragoza, Spain
| |
Collapse
|
161
|
Stem cell therapy for glaucoma: science or snake oil? Surv Ophthalmol 2014; 60:93-105. [PMID: 25132498 DOI: 10.1016/j.survophthal.2014.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 06/30/2014] [Accepted: 07/09/2014] [Indexed: 01/15/2023]
Abstract
In recent years there has been substantial progress in developing stem cell treatments for glaucoma. As a downstream approach that targets the underlying susceptibility of retinal ganglion and trabecular meshwork cells, stem cell therapy has the potential to both replace lost, and protect damaged, cells by secreting neurotrophic factors. A variety of sources, including embryonic cells, adult cells derived from the central nervous system, and induced pluripotent stem cells show promise as therapeutic approaches. Even though safety concerns and ethical controversies have limited clinical implementation, some institutions have already commercialized stem cell therapy and are using direct-to-consumer advertising to attract patients with glaucoma. We review the progress of stem cell therapy and its current commercial availability.
Collapse
|
162
|
Induced pluripotent stem (iPS) cells: A new source for cell-based therapeutics? J Control Release 2014; 185:37-44. [DOI: 10.1016/j.jconrel.2014.04.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 12/18/2022]
|
163
|
Gill KP, Hewitt AW, Davidson KC, Pébay A, Wong RCB. Methods of Retinal Ganglion Cell Differentiation From Pluripotent Stem Cells. Transl Vis Sci Technol 2014. [DOI: 10.1167/tvst.3.4.2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
164
|
Heller JP, Martin KR. Enhancing RPE Cell-Based Therapy Outcomes for AMD: The Role of Bruch's Membrane. Transl Vis Sci Technol 2014. [DOI: 10.1167/tvst.3.4.4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
165
|
Gill KP, Hewitt AW, Davidson KC, Pébay A, Wong RCB. Methods of Retinal Ganglion Cell Differentiation From Pluripotent Stem Cells. Transl Vis Sci Technol 2014; 3:7. [PMID: 25774327 DOI: 10.1167/tvst.3.3.7] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 04/26/2014] [Indexed: 12/22/2022] Open
Abstract
Glaucoma, the worldwide leading cause of irreversible blindness, is characterized by progressive degeneration of the optic nerve and loss of retinal ganglion cells. Research into glaucoma pathogenesis has been hampered by difficulties in isolating and culturing retinal ganglion cells in vitro. However, recent improvements in laboratory techniques have enabled the generation of a variety of mature cell types from pluripotent stem cells, including retinal ganglion cells. Indeed, stem cell-based approaches have the potential to revolutionize the field by providing an unlimited source of cells for replacement therapies and by enabling development of in vitro disease models for drug screening and research. Consequently, research aimed at directing pluripotent stem cells to differentiate into retinal ganglion cells has expanded dramatically during the past decade, resulting in significant advances in technique and efficiency. In this paper, we review the methodology for retinal ganglion cell differentiation from pluripotent stem cells of both mouse and human origin and summarize how these techniques have opened up new avenues for modelling glaucoma. Generation of stem cell-derived retinal ganglion cells will have significant translational values, providing an in vitro platform to study the mechanisms responsible for pathogenesis and for drug screening to improve treatment options, as well as for the development of cell therapies for optic neuropathies such as glaucoma.
Collapse
Affiliation(s)
- Katherine P Gill
- Department of Ophthalmology, University of Melbourne, Melbourne East, VIC, Australia
| | - Alex W Hewitt
- Department of Ophthalmology, University of Melbourne, Melbourne East, VIC, Australia
| | - Kathryn C Davidson
- Department of Ophthalmology, University of Melbourne, Melbourne East, VIC, Australia ; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital Melbourne East, VIC, Australia
| | - Alice Pébay
- Department of Ophthalmology, University of Melbourne, Melbourne East, VIC, Australia ; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital Melbourne East, VIC, Australia
| | - Raymond C B Wong
- Department of Ophthalmology, University of Melbourne, Melbourne East, VIC, Australia ; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital Melbourne East, VIC, Australia
| |
Collapse
|
166
|
Photoreceptor replacement therapy: Challenges presented by the diseased recipient retinal environment. Vis Neurosci 2014; 31:333-44. [DOI: 10.1017/s0952523814000200] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractVision loss caused by the death of photoreceptors is the leading cause of irreversible blindness in the developed world. Rapid advances in stem cell biology and techniques in cell transplantation have made photoreceptor replacement by transplantation a very plausible therapeutic strategy. These advances include the demonstration of restoration of vision following photoreceptor transplantation and the generation of transplantable populations of donor cells from stem cells. In this review, we present a brief overview of the recent progress in photoreceptor transplantation. We then consider in more detail some of the challenges presented by the degenerating retinal environment that must play host to these transplanted cells, how these may influence transplanted photoreceptor cell integration and survival, and some of the progress in developing strategies to circumnavigate these issues.
Collapse
|
167
|
Bertolotti E, Neri A, Camparini M, Macaluso C, Marigo V. Stem cells as source for retinal pigment epithelium transplantation. Prog Retin Eye Res 2014; 42:130-44. [PMID: 24933042 DOI: 10.1016/j.preteyeres.2014.06.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/30/2014] [Accepted: 06/05/2014] [Indexed: 12/27/2022]
Abstract
Inherited maculopathies, age related macular degeneration and some forms of retinitis pigmentosa are associated with impaired function or loss of the retinal pigment epithelium (RPE). Among potential treatments, transplantation approaches are particularly promising. The arrangement of RPE cells in a well-defined tissue layer makes the RPE amenable to cell or tissue sheet transplantation. Different cell sources have been suggested for RPE transplantation but the development of a clinical protocol faces several obstacles. The source should provide a sufficient number of cells to at least recover the macula area. Secondly, cells should be plastic enough to be able to integrate in the host tissue. Tissue sheets should be considered as well, but the substrate on which RPE cells are cultured needs to be carefully evaluated. Immunogenicity can also be an obstacle for effective transplantation as well as tumorigenicity of not fully differentiated cells. Finally, ethical concerns may represent drawbacks when embryo-derived cells are proposed for RPE transplantation. Here we discuss different cell sources that became available in recent years and their different properties. We also present data on a new source of human RPE. We provide a protocol for RPE differentiation of retinal stem cells derived from adult ciliary bodies of post-mortem donors. We show molecular characterization of the in vitro differentiated RPE tissue and demonstrate its functionality based on a phagocytosis assay. This new source may provide tissue for allogenic transplantation based on best matches through histocompatibility testing.
Collapse
Affiliation(s)
- Evelina Bertolotti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto Neri
- Ophthalmology, S.Bi.Bi.T. Department, University of Parma, Parma, Italy
| | - Monica Camparini
- Ophthalmology, S.Bi.Bi.T. Department, University of Parma, Parma, Italy
| | - Claudio Macaluso
- Ophthalmology, S.Bi.Bi.T. Department, University of Parma, Parma, Italy
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
168
|
Generation of three-dimensional retinal tissue with functional photoreceptors from human iPSCs. Nat Commun 2014; 5:4047. [PMID: 24915161 PMCID: PMC4370190 DOI: 10.1038/ncomms5047] [Citation(s) in RCA: 691] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 05/05/2014] [Indexed: 12/17/2022] Open
Abstract
Many forms of blindness result from the dysfunction or loss of retinal photoreceptors. Induced pluripotent stem cells (iPSCs) hold great potential for the modelling of these diseases or as potential therapeutic agents. However, to fulfill this promise, a remaining challenge is to induce human iPSC to recreate in vitro key structural and functional features of the native retina, in particular the presence of photoreceptors with outer-segment discs and light sensitivity. Here we report that hiPSC can, in a highly autonomous manner, recapitulate spatiotemporally each of the main steps of retinal development observed in vivo and form three-dimensional retinal cups that contain all major retinal cell types arranged in their proper layers. Moreover, the photoreceptors in our hiPSC-derived retinal tissue achieve advanced maturation, showing the beginning of outer-segment disc formation and photosensitivity. This success brings us one step closer to the anticipated use of hiPSC for disease modelling and open possibilities for future therapies.
Collapse
|
169
|
Heller JP, Martin KR. Enhancing RPE Cell-Based Therapy Outcomes for AMD: The Role of Bruch's Membrane. Transl Vis Sci Technol 2014; 3:11. [PMID: 25068093 PMCID: PMC4108298 DOI: 10.1167/tvst.3.3.11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 02/09/2014] [Indexed: 12/22/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of legal blindness in older people in the developed world. The disease involves damage to the part of the retina responsible for central vision. Degeneration of retinal pigment epithelial (RPE) cells, photoreceptors, and choriocapillaris may contribute to visual loss. Over the past decades, scientists and clinicians have tried to replace lost RPE cells in patients with AMD using cells from different sources. In recent years, advances in generating RPE cells from stem cells have been made and clinical trials are currently evaluating the safety and efficiency of replacing the degenerated RPE cell layer with stem cell-derived RPE cells. However, the therapeutic success of transplantation of stem cell-derived RPE cells may be limited unless the transplanted cells can adhere and survive in the long term in the diseased eye. One hallmark of AMD is the altered extracellular environment of Bruch's membrane to which the grafted cells have to adhere. Here, we discuss recent approaches to overcome the inhibitory environment of the diseased eye and to enhance the survival rate of transplanted RPE cells. Our aim is to highlight novel approaches that may have the potential to improve the efficacy of RPE transplantation for AMD in the future.
Collapse
Affiliation(s)
- Janosch P. Heller
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, United Kingdom
| | - Keith R. Martin
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
- Department of Ophthalmology, NIHR Biomedical Research Centre and Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, United Kingdom
| |
Collapse
|
170
|
Wright LS, Phillips MJ, Pinilla I, Hei D, Gamm DM. Induced pluripotent stem cells as custom therapeutics for retinal repair: progress and rationale. Exp Eye Res 2014; 123:161-72. [PMID: 24534198 PMCID: PMC4047146 DOI: 10.1016/j.exer.2013.12.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/25/2013] [Accepted: 12/02/2013] [Indexed: 12/17/2022]
Abstract
Human pluripotent stem cells have made a remarkable impact on science, technology and medicine by providing a potentially unlimited source of human cells for basic research and clinical applications. In recent years, knowledge gained from the study of human embryonic stem cells and mammalian somatic cell reprogramming has led to the routine production of human induced pluripotent stem cells (hiPSCs) in laboratories worldwide. hiPSCs show promise for use in transplantation, high throughput drug screening, "disease-in-a-dish" modeling, disease gene discovery, and gene therapy testing. This review will focus on the first application, beginning with a discussion of methods for producing retinal lineage cells that are lost in inherited and acquired forms of retinal degenerative disease. The selection of appropriate hiPSC-derived donor cell type(s) for transplantation will be discussed, as will the caveats and prerequisite steps to formulating a clinical Good Manufacturing Practice (cGMP) product for clinical trials.
Collapse
Affiliation(s)
- Lynda S Wright
- Waisman Center, University of Wisconsin, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
| | - M Joseph Phillips
- Waisman Center, University of Wisconsin, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
| | - Isabel Pinilla
- Department of Ophthalmology, Lozano Blesa Hospital and Aragones Health Sciences Institute, Zaragoza, Spain
| | - Derek Hei
- Waisman Center, University of Wisconsin, Madison, WI, USA
| | - David M Gamm
- Waisman Center, University of Wisconsin, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
171
|
Derivation of traceable and transplantable photoreceptors from mouse embryonic stem cells. Stem Cell Reports 2014; 2:853-65. [PMID: 24936471 PMCID: PMC4050344 DOI: 10.1016/j.stemcr.2014.04.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 04/15/2014] [Accepted: 04/16/2014] [Indexed: 01/12/2023] Open
Abstract
Retinal degenerative diseases resulting in the loss of photoreceptors are one of the major causes of blindness. Photoreceptor replacement therapy is a promising treatment because the transplantation of retina-derived photoreceptors can be applied now to different murine retinopathies to restore visual function. To have an unlimited source of photoreceptors, we derived a transgenic embryonic stem cell (ESC) line in which the Crx-GFP transgene is expressed in photoreceptors and assessed the capacity of a 3D culture protocol to produce integration-competent photoreceptors. This culture system allows the production of a large number of photoreceptors recapitulating the in vivo development. After transplantation, integrated cells showed the typical morphology of mature rods bearing external segments and ribbon synapses. We conclude that a 3D protocol coupled with ESCs provides a safe and renewable source of photoreceptors displaying a development and transplantation competence comparable to photoreceptors from age-matched retinas. De novo isolation of Crx-GFP embryonic stem cell lines to trace photoreceptors 3D culture system fine-tuning to generate many integration-competent photoreceptors Revealing in-vitro- and in-vivo-developing retina similarities Characterization of the most appropriate stage to transplant photoreceptors
Collapse
|
172
|
Abstract
Blindness represents an increasing global problem with significant social and economic impact upon affected patients and society as a whole. In Europe, approximately one in 30 individuals experience sight loss and 75% of those are unemployed, a social burden which is very likely to increase as the population of Europe ages. Diseases affecting the retina account for approximately 26% of blindness globally and 70% of blindness in the United Kingdom. To date, there are no treatments to restore lost retinal cells and improve visual function, highlighting an urgent need for new therapeutic approaches. A pioneering breakthrough has demonstrated the ability to generate synthetic retina from pluripotent stem cells under laboratory conditions, a finding with immense relevance for basic research, in vitro disease modeling, drug discovery, and cell replacement therapies. This review summarizes the current achievements in pluripotent stem cell differentiation toward retinal cells and highlights the steps that need to be completed in order to generate human synthetic retinae with high efficiency and reproducibly from patient-specific pluripotent stem cells.
Collapse
|
173
|
Garita-Hernández M, Diaz-Corrales F, Lukovic D, González-Guede I, Diez-Lloret A, Valdés-Sánchez ML, Massalini S, Erceg S, Bhattacharya SS. Hypoxia increases the yield of photoreceptors differentiating from mouse embryonic stem cells and improves the modeling of retinogenesis in vitro. Stem Cells 2014; 31:966-78. [PMID: 23362204 DOI: 10.1002/stem.1339] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 11/23/2012] [Indexed: 12/19/2022]
Abstract
Retinitis pigmentosa (RP), a genetically heterogeneous group of diseases together with age-related macular degeneration (AMD), are the leading causes of permanent blindness and are characterized by the progressive dysfunction and death of the light sensing photoreceptors of the retina. Due to the limited regeneration capacity of the mammalian retina, the scientific community has invested significantly in trying to obtain retinal progenitor cells from embryonic stem cells (ESC). These represent an unlimited source of retinal cells, but it has not yet been possible to achieve specific populations, such as photoreceptors, efficiently enough to allow them to be used safely in the future as cell therapy of RP or AMD. In this study, we generated a high yield of photoreceptors from directed differentiation of mouse ESC (mESC) by recapitulating crucial phases of retinal development. We present a new protocol of differentiation, involving hypoxia and taking into account extrinsic and intrinsic cues. These include niche-specific conditions as well as the manipulation of the signaling pathways involved in retinal development. Our results show that hypoxia promotes and improves the differentiation of mESC toward photoreceptors. Different populations of retinal cells are increased in number under the hypoxic conditions applied, such as Crx-positive cells, S-Opsin-positive cells, and double positive cells for Rhodopsin and Recoverin, as shown by immunofluorescence analysis. For the first time, this manuscript reports the high efficiency of differentiation in vivo and the expression of mature rod photoreceptor markers in a large number of differentiated cells, transplanted in the subretinal space of wild-type mice.
Collapse
Affiliation(s)
- Marcela Garita-Hernández
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Assawachananont J, Mandai M, Okamoto S, Yamada C, Eiraku M, Yonemura S, Sasai Y, Takahashi M. Transplantation of embryonic and induced pluripotent stem cell-derived 3D retinal sheets into retinal degenerative mice. Stem Cell Reports 2014; 2:662-74. [PMID: 24936453 PMCID: PMC4050483 DOI: 10.1016/j.stemcr.2014.03.011] [Citation(s) in RCA: 245] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 03/27/2014] [Accepted: 03/27/2014] [Indexed: 02/09/2023] Open
Abstract
In this article, we show that mouse embryonic stem cell- or induced pluripotent stem cell-derived 3D retinal tissue developed a structured outer nuclear layer (ONL) with complete inner and outer segments even in an advanced retinal degeneration model (rd1) that lacked ONL. We also observed host-graft synaptic connections by immunohistochemistry. This study provides a "proof of concept" for retinal sheet transplantation therapy for advanced retinal degenerative diseases.
Collapse
Affiliation(s)
- Juthaporn Assawachananont
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan ; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Satoshi Okamoto
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan ; Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Chikako Yamada
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Mototsugu Eiraku
- Organogenesis and Neurogenesis Group, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Shigenobu Yonemura
- Electron Microscope Laboratory, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Yoshiki Sasai
- Organogenesis and Neurogenesis Group, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| |
Collapse
|
175
|
Homma K, Okamoto S, Mandai M, Gotoh N, Rajasimha HK, Chang YS, Chen S, Li W, Cogliati T, Swaroop A, Takahashi M. Developing rods transplanted into the degenerating retina of Crx-knockout mice exhibit neural activity similar to native photoreceptors. Stem Cells 2014; 31:1149-59. [PMID: 23495178 DOI: 10.1002/stem.1372] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 01/15/2013] [Accepted: 01/29/2013] [Indexed: 12/21/2022]
Abstract
Replacement of dysfunctional or dying photoreceptors offers a promising approach for retinal neurodegenerative diseases, including age-related macular degeneration and retinitis pigmentosa. Several studies have demonstrated the integration and differentiation of developing rod photoreceptors when transplanted in wild-type or degenerating retina; however, the physiology and function of the donor cells are not adequately defined. Here, we describe the physiological properties of developing rod photoreceptors that are tagged with green fluorescent protein (GFP) driven by the promoter of rod differentiation factor, Nrl. GFP-tagged developing rods show Ca(2 +) responses and rectifier outward currents that are smaller than those observed in fully developed photoreceptors, suggesting their immature developmental state. These immature rods also exhibit hyperpolarization-activated current (Ih ) induced by the activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. When transplanted into the subretinal space of wild-type or retinal degeneration mice, GFP-tagged developing rods can integrate into the photoreceptor outer nuclear layer in wild-type mouse retina and exhibit Ca(2 +) responses and membrane current comparable to native rod photoreceptors. A proportion of grafted rods develop rhodopsin-positive outer segment-like structures within 2 weeks after transplantation into the retina of Crx-knockout mice and produce rectifier outward current and Ih upon membrane depolarization and hyperpolarization. GFP-positive rods derived from induced pluripotent stem (iPS) cells also display similar membrane current Ih as native developing rod photoreceptors, express rod-specific phototransduction genes, and HCN-1 channels. We conclude that Nrl-promoter-driven GFP-tagged donor photoreceptors exhibit physiological characteristics of rods and that iPS cell-derived rods in vitro may provide a renewable source for cell-replacement therapy.
Collapse
Affiliation(s)
- Kohei Homma
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Human Pluripotent Stem Cell-Derived Retinal Pigmented Epithelium in Retinal Treatment: from Bench to Bedside. Mol Neurobiol 2014; 50:597-612. [DOI: 10.1007/s12035-014-8684-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/17/2014] [Indexed: 01/23/2023]
|
177
|
Al-Shamekh S, Goldberg JL. Retinal repair with induced pluripotent stem cells. Transl Res 2014; 163:377-86. [PMID: 24291154 PMCID: PMC4073787 DOI: 10.1016/j.trsl.2013.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/03/2013] [Accepted: 11/04/2013] [Indexed: 12/28/2022]
Abstract
Retinal degeneration such as age-related macular degeneration and other inherited forms, such as Stargardt's disease and retinitis pigmentosa, and optic neuropathies including glaucoma and ischemic optic neuropathy are major causes of vision loss and blindness worldwide. Damage to retinal pigment epithelial cells and photoreceptors in the former, and to retinal ganglion cell axons in the optic nerve and their cell bodies in the retina in the latter diseases lead to the eventual death of these retinal cells, and in humans there is no endogenous replacement or repair. Cell replacement therapies provide 1 avenue to restore function in these diseases, particularly in the case of retinal repair, although there are considerable issues to overcome, including the differentiation and integration of the transplanted cells. What stem cell sources could be used for such therapies? One promising source is induced pluripotent stem cells (iPSCs), which could be drawn from an individual patient needing therapy, or generated and banked from select donors. We review developing research in the use of iPSCs for retinal cell replacement therapy.
Collapse
Affiliation(s)
- Shomoukh Al-Shamekh
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Fla; Department of Ophthalmology, King Abdulaziz University Hospital, King Saud University, Riyadh, Saudi Arabia
| | - Jeffrey L Goldberg
- Shiley Eye Center, University of California, San Diego, Calif; Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Fla.
| |
Collapse
|
178
|
Srikanth P, Young-Pearse TL. Stem cells on the brain: modeling neurodevelopmental and neurodegenerative diseases using human induced pluripotent stem cells. J Neurogenet 2014; 28:5-29. [PMID: 24628482 PMCID: PMC4285381 DOI: 10.3109/01677063.2014.881358] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seven years have passed since the initial report of the generation of induced pluripotent stem cells (iPSCs) from adult human somatic cells, and in the intervening time the field of neuroscience has developed numerous disease models using this technology. Here, we review progress in the field and describe both the advantages and potential pitfalls of modeling neurodegenerative and neurodevelopmental diseases using this technology. We include tables with information on neural differentiation protocols and studies that developed human iPSC lines to model neurological diseases. We also discuss how one can: investigate effects of genetic mutations with iPSCs, examine cell fate-specific phenotypes, best determine the specificity of a phenotype, and bring in vivo relevance to this in vitro technique.
Collapse
Affiliation(s)
- Priya Srikanth
- Center for Neurologic Diseases, Brigham and Women's Hospital , Boston, Massachusetts , USA
| | | |
Collapse
|
179
|
Krohne TU, Westenskow PD, Kurihara T, Friedlander DF, Lehmann M, Dorsey AL, Li W, Zhu S, Schultz A, Wang J, Siuzdak G, Ding S, Friedlander M. Generation of retinal pigment epithelial cells from small molecules and OCT4 reprogrammed human induced pluripotent stem cells. Stem Cells Transl Med 2014; 1:96-109. [PMID: 22532929 DOI: 10.5966/sctm.2011-0057] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Autologous retinal pigment epithelium (RPE) grafts derived from induced pluripotent stem cells (iPSCs) may be used to cure blinding diseases in which RPE dysfunction results in photoreceptor degeneration. Four-, two-, and one-factor-derived iPSCs (4F-, 2F-, and 1F-iPSCs, respectively) were differentiated into fully functional cuboidal pigmented cells in polarized monolayers that express RPE-specific markers. 1F-iPSCs-RPE (1F-iPS-RPE) strongly resembles primary human fetal RPE (hfRPE) based on proteomic and untargeted metabolomic analyses, and using novel in vivo imaging technology coupled with electroretinography, we demonstrated that 1F-iPS-RPE mediate anatomical and functional rescue of photoreceptors after transplantation in an animal model of RPE-mediated retinal degeneration. 1F-iPS0RPE cells were injected subretinally as a suspension and formed a monolayer dispersed between host RPE cells. Furthermore, 1F-iPS-RPE do not simply provide trophic support to rescue photoreceptors as previously speculated but actually phagocytose photoreceptor outer segments in vivo and maintain visual cycling. Thus, 1f-iPS-RPE grafts may be superior to conventional iPS-RPE for clinical use because 1F-IPS-RPE closely resemble hfRPE, mediate anatomical and functional photoreceptor rescue in vivo, and are generated using a reduced number of potentially oncogenic reprogramming factors.
Collapse
Affiliation(s)
- Tim U Krohne
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Characterization of human induced pluripotent stem cell-derived retinal pigment epithelium cell sheets aiming for clinical application. Stem Cell Reports 2014; 2:205-18. [PMID: 24527394 PMCID: PMC3923225 DOI: 10.1016/j.stemcr.2013.12.007] [Citation(s) in RCA: 434] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/11/2013] [Accepted: 12/11/2013] [Indexed: 02/06/2023] Open
Abstract
Age-related macular degeneration (AMD) causes severe visual impairment due in part to age-dependent impairment of retinal pigment epithelium (RPE). It has been suggested that autologous human induced pluripotent stem cells (hiPSCs) may represent a useful cell source for the generation of graft RPE. We generated hiPSC-derived RPE (hiPSC-RPE) cell sheets optimized to meet clinical use requirements, including quality, quantity, consistency, and safety. These cell sheets are generated as a monolayer of cells without any artificial scaffolds, express typical RPE markers, form tight junctions that exhibit polarized secretion of growth factors, and show phagocytotic ability and gene-expression patterns similar to those of native RPE. Additionally, upon transplantation, autologous nonhuman primate iPSC-RPE cell sheets showed no immune rejection or tumor formation. These results suggest that autologous hiPSC-RPE cell sheets may serve as a useful form of graft for use in tissue replacement therapy for AMD. hiPSC-RPE cell sheets show characteristics similar to those of native RPE A hiPSC-RPE sheet is sufficient to transplant a patient All hiPSC-RPEs show gene-expression patterns similar to native RPE Autologous nonhuman primate iPSC-RPE cell sheets show no rejection or tumor
Collapse
|
181
|
Sorkio A, Hongisto H, Kaarniranta K, Uusitalo H, Juuti-Uusitalo K, Skottman H. Structure and barrier properties of human embryonic stem cell-derived retinal pigment epithelial cells are affected by extracellular matrix protein coating. Tissue Eng Part A 2014; 20:622-34. [PMID: 24044751 DOI: 10.1089/ten.tea.2013.0049] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Extracellular matrix (ECM) interactions play a vital role in cell morphology, migration, proliferation, and differentiation of cells. We investigated the role of ECM proteins on the structure and function of human embryonic stem cell-derived retinal pigment epithelial (hESC-RPE) cells during their differentiation and maturation from hESCs into RPE cells in adherent differentiation cultures on several human ECM proteins found in native human Bruch's membrane, namely, collagen I, collagen IV, laminin, fibronectin, and vitronectin, as well as on commercial substrates of xeno-free CELLstart™ and Matrigel™. Cell pigmentation, expression of RPE-specific proteins, fine structure, as well as the production of basal lamina by hESC-RPE on different protein coatings were evaluated after 140 days of differentiation. The integrity of hESC-RPE epithelium and barrier properties on different coatings were investigated by measuring transepithelial resistance. All coatings supported the differentiation of hESC-RPE cells as demonstrated by early onset of cell pigmentation and further maturation to RPE monolayers after enrichment. Mature RPE phenotype was verified by RPE-specific gene and protein expression, correct epithelial polarization, and phagocytic activity. Significant differences were found in the degree of RPE cell pigmentation and tightness of epithelial barrier between different coatings. Further, the thickness of self-assembled basal lamina and secretion of the key ECM proteins found in the basement membrane of the native RPE varied between hESC-RPE cultured on compared protein coatings. In conclusion, this study shows that the cell culture substrate has a major effect on the structure and basal lamina production during the differentiation and maturation of hESC-RPE potentially influencing the success of cell integrations and survival after cell transplantation.
Collapse
Affiliation(s)
- Anni Sorkio
- 1 Institute of Biomedical Technology, University of Tampere , Tampere, Finland
| | | | | | | | | | | |
Collapse
|
182
|
Layer PG, Araki M, Vogel-Höpker A. New concepts for reconstruction of retinal and pigment epithelial tissues. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/eop.10.42] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
183
|
Kador KE, Goldberg JL. Scaffolds and stem cells: delivery of cell transplants for retinal degenerations. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 7:459-470. [PMID: 23585772 DOI: 10.1586/eop.12.56] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Retinal degenerations and optic neuropathies often lead to death of photoreceptors or retinal ganglion cells, respectively. Stem cell therapies are showing promise for these diseases in preclinical models and are beginning to transition into human trials, but cell delivery and integration remain major challenges. Focusing on photoreceptor- and progenitor-directed approaches, in this article, the authors review how advances in tissue engineering and cell scaffold design are enhancing cell therapies for retinal degeneration.
Collapse
Affiliation(s)
- Karl E Kador
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, 1501 NW 10th Avenue, BRB 826, FL 33136, USA
| | | |
Collapse
|
184
|
Muñiz A, Greene WA, Plamper ML, Choi JH, Johnson AJ, Tsin AT, Wang HC. Retinoid uptake, processing, and secretion in human iPS-RPE support the visual cycle. Invest Ophthalmol Vis Sci 2014; 55:198-209. [PMID: 24255038 DOI: 10.1167/iovs.13-11740] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Retinal pigmented epithelium derived from human induced pluripotent stem (iPS) cells (iPS-RPE) may be a source of cells for transplantation. For this reason, it is essential to determine the functional competence of iPS-RPE. One key role of the RPE is uptake and processing of retinoids via the visual cycle. The purpose of this study is to investigate the expression of visual cycle proteins and the functional ability of the visual cycle in iPS-RPE. METHODS iPS-RPE was derived from human iPS cells. Immunocytochemistry, RT-PCR, and Western blot analysis were used to detect expression of RPE genes lecithin-retinol acyl transferase (LRAT), RPE65, cellular retinaldehyde-binding protein (CRALBP), and pigment epithelium-derived factor (PEDF). All-trans retinol was delivered to cultured cells or whole cell homogenate to assess the ability of the iPS-RPE to process retinoids. RESULTS Cultured iPS-RPE expresses visual cycle genes LRAT, CRALBP, and RPE65. After incubation with all-trans retinol, iPS-RPE synthesized up to 2942 ± 551 pmol/mg protein all-trans retinyl esters. Inhibition of LRAT with N-ethylmaleimide (NEM) prevented retinyl ester synthesis. Significantly, after incubation with all-trans retinol, iPS-RPE released 188 ± 88 pmol/mg protein 11-cis retinaldehyde into the culture media. CONCLUSIONS iPS-RPE develops classic RPE characteristics and maintains expression of visual cycle proteins. The results of this study confirm that iPS-RPE possesses the machinery to process retinoids for support of visual pigment regeneration. Inhibition of all-trans retinyl ester accumulation by NEM confirms LRAT is active in iPS-RPE. Finally, the detection of 11-cis retinaldehyde in the culture medium demonstrates the cells' ability to process retinoids through the visual cycle. This study demonstrates expression of key visual cycle machinery and complete visual cycle activity in iPS-RPE.
Collapse
Affiliation(s)
- Alberto Muñiz
- Ocular Trauma, U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, Houston, Texas
| | | | | | | | | | | | | |
Collapse
|
185
|
Pearson RA. Advances in repairing the degenerate retina by rod photoreceptor transplantation. Biotechnol Adv 2014; 32:485-91. [PMID: 24412415 PMCID: PMC4070022 DOI: 10.1016/j.biotechadv.2014.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 09/26/2013] [Accepted: 01/01/2014] [Indexed: 02/01/2023]
Abstract
Despite very different aetiologies, age-related macular degeneration (AMD) and most inherited retinal disorders culminate in the same final common pathway, loss of the light-sensitive photoreceptors. There are few clinical treatments and none can reverse the loss of vision. Photoreceptor replacement by transplantation is proposed as a broad treatment strategy applicable to all degenerations. The past decade has seen a number of landmark achievements in this field, which together provide strong justification for continuing investigation into photoreceptor replacement strategies. These include proof of principle for restoring vision by rod-photoreceptor transplantation in mice with congenital stationary night blindness and advances in stem cell biology, which have led to the generation of complete optic structures in vitro from embryonic stem cells. The latter represents enormous potential for generating suitable and renewable donor cells with which to achieve the former. However, there are still challenges presented by the degenerating recipient retinal environment that must be addressed as we move to translating these technologies towards clinical application.
Collapse
Affiliation(s)
- Rachael A Pearson
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
186
|
Stanzel BV, Liu Z, Somboonthanakij S, Wongsawad W, Brinken R, Eter N, Corneo B, Holz FG, Temple S, Stern JH, Blenkinsop TA. Human RPE stem cells grown into polarized RPE monolayers on a polyester matrix are maintained after grafting into rabbit subretinal space. Stem Cell Reports 2014; 2:64-77. [PMID: 24511471 PMCID: PMC3916756 DOI: 10.1016/j.stemcr.2013.11.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 11/07/2013] [Accepted: 11/08/2013] [Indexed: 12/22/2022] Open
Abstract
Transplantation of the retinal pigment epithelium (RPE) is being developed as a cell-replacement therapy for age-related macular degeneration. Human embryonic stem cell (hESC) and induced pluripotent stem cell (iPSC)-derived RPE are currently translating toward clinic. We introduce the adult human RPE stem cell (hRPESC) as an alternative RPE source. Polarized monolayers of adult hRPESC-derived RPE grown on polyester (PET) membranes had near-native characteristics. Trephined pieces of RPE monolayers on PET were transplanted subretinally in the rabbit, a large-eyed animal model. After 4 days, retinal edema was observed above the implant, detected by spectral domain optical coherence tomography (SD-OCT) and fundoscopy. At 1 week, retinal atrophy overlying the fetal or adult transplant was observed, remaining stable thereafter. Histology obtained 4 weeks after implantation confirmed a continuous polarized human RPE monolayer on PET. Taken together, the xeno-RPE survived with retained characteristics in the subretinal space. These experiments support that adult hRPESC-derived RPE are a potential source for transplantation therapies. Adult hRPESC-derived RPE had comparable in vitro characteristics to fetal hRPE hRPE monolayers survived 4 weeks on PET carriers under the rabbit retina Better xenograft survival may be due to the maintained hRPE cell polarity Atrophy of the retina overlaying the hRPE xenograft remains a future challenge
Collapse
Affiliation(s)
- Boris V Stanzel
- Department of Ophthalmology, University of Bonn, Bonn 53127, Germany
| | - Zengping Liu
- Department of Ophthalmology, University of Bonn, Bonn 53127, Germany
| | - Sudawadee Somboonthanakij
- Department of Ophthalmology, University of Bonn, Bonn 53127, Germany ; Mettapracharak Eye Institute, Raikhing, Nakhon Pathom 73210, Thailand
| | - Warapat Wongsawad
- Department of Ophthalmology, University of Bonn, Bonn 53127, Germany ; Mettapracharak Eye Institute, Raikhing, Nakhon Pathom 73210, Thailand
| | - Ralf Brinken
- Department of Ophthalmology, University of Bonn, Bonn 53127, Germany
| | - Nicole Eter
- Department of Ophthalmology, University of Muenster, Muenster 48149, Germany
| | | | - Frank G Holz
- Department of Ophthalmology, University of Bonn, Bonn 53127, Germany
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | | | | |
Collapse
|
187
|
Westenskow PD, Kurihara T, Friedlander M. Utilizing stem cell-derived RPE cells as a therapeutic intervention for age-related macular degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:323-9. [PMID: 24664714 DOI: 10.1007/978-1-4614-3209-8_41] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Degeneration or dysfunction of the retinal pigment epithelium (RPE) can induce secondary photoreceptor atrophy and catastrophic vision loss in patients with age-related macular degeneration (AMD). AMD is the leading cause of vision loss in the elderly in industrialized countries and no cure exists for the "dry" or atrophic form to date. However, recent pre-clinical data from several groups suggests that embryonic stem cell-derived RPE cell transplantation may prevent photoreceptor degeneration in animal models of RPE degeneration. Another approach may be to derive RPE cells from autologous induced pluripotent stem cells (iPSCs) reprogrammed from dermal tissue. However, the safety of this approach has been questioned on several levels. In this chapter we will summarize work reported by several groups, including our own, that clearly demonstrate that transplanted RPE cells can provide anatomical and functional photoreceptor rescue in animal models of retinal degeneration. We will also discuss some of the prevailing concerns and challenges associated with this technique.
Collapse
Affiliation(s)
- Peter D Westenskow
- Department of Cell Biology, The Scripps Research Institute, MB 216, 10550 N. Torrey Pines Rd, 92014, La Jolla, CA, USA,
| | | | | |
Collapse
|
188
|
Munisi HI, Xie Z, Sengoku S. Exploring innovation in stem cell and regenerative medicine in Japan: the power of the consortium-based approach. Regen Med 2014; 9:467-77. [PMID: 25159064 DOI: 10.2217/rme.14.18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
This article describes a recent trend in Japanese research, development and commercialization toward the application of stem cell technologies. Japan is the world's third largest economy and has a significant national presence in the pharmaceutical and biotechnology businesses; as such, stem cell R&D is abundant in the country. As indicated by the second largest share of patent applications worldwide, Japan had been expected to assert significant added value in the commercialization and industrial application of stem cell technologies; however, difficulties have impeded clinical development in this area, particularly the very small number of clinical trials and approved products for regenerative medicine or cell therapy. To address this 'Japan paradox', this report provides an overview of approaches for the commercialization of stem cell technologies in areas such as drug discovery, cell therapy and regenerative medicine, by discussing representative case examples of listed firms.
Collapse
Affiliation(s)
- Hawa Issa Munisi
- The Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
189
|
Yip HK. Retinal stem cells and regeneration of vision system. Anat Rec (Hoboken) 2013; 297:137-60. [PMID: 24293400 DOI: 10.1002/ar.22800] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/14/2022]
Abstract
The vertebrate retina is a well-characterized model for studying neurogenesis. Retinal neurons and glia are generated in a conserved order from a pool of mutlipotent progenitor cells. During retinal development, retinal stem/progenitor cells (RPC) change their competency over time under the influence of intrinsic (such as transcriptional factors) and extrinsic factors (such as growth factors). In this review, we summarize the roles of these factors, together with the understanding of the signaling pathways that regulate eye development. The information about the interactions between intrinsic and extrinsic factors for retinal cell fate specification is useful to regenerate specific retinal neurons from RPCs. Recent studies have identified RPCs in the retina, which may have important implications in health and disease. Despite the recent advances in stem cell biology, our understanding of many aspects of RPCs in the eye remains limited. PRCs are present in the developing eye of all vertebrates and remain active in lower vertebrates throughout life. In mammals, however, PRCs are quiescent and exhibit very little activity and thus have low capacity for retinal regeneration. A number of different cellular sources of RPCs have been identified in the vertebrate retina. These include PRCs at the retinal margin, pigmented cells in the ciliary body, iris, and retinal pigment epithelium, and Müller cells within the retina. Because PRCs can be isolated and expanded from immature and mature eyes, it is possible now to study these cells in culture and after transplantation in the degenerated retinal tissue. We also examine current knowledge of intrinsic RPCs, and human embryonic stems and induced pluripotent stem cells as potential sources for cell transplant therapy to regenerate the diseased retina.
Collapse
Affiliation(s)
- Henry K Yip
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Adminstrative Region, People's Republic of China; Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Adminstrative Region, People's Republic of China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong Special Adminstrative Region, People's Republic of China
| |
Collapse
|
190
|
Borooah S, Phillips M, Bilican B, Wright A, Wilmut I, Chandran S, Gamm D, Dhillon B. Using human induced pluripotent stem cells to treat retinal disease. Prog Retin Eye Res 2013; 37:163-81. [PMID: 24104210 PMCID: PMC3841575 DOI: 10.1016/j.preteyeres.2013.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 09/06/2013] [Accepted: 09/16/2013] [Indexed: 02/08/2023]
Abstract
The eye is an ideal target for exploiting the potential of human induced pluripotent stem cell (hiPSC) technology in order to understand disease pathways and explore novel therapeutic strategies for inherited retinal disease. The aim of this article is to map the pathway from state-of-the art laboratory-based discoveries to realising the translational potential of this emerging technique. We describe the relevance and routes to establishing hiPSCs in selected models of human retinal disease. Additionally, we define pathways for applying hiPSC technology in treating currently incurable, progressive and blinding retinal disease.
Collapse
Affiliation(s)
- S. Borooah
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
- Ophthalmology, School of Clinical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - M.J. Phillips
- Waisman Center, University of Wisconsin School of Medicine and Public Health, 1500 Highland Ave, Madison, WI 53705, USA
| | - B. Bilican
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - A.F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - I. Wilmut
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - S. Chandran
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - D. Gamm
- Waisman Center, University of Wisconsin School of Medicine and Public Health, 1500 Highland Ave, Madison, WI 53705, USA
- Department of Ophthalmology and Visual Sciences, McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, 1500 Highland Ave, Madison, WI 53705, USA
| | - B. Dhillon
- Ophthalmology, School of Clinical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
191
|
Singh R, Phillips MJ, Kuai D, Meyer J, Martin JM, Smith MA, Perez ET, Shen W, Wallace KA, Capowski EE, Wright LS, Gamm DM. Functional analysis of serially expanded human iPS cell-derived RPE cultures. Invest Ophthalmol Vis Sci 2013; 54:6767-78. [PMID: 24030465 DOI: 10.1167/iovs.13-11943] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE To determine the effects of serial expansion on the cellular, molecular, and functional properties of human iPS cell (hiPSC)-derived RPE cultures. METHODS Fibroblasts obtained from four individuals were reprogrammed into hiPSCs and differentiated to RPE cells using previously described methods. Patches of deeply pigmented hiPSC-RPE were dissected, dissociated, and grown in culture until they re-formed pigmented monolayers. Subsequent passages were obtained by repeated dissociation, expansion, and maturation of RPE into pigmented monolayers. Gene and protein expression profiles and morphological and functional characteristics of hiPSC-RPE at different passages were compared with each other and to human fetal RPE (hfRPE). RESULTS RPE from all four hiPSC lines could be expanded more than 1000-fold when serially passaged as pigmented monolayer cultures. Importantly, expansion of hiPSC-RPE monolayers over the first three passages (P1-P3) resulted in decreased expression of pluripotency and neuroretinal markers and maintenance of characteristic morphological features and gene and protein expression profiles. Furthermore, P1 to P3 hiPSC-RPE monolayers reliably demonstrated functional tight junctions, G-protein-coupled receptor-mediated calcium transients, phagocytosis and degradation of photoreceptor outer segments, and polarized secretion of biomolecules. In contrast, P4 hiPSC-RPE cells failed to form monolayers and possessed altered morphological and functional characteristics and gene expression levels. CONCLUSIONS Highly differentiated, pigmented hiPSC-RPE monolayers can undergo limited serial expansion while retaining key cytological and functional attributes. However, passaging hiPSC-RPE cultures beyond senescence leads to loss of such features. Our findings support limited, controlled passaging of patient-specific hiPSC-RPE to procure cells needed for in vitro disease modeling, drug screening, and cellular transplantation.
Collapse
Affiliation(s)
- Ruchira Singh
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Induction of pluripotency. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:5-25. [PMID: 23696349 DOI: 10.1007/978-94-007-6621-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The molecular and phenotypic irreversibility of mammalian cell differentiation was a fundamental principle of developmental biology at least until the 1980s, despite numerous reports dating back to the 1950s of the induction of pluripotency in amphibian cells by nuclear transfer (NT). Landmark reports in the 1980s and 1990s in sheep progressively challenged this dogmatic assumption; firstly, embryonic development of reconstructed embryos comprising whole (donor) blastomeres fused to enucleated oocytes, and famously, the cloning of Dolly from a terminally differentiated cell. Thus, the intrinsic ability of oocyte-derived factors to reverse the differentiated phenotype was confirmed. The concomitant elucidation of methods for human embryonic stem cell isolation and cultivation presented opportunities for therapeutic cell replacement strategies, particularly through NT of patient nuclei to enucleated oocytes for subsequent isolation of patient-specific (autologous), pluripotent cells from the resulting blastocysts. Associated logistical limitations of working with human oocytes, in addition to ethical and moral objections prompted exploration of alternative approaches to generate autologous stem cells for therapy, utilizing the full repertoire of factors characteristic of pluripotency, primarily through cell fusion and use of pluripotent cell extracts. Stunningly, in 2006, Japanese scientists described somatic cell reprogramming through delivery of four key factors (identified through a deductive approach from 24 candidate genes). Although less efficient than previous approaches, much of current stem cell research adopts this focused approach to cell reprogramming and (autologous) cell therapy. This chapter is a quasi-historical commentary of the various aforementioned approaches for the induction of pluripotency in lineage-committed cells, and introduces transcriptional and epigenetic changes occurring during reprogramming.
Collapse
|
193
|
Tucker BA, Mullins RF, Streb LM, Anfinson K, Eyestone ME, Kaalberg E, Riker MJ, Drack AV, Braun TA, Stone EM. Patient-specific iPSC-derived photoreceptor precursor cells as a means to investigate retinitis pigmentosa. eLife 2013; 2:e00824. [PMID: 23991284 PMCID: PMC3755341 DOI: 10.7554/elife.00824] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/24/2013] [Indexed: 12/11/2022] Open
Abstract
Next-generation and Sanger sequencing were combined to identify disease-causing USH2A mutations in an adult patient with autosomal recessive RP. Induced pluripotent stem cells (iPSCs), generated from the patient's keratinocytes, were differentiated into multi-layer eyecup-like structures with features of human retinal precursor cells. The inner layer of the eyecups contained photoreceptor precursor cells that expressed photoreceptor markers and exhibited axonemes and basal bodies characteristic of outer segments. Analysis of the USH2A transcripts of these cells revealed that one of the patient's mutations causes exonification of intron 40, a translation frameshift and a premature stop codon. Western blotting revealed upregulation of GRP78 and GRP94, suggesting that the patient's other USH2A variant (Arg4192His) causes disease through protein misfolding and ER stress. Transplantation into 4-day-old immunodeficient Crb1 (-/-) mice resulted in the formation of morphologically and immunohistochemically recognizable photoreceptor cells, suggesting that the mutations in this patient act via post-developmental photoreceptor degeneration. DOI:http://dx.doi.org/10.7554/eLife.00824.001.
Collapse
Affiliation(s)
- Budd A Tucker
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Robert F Mullins
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Luan M Streb
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Kristin Anfinson
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Mari E Eyestone
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Emily Kaalberg
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Megan J Riker
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Arlene V Drack
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Terry A Braun
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
- Department of Biomedical Engineering, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Edwin M Stone
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, United States
- Howard Hughes Medical Institute, University of Iowa, Iowa City, United States
| |
Collapse
|
194
|
Satarian L, Javan M, Kiani S, Hajikaram M, Mirnajafi-Zadeh J, Baharvand H. Engrafted human induced pluripotent stem cell-derived anterior specified neural progenitors protect the rat crushed optic nerve. PLoS One 2013; 8:e71855. [PMID: 23977164 PMCID: PMC3747054 DOI: 10.1371/journal.pone.0071855] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 07/05/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Degeneration of retinal ganglion cells (RGCs) is a common occurrence in several eye diseases. This study examined the functional improvement and protection of host RGCs in addition to the survival, integration and neuronal differentiation capabilities of anterior specified neural progenitors (NPs) following intravitreal transplantation. METHODOLOGY/PRINCIPAL FINDINGS NPs were produced under defined conditions from human induced pluripotent stem cells (hiPSCs) and transplanted into rats whose optic nerves have been crushed (ONC). hiPSCs were induced to differentiate into anterior specified NPs by the use of Noggin and retinoic acid. The hiPSC-NPs were labeled by green fluorescent protein or a fluorescent tracer 1,1' -dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI) and injected two days after induction of ONC in hooded rats. Functional analysis according to visual evoked potential recordings showed significant amplitude recovery in animals transplanted with hiPSC-NPs. Retrograde labeling by an intra-collicular DiI injection showed significantly higher numbers of RGCs and spared axons in ONC rats treated with hiPSC-NPs or their conditioned medium (CM). The analysis of CM of hiPSC-NPs showed the secretion of ciliary neurotrophic factor, basic fibroblast growth factor, and insulin-like growth factor. Optic nerve of cell transplanted groups also had increased GAP43 immunoreactivity and myelin staining by FluoroMyelin™ which imply for protection of axons and myelin. At 60 days post-transplantation hiPSC-NPs were integrated into the ganglion cell layer of the retina and expressed neuronal markers. CONCLUSIONS/SIGNIFICANCE The transplantation of anterior specified NPs may improve optic nerve injury through neuroprotection and differentiation into neuronal lineages. These NPs possibly provide a promising new therapeutic approach for traumatic optic nerve injuries and loss of RGCs caused by other diseases.
Collapse
Affiliation(s)
- Leila Satarian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Kiani
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Hajikaram
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| |
Collapse
|
195
|
Steele-Stallard HB, Le Quesne Stabej P, Lenassi E, Luxon LM, Claustres M, Roux AF, Webster AR, Bitner-Glindzicz M. Screening for duplications, deletions and a common intronic mutation detects 35% of second mutations in patients with USH2A monoallelic mutations on Sanger sequencing. Orphanet J Rare Dis 2013; 8:122. [PMID: 23924366 PMCID: PMC3751126 DOI: 10.1186/1750-1172-8-122] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/04/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Usher Syndrome is the leading cause of inherited deaf-blindness. It is divided into three subtypes, of which the most common is Usher type 2, and the USH2A gene accounts for 75-80% of cases. Despite recent sequencing strategies, in our cohort a significant proportion of individuals with Usher type 2 have just one heterozygous disease-causing mutation in USH2A, or no convincing disease-causing mutations across nine Usher genes. The purpose of this study was to improve the molecular diagnosis in these families by screening USH2A for duplications, heterozygous deletions and a common pathogenic deep intronic variant USH2A: c.7595-2144A>G. METHODS Forty-nine Usher type 2 or atypical Usher families who had missing mutations (mono-allelic USH2A or no mutations following Sanger sequencing of nine Usher genes) were screened for duplications/deletions using the USH2A SALSA MLPA reagent kit (MRC-Holland). Identification of USH2A: c.7595-2144A>G was achieved by Sanger sequencing. Mutations were confirmed by a combination of reverse transcription PCR using RNA extracted from nasal epithelial cells or fibroblasts, and by array comparative genomic hybridisation with sequencing across the genomic breakpoints. RESULTS Eight mutations were identified in 23 Usher type 2 families (35%) with one previously identified heterozygous disease-causing mutation in USH2A. These consisted of five heterozygous deletions, one duplication, and two heterozygous instances of the pathogenic variant USH2A: c.7595-2144A>G. No variants were found in the 15 Usher type 2 families with no previously identified disease-causing mutations. In 11 atypical families, none of whom had any previously identified convincing disease-causing mutations, the mutation USH2A: c.7595-2144A>G was identified in a heterozygous state in one family. All five deletions and the heterozygous duplication we report here are novel. This is the first time that a duplication in USH2A has been reported as a cause of Usher syndrome. CONCLUSIONS We found that 8 of 23 (35%) of 'missing' mutations in Usher type 2 probands with only a single heterozygous USH2A mutation detected with Sanger sequencing could be attributed to deletions, duplications or a pathogenic deep intronic variant. Future mutation detection strategies and genetic counselling will need to take into account the prevalence of these types of mutations in order to provide a more comprehensive diagnostic service.
Collapse
|
196
|
Iwamoto H, Ojima T, Hayata K, Katsuda M, Miyazawa M, Iida T, Nakamura M, Nakamori M, Iwahashi M, Yamaue H. Antitumor immune response of dendritic cells (DCs) expressing tumor-associated antigens derived from induced pluripotent stem cells: in comparison to bone marrow-derived DCs. Int J Cancer 2013; 134:332-41. [PMID: 23824921 DOI: 10.1002/ijc.28367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 06/19/2013] [Indexed: 01/06/2023]
Abstract
It is generally accepted that the difficulty in obtaining a sufficient number of functional dendritic cells (DCs) is a serious problem in DC-based immunotherapy. Therefore, we used the induced pluripotent stem (iPS) cell-derived DCs (iPSDCs). If the therapeutic efficacy of iPSDCs is equivalent to that of bone marrow-derived DCs (BMDCs), then the aforementioned problems may be solved. In our study, we induced iPSDCs from iPS cells and examined the capacity for maturation of iPSDCs compared to that of BMDCs in addition to the capacity for migration of iPSDCs to regional lymph nodes. We adenovirally transduced the hgp100 gene, natural tumor antigens, into DCs and immunized mice once with the genetically modified DCs. The cytotoxic activity of CD8 (+) cytotoxic T lymphocytes (CTLs) was assayed using a (51) Cr-release assay. The therapeutic efficacy of the vaccination was examined in a subcutaneous tumor model. Our results showed that iPSDCs have an equal capacity to BMDCs in terms of maturation and migration. Furthermore, hgp100-specific CTLs were generated in mice immunized with genetically modified iPSDCs. These CTLs exhibited as high a level of cytotoxicity against B16 cells as BMDCs. Moreover, vaccination with the genetically modified iPSDCs achieved as high a level of therapeutic efficacy as vaccination with BMDCs. Our study clarified experimentally that genetically modified iPSDCs have an equal capacity to BMDCs in terms of tumor-associated antigen-specific therapeutic antitumor immunity. This vaccination strategy may therefore be useful for future clinical application as a cancer vaccine.
Collapse
Affiliation(s)
- Hiromitsu Iwamoto
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, 641-8510, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Gonzalez-Cordero A, West EL, Pearson RA, Duran Y, Carvalho LS, Chu CJ, Naeem A, Blackford SJI, Georgiadis A, Lakowski J, Hubank M, Smith AJ, Bainbridge JWB, Sowden JC, Ali RR. Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina. Nat Biotechnol 2013; 31:741-7. [PMID: 23873086 PMCID: PMC3826328 DOI: 10.1038/nbt.2643] [Citation(s) in RCA: 282] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 06/22/2013] [Indexed: 11/09/2022]
Abstract
Irreversible blindness caused by loss of photoreceptors may be amenable to cell therapy. We previously demonstrated retinal repair and restoration of vision through transplantation of photoreceptor precursors obtained from postnatal retinas into visually impaired adult mice. Considerable progress has been made in differentiating embryonic stem cells (ESCs) in vitro toward photoreceptor lineages. However, the capability of ESC-derived photoreceptors to integrate after transplantation has not been demonstrated unequivocally. Here, to isolate photoreceptor precursors fit for transplantation, we adapted a recently reported three-dimensional (3D) differentiation protocol that generates neuroretina from mouse ESCs. We show that rod precursors derived by this protocol and selected via a GFP reporter under the control of a Rhodopsin promoter integrate within degenerate retinas of adult mice and mature into outer segment-bearing photoreceptors. Notably, ESC-derived precursors at a developmental stage similar to postnatal days 4-8 integrate more efficiently compared with cells at other stages. This study shows conclusively that ESCs can provide a source of photoreceptors for retinal cell transplantation.
Collapse
Affiliation(s)
- Anai Gonzalez-Cordero
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Emma L. West
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Rachael A. Pearson
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Yanai Duran
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Livia S. Carvalho
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Colin J. Chu
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Arifa Naeem
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Samuel J. I. Blackford
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Anastasios Georgiadis
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Jorn Lakowski
- Developmental Biology Unit, University College London, 30 Guilford Street, London, WC1N 1EH UK
| | - Mike Hubank
- UCL Genomics, University College London, 30 Guilford Street, London, WC1N 1EH UK
| | - Alexander J. Smith
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - James W. B. Bainbridge
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Jane C. Sowden
- Developmental Biology Unit, University College London, 30 Guilford Street, London, WC1N 1EH UK
| | - Robin R. Ali
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
- Molecular Immunology Unit, Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH UK
| |
Collapse
|
198
|
Ramsden CM, Powner MB, Carr AJF, Smart MJK, da Cruz L, Coffey PJ. Stem cells in retinal regeneration: past, present and future. Development 2013; 140:2576-85. [PMID: 23715550 DOI: 10.1242/dev.092270] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell therapy for retinal disease is under way, and several clinical trials are currently recruiting. These trials use human embryonic, foetal and umbilical cord tissue-derived stem cells and bone marrow-derived stem cells to treat visual disorders such as age-related macular degeneration, Stargardt's disease and retinitis pigmentosa. Over a decade of analysing the developmental cues involved in retinal generation and stem cell biology, coupled with extensive surgical research, have yielded differing cellular approaches to tackle these retinopathies. Here, we review these various stem cell-based approaches for treating retinal diseases and discuss future directions and challenges for the field.
Collapse
Affiliation(s)
- Conor M Ramsden
- The London Project to Cure Blindness, Division of ORBIT, Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | | | | | | | | | | |
Collapse
|
199
|
Gonzalez-Cordero A, West EL, Pearson RA, Duran Y, Carvalho LS, Chu CJ, Naeem A, Blackford SJI, Georgiadis A, Lakowski J, Hubank M, Smith AJ, Bainbridge JWB, Sowden JC, Ali RR. Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina. Nat Biotechnol 2013. [PMID: 23873086 DOI: 10.1038/nbt.2643]] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Irreversible blindness caused by loss of photoreceptors may be amenable to cell therapy. We previously demonstrated retinal repair and restoration of vision through transplantation of photoreceptor precursors obtained from postnatal retinas into visually impaired adult mice. Considerable progress has been made in differentiating embryonic stem cells (ESCs) in vitro toward photoreceptor lineages. However, the capability of ESC-derived photoreceptors to integrate after transplantation has not been demonstrated unequivocally. Here, to isolate photoreceptor precursors fit for transplantation, we adapted a recently reported three-dimensional (3D) differentiation protocol that generates neuroretina from mouse ESCs. We show that rod precursors derived by this protocol and selected via a GFP reporter under the control of a Rhodopsin promoter integrate within degenerate retinas of adult mice and mature into outer segment-bearing photoreceptors. Notably, ESC-derived precursors at a developmental stage similar to postnatal days 4-8 integrate more efficiently compared with cells at other stages. This study shows conclusively that ESCs can provide a source of photoreceptors for retinal cell transplantation.
Collapse
Affiliation(s)
- Anai Gonzalez-Cordero
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Emma L West
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Rachael A Pearson
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Yanai Duran
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Livia S Carvalho
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Colin J Chu
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Arifa Naeem
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Samuel J I Blackford
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Anastasios Georgiadis
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Jorn Lakowski
- Developmental Biology Unit, University College London, 30 Guilford Street, London, WC1N 1EH UK
| | - Mike Hubank
- UCL Genomics, University College London, 30 Guilford Street, London, WC1N 1EH UK
| | - Alexander J Smith
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - James W B Bainbridge
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Jane C Sowden
- Developmental Biology Unit, University College London, 30 Guilford Street, London, WC1N 1EH UK
| | - Robin R Ali
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK.,Molecular Immunology Unit, Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH UK
| |
Collapse
|
200
|
Mekala SR, Vauhini V, Nagarajan U, Maddileti S, Gaddipati S, Mariappan I. Derivation, characterization and retinal differentiation of induced pluripotent stem cells. J Biosci 2013; 38:123-34. [PMID: 23385820 DOI: 10.1007/s12038-012-9296-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Millions of people world over suffer visual disability due to retinal dystrophies which can be age-related or a genetic disorder resulting in gradual degeneration of the retinal pigmented epithelial (RPE) cells and photoreceptors. Therefore, cell replacement therapy offers a great promise in treating such diseases. Since the adult retina does not harbour any stem cells, alternative stem cell sources like the embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) offer a great promise for generating different cell types of the retina. Here, we report the derivation of four iPSC lines from mouse embryonic fibroblasts (MEFs) using a cocktail of recombinant retroviruses carrying the genes for Oct4, Sox2, Klf4 and cMyc. The iPS clone MEF-4F3 was further characterized for stemness marker expression and stable reprogramming by immunocytochemistry, FACS and RT-PCR analysis. Methylation analysis of the nanog promoter confirmed the reprogrammed epigenetic state. Pluripotency was confirmed by embryoid body (EB) formation and lineage-specific marker expression. Also, upon retinal differentiation, patches of pigmented cells with typical cobble-stone phenotype similar to RPE cells are generated within 6 weeks and they expressed ZO-1 (tight junction protein), RPE65 and bestrophin (mature RPE markers) and showed phagocytic activity by the uptake of fluorescent latex beads.
Collapse
Affiliation(s)
- Subba Rao Mekala
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Prof Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, C-TRACER, L.V. Prasad Eye Institute, Hyderabad, India
| | | | | | | | | | | |
Collapse
|