301
|
Liu Y, Wang J, Luo Y, Chen S, Lewallen M, Xie T. Stem Cells and Ocular Tissue Regeneration. ASIA-PACIFIC JOURNAL OF OPHTHALMOLOGY (PHILADELPHIA, PA.) 2013; 2:111-8. [PMID: 26108048 DOI: 10.1097/apo.0b013e31828615b7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE Millions worldwide have visual impairments caused by dysfunctional eye components, including cornea, lens, retina, and optic nerve, or the visual cortex in the brain. Insufficient cornea donation and inherent artificial lens problems demand alternative treatment strategies for cornea diseases and cataracts, whereas retinal degenerative diseases, including glaucoma, macular degeneration, and retinitis pigmentosa, still lack effective treatments. Stem cells have been investigated for their potential in various eye-specific pathologies to replace lost retinal ganglion cells and photoreceptors in retinal degenerative diseases and toward engineering transplantable patient-specific cornea or lens. DESIGN Many stem cell types, including putative resident eye stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells, have been investigated for their potential to generate specific cell types in the eye in culture and after transplantation and to engineer eye tissues in combination with structural scaffolds. METHOD Cultured stem cells and in vitro differentiated eye-specific cells are transplanted into different locations of the eye to test their ability to produce functional cells for supporting eye functions. In addition, stem cells have been directly tested in vitro for their capacity to engineer eye-specific tissues. RESULTS Different stem cell types have been shown to have distinct capacities to produce eye-specific cells or even the entire retina. CONCLUSIONS Stem cells offer great hope for treating various eye pathologies. Despite recent progress, many challenges must still be overcome before the era of stem cell-based therapy in the eye truly arrives.
Collapse
Affiliation(s)
- Yizhi Liu
- From the *State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, PR China; and †Stowers Institute for Medical Research, Kansas City, MO
| | | | | | | | | | | |
Collapse
|
302
|
Zhao J, Luo J, Zhang K. Is Era of Ocular Regeneration Near? ASIA-PACIFIC JOURNAL OF OPHTHALMOLOGY (PHILADELPHIA, PA.) 2013; 2:71-2. [PMID: 26108040 DOI: 10.1097/apo.0b013e318290e20c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Jiagang Zhao
- From the *Department of Ophthalmology and Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China; Department of Ophthalmology,†Institute for Genomic Medicine and Shiley Eye Center, University of California San Diego, La Jolla, CA; ‡Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China; §Veterans Administration Healthcare System, San Diego, CA
| | | | | |
Collapse
|
303
|
Nasonkin IO, Merbs SL, Lazo K, Oliver VF, Brooks M, Patel K, Enke RA, Nellissery J, Jamrich M, Le YZ, Bharti K, Fariss RN, Rachel RA, Zack DJ, Rodriguez-Boulan EJ, Swaroop A. Conditional knockdown of DNA methyltransferase 1 reveals a key role of retinal pigment epithelium integrity in photoreceptor outer segment morphogenesis. Development 2013; 140:1330-41. [PMID: 23406904 DOI: 10.1242/dev.086603] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Dysfunction or death of photoreceptors is the primary cause of vision loss in retinal and macular degenerative diseases. As photoreceptors have an intimate relationship with the retinal pigment epithelium (RPE) for exchange of macromolecules, removal of shed membrane discs and retinoid recycling, an improved understanding of the development of the photoreceptor-RPE complex will allow better design of gene- and cell-based therapies. To explore the epigenetic contribution to retinal development we generated conditional knockout alleles of DNA methyltransferase 1 (Dnmt1) in mice. Conditional Dnmt1 knockdown in early eye development mediated by Rx-Cre did not produce lamination or cell fate defects, except in cones; however, the photoreceptors completely lacked outer segments despite near normal expression of phototransduction and cilia genes. We also identified disruption of RPE morphology and polarization as early as E15.5. Defects in outer segment biogenesis were evident with Dnmt1 exon excision only in RPE, but not when excision was directed exclusively to photoreceptors. We detected a reduction in DNA methylation of LINE1 elements (a measure of global DNA methylation) in developing mutant RPE as compared with neural retina, and of Tuba3a, which exhibited dramatically increased expression in mutant retina. These results demonstrate a unique function of DNMT1-mediated DNA methylation in controlling RPE apicobasal polarity and neural retina differentiation. We also establish a model to study the epigenetic mechanisms and signaling pathways that guide the modulation of photoreceptor outer segment morphogenesis by RPE during retinal development and disease.
Collapse
Affiliation(s)
- Igor O Nasonkin
- 1Neurobiology-Neurodegeneration and Repair Laboratory (N-NRL), National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
304
|
Abstract
Studies on stem cell aging are uncovering molecular mechanisms of regenerative decline, providing new insight into potential rejuvenating therapies.
Collapse
|
305
|
Yang J, Nong E, Tsang SH. Induced pluripotent stem cells and retinal degeneration treatment. EXPERT REVIEW OF OPHTHALMOLOGY 2013. [DOI: 10.1586/eop.12.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
306
|
Reversal of end-stage retinal degeneration and restoration of visual function by photoreceptor transplantation. Proc Natl Acad Sci U S A 2013; 110:1101-6. [PMID: 23288902 DOI: 10.1073/pnas.1119416110] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
One strategy to restore vision in retinitis pigmentosa and age-related macular degeneration is cell replacement. Typically, patients lose vision when the outer retinal photoreceptor layer is lost, and so the therapeutic goal would be to restore vision at this stage of disease. It is not currently known if a degenerate retina lacking the outer nuclear layer of photoreceptor cells would allow the survival, maturation, and reconnection of replacement photoreceptors, as prior studies used hosts with a preexisting outer nuclear layer at the time of treatment. Here, using a murine model of severe human retinitis pigmentosa at a stage when no host rod cells remain, we show that transplanted rod precursors can reform an anatomically distinct and appropriately polarized outer nuclear layer. A trilaminar organization was returned to rd1 hosts that had only two retinal layers before treatment. The newly introduced precursors were able to resume their developmental program in the degenerate host niche to become mature rods with light-sensitive outer segments, reconnecting with host neurons downstream. Visual function, assayed in the same animals before and after transplantation, was restored in animals with zero rod function at baseline. These observations suggest that a cell therapy approach may reconstitute a light-sensitive cell layer de novo and hence repair a structurally damaged visual circuit. Rather than placing discrete photoreceptors among preexisting host outer retinal cells, total photoreceptor layer reconstruction may provide a clinically relevant model to investigate cell-based strategies for retinal repair.
Collapse
|
307
|
|
308
|
|
309
|
|
310
|
|
311
|
Gullapalli VK, Khodair MA, Wang H, Sugino IK, Madreperla S, Zarbin MA. Transplantation Frontiers. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00125-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
312
|
Osakada F, Takahashi M. Stem Cells in the Developing and Adult Nervous System. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
313
|
Ng TK, Lam DSC, Cheung HS. Prospects of Stem Cells for Retinal Diseases. ASIA-PACIFIC JOURNAL OF OPHTHALMOLOGY (PHILADELPHIA, PA.) 2013; 2:57-63. [PMID: 26107868 DOI: 10.1097/apo.0b013e31827e3e5d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Retinal diseases, including glaucoma, retinitis pigmentosa, diabetic retinopathy, and age-related macular degeneration, are the leading causes of irreversible visual impairment and blindness in developed countries. Traditional and current treatment regimens are based on surgical or medical interventions to slow down the disease progression. However, the number of retinal cells would continue to diminish, and the diseases could not be completely cured. There is an emerging role of stem cells in retinal research. The stem cell therapy on retinal diseases is based on 2 theories: cell replacement therapy and neuroprotective effect. The former hypothesizes that new retinal cells could be regenerated from stem cells to substitute the damaged cells in the diseased retina, whereas the latter believes that the paracrine effects of stem cells modulate the microenvironments of the diseased retina so as to protect the retinal neurons. This article summarizes the choice of stem cells in retinal research. Moreover, the current progress of retinal research on stem cells and the clinical applications of stem cells on retinal diseases are reviewed. In addition, potential challenges and future prospects of retinal stem cell research are discussed.
Collapse
Affiliation(s)
- Tsz Kin Ng
- From the *Geriatric Research, Education and Clinical Center, Miami Veterans Affairs Medical Center, Miami, FL; †State Key Laboratory in Ophthalmology & Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China; and ‡Department of Biomedical Engineering, College of Engineering, University of Miami, Coral Gables, FL
| | | | | |
Collapse
|
314
|
Reh TA. The Development of the Retina. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
315
|
Wang L, Lang LL, Wang Y, Shi S, Liu L. Prostaglandin E2Enhances Proliferation, Dedifferentiation and Stem-Like Properties of Rat Retinal Müller Glial Cells in vitro. Ophthalmic Res 2013; 49:100-7. [DOI: 10.1159/000345256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/15/2012] [Indexed: 01/08/2023]
|
316
|
Repair of the degenerate retina by photoreceptor transplantation. Proc Natl Acad Sci U S A 2012; 110:354-9. [PMID: 23248312 DOI: 10.1073/pnas.1212677110] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Despite different aetiologies, age-related macular degeneration and most inherited retinal disorders culminate in the same final common pathway, the loss of photoreceptors. There are few treatments and none reverse the loss of vision. Photoreceptor replacement by transplantation is proposed as a broad treatment strategy applicable to all degenerations. Recently, we demonstrated restoration of vision following rod-photoreceptor transplantation into a mouse model of stationary night-blindness, raising the critical question of whether photoreceptor replacement is equally effective in different types and stages of degeneration. We present a comprehensive assessment of rod-photoreceptor transplantation across six murine models of inherited photoreceptor degeneration. Transplantation is feasible in all models examined but disease type has a major impact on outcome, as assessed both by the morphology and number of integrated rod-photoreceptors. Integration can increase (Prph2(+/Δ307)), decrease (Crb1(rd8/rd8), Gnat1(-/-), Rho(-/-)), or remain constant (PDE6β(rd1/rd1), Prph2(rd2/rd2)) with disease progression, depending upon the gene defect, with no correlation with severity. Robust integration is possible even in late-stage disease. Glial scarring and outer limiting membrane integrity, features that change with degeneration, significantly affect transplanted photoreceptor integration. Combined breakdown of these barriers markedly increases integration in a model with an intact outer limiting membrane, strong gliotic response, and otherwise poor transplantation outcome (Rho(-/-)), leading to an eightfold increase in integration and restoration of visual function. Thus, it is possible to achieve robust integration across a broad range of inherited retinopathies. Moreover, transplantation outcome can be improved by administering appropriate, tailored manipulations of the recipient environment.
Collapse
|
317
|
Li F, Niyibizi C. Cells derived from murine induced pluripotent stem cells (iPSC) by treatment with members of TGF-beta family give rise to osteoblasts differentiation and form bone in vivo. BMC Cell Biol 2012; 13:35. [PMID: 23241430 PMCID: PMC3541062 DOI: 10.1186/1471-2121-13-35] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 12/06/2012] [Indexed: 01/17/2023] Open
Abstract
Background Induced pluripotent stem cells (iPSC) are generated by reprogramming somatic cells into embryonic like state (ESC) using defined factors. There is great interest in these cells because of their potential for application in regenerative medicine. Results iPSC reprogrammed from murine tail tip fibroblasts were exposed to retinoic acid alone (RA) or in combination with TGF-β1 and 3, basic fibroblast growth factor (bFGF) or bone morphogenetic protein 2 (BMP-2). The resulting cells expressed selected putative mesenchymal stem cells (MSCs) markers; differentiated toward osteoblasts and adipocytic cell lineages in vitro at varying degrees. TGF-beta1 and 3 derived-cells possessed higher potential to give rise to osteoblasts than bFGF or BMP-2 derived-cells while BMP-2 derived cells exhibited a higher potential to differentiate toward adipocytic lineage. TGF-β1 in combination with RA derived-cells seeded onto HA/TCP ceramics and implanted in mice deposited typical bone. Immunofluorescence staining for bone specific proteins in cell seeded scaffolds tissue sections confirmed differentiation of the cells into osteoblasts in vivo. Conclusions The results demonstrate that TGF-beta family of proteins could potentially be used to generate murine iPSC derived-cells with potential for osteoblasts differentiation and bone formation in vivo and thus for application in musculoskeletal tissue repair and regeneration.
Collapse
Affiliation(s)
- Feng Li
- Department of Orthopaedics and Rehabilitation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | |
Collapse
|
318
|
Abstract
Three embryonic tissue sources-the neural ectoderm, the surface ectoderm, and the periocular mesenchyme-contribute to the formation of the mammalian eye. For this reason, the developing eye has presented an invaluable system for studying the interactions among cells and, more recently, genes, in specifying cell fate. This article describes how the eye primordium is specified in the anterior neural plate by four eye field transcription factors and how the optic vesicle becomes regionalized into three distinct tissue types. Specific attention is given to how cross talk between the optic vesicle and surface ectoderm contributes to lens and optic cup formation. This article also describes how signaling networks and cell movements set up axes in the optic cup and establish the multiple cell fates important for vision. How multipotent retinal progenitor cells give rise to the six neuronal and one glial cell type in the mature retina is also explained. Finally, the history and progress of cellular therapeutics for the treatment of degenerative eye disease is outlined. Throughout this article, special attention is given to how disruption of gene function causes ocular malformation in humans. Indeed, the accessibility of the eye has contributed much to our understanding of the basic processes involved in mammalian development.
Collapse
Affiliation(s)
- Whitney Heavner
- UNC Neuroscience Center, Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
319
|
Gregory-Evans CY, Wallace VA, Gregory-Evans K. Gene networks: dissecting pathways in retinal development and disease. Prog Retin Eye Res 2012; 33:40-66. [PMID: 23128416 DOI: 10.1016/j.preteyeres.2012.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 10/18/2012] [Accepted: 10/19/2012] [Indexed: 01/21/2023]
Abstract
During retinal neurogenesis, diverse cellular subtypes originate from multipotent neural progenitors in a spatiotemporal order leading to a highly specialized laminar structure combined with a distinct mosaic architecture. This is driven by the combinatorial action of transcription factors and signaling molecules which specify cell fate and differentiation. The emerging approach of gene network analysis has allowed a better understanding of the functional relationships between genes expressed in the developing retina. For instance, these gene networks have identified transcriptional hubs that have revealed potential targets and pathways for the development of therapeutic options for retinal diseases. Much of the current knowledge has been informed by targeted gene deletion experiments and gain-of-functional analysis. In this review we will provide an update on retinal development gene networks and address the wider implications for future disease therapeutics.
Collapse
Affiliation(s)
- Cheryl Y Gregory-Evans
- Department of Ophthalmology, University of British Columbia, Vancouver, BC V5Z 3N9, Canada.
| | | | | |
Collapse
|
320
|
Seiler MJ, Aramant RB. Cell replacement and visual restoration by retinal sheet transplants. Prog Retin Eye Res 2012; 31:661-87. [PMID: 22771454 PMCID: PMC3472113 DOI: 10.1016/j.preteyeres.2012.06.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 06/19/2012] [Accepted: 06/23/2012] [Indexed: 12/18/2022]
Abstract
Retinal diseases such as age-related macular degeneration (ARMD) and retinitis pigmentosa (RP) affect millions of people. Replacing lost cells with new cells that connect with the still functional part of the host retina might repair a degenerating retina and restore eyesight to an unknown extent. A unique model, subretinal transplantation of freshly dissected sheets of fetal-derived retinal progenitor cells, combined with its retinal pigment epithelium (RPE), has demonstrated successful results in both animals and humans. Most other approaches are restricted to rescue endogenous retinal cells of the recipient in earlier disease stages by a 'nursing' role of the implanted cells and are not aimed at neural retinal cell replacement. Sheet transplants restore lost visual responses in several retinal degeneration models in the superior colliculus (SC) corresponding to the location of the transplant in the retina. They do not simply preserve visual performance - they increase visual responsiveness to light. Restoration of visual responses in the SC can be directly traced to neural cells in the transplant, demonstrating that synaptic connections between transplant and host contribute to the visual improvement. Transplant processes invade the inner plexiform layer of the host retina and form synapses with presumable host cells. In a Phase II trial of RP and ARMD patients, transplants of retina together with its RPE improved visual acuity. In summary, retinal progenitor sheet transplantation provides an excellent model to answer questions about how to repair and restore function of a degenerating retina. Supply of fetal donor tissue will always be limited but the model can set a standard and provide an informative base for optimal cell replacement therapies such as embryonic stem cell (ESC)-derived therapy.
Collapse
Affiliation(s)
- Magdalene J Seiler
- Department of Anatomy & Neurobiology, Reeve-Irvine Research Center, Sue & Bill Gross Stem Cell Research Center, University of California at Irvine, 1101 Gross Hall, 845 Health Science Rd., Irvine, CA 92697-4265, USA.
| | | |
Collapse
|
321
|
Polosukhina A, Litt J, Tochitsky I, Nemargut J, Sychev Y, De Kouchkovsky I, Huang T, Borges K, Trauner D, Van Gelder RN, Kramer RH. Photochemical restoration of visual responses in blind mice. Neuron 2012; 75:271-82. [PMID: 22841312 DOI: 10.1016/j.neuron.2012.05.022] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2012] [Indexed: 11/25/2022]
Abstract
Retinitis pigmentosa (RP) and age-related macular degeneration (AMD) are degenerative blinding diseases caused by the death of rods and cones, leaving the remainder of the visual system intact but largely unable to respond to light. Here, we show that AAQ, a synthetic small molecule photoswitch, can restore light sensitivity to the retina and behavioral responses in vivo in mouse models of RP, without exogenous gene delivery. Brief application of AAQ bestows prolonged light sensitivity on multiple types of retinal neurons, resulting in synaptically amplified responses and center-surround antagonism in arrays of retinal ganglion cells (RGCs). Intraocular injection of AAQ restores the pupillary light reflex and locomotory light avoidance behavior in mice lacking retinal photoreceptors, indicating reconstitution of light signaling to brain circuits. AAQ and related photoswitch molecules present a potential drug strategy for restoring retinal function in degenerative blinding diseases.
Collapse
Affiliation(s)
- Aleksandra Polosukhina
- Vision Science Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
Becker S, Jayaram H, Limb GA. Recent Advances towards the Clinical Application of Stem Cells for Retinal Regeneration. Cells 2012; 1:851-73. [PMID: 24710533 PMCID: PMC3901131 DOI: 10.3390/cells1040851] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 09/29/2012] [Accepted: 10/10/2012] [Indexed: 01/10/2023] Open
Abstract
Retinal degenerative diseases constitute a major cause of irreversible blindness in the world. Stem cell-based therapies offer hope for these patients at risk of or suffering from blindness due to the deterioration of the neural retina. Various sources of stem cells are currently being investigated, ranging from human embryonic stem cells to adult-derived induced pluripotent stem cells as well as human Müller stem cells, with the first clinical trials to investigate the safety and tolerability of human embryonic stem cell-derived retinal pigment epithelium cells having recently commenced. This review aims to summarize the latest advances in the development of stem cell strategies for the replacement of retinal neurons and their supportive cells, the retinal pigment epithelium (RPE) affected by retinal degenerative conditions. Particular emphasis will be given to the advances in stem cell transplantation and the challenges associated with their translation into clinical practice.
Collapse
Affiliation(s)
- Silke Becker
- Ocular Biology and Therapeutics, Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| | - Hari Jayaram
- Ocular Biology and Therapeutics, Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| | - G Astrid Limb
- Ocular Biology and Therapeutics, Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
323
|
Barak Y, Heroman WJ, Tezel TH. The past, present, and future of exudative age-related macular degeneration treatment. Middle East Afr J Ophthalmol 2012; 19:43-51. [PMID: 22346114 PMCID: PMC3277024 DOI: 10.4103/0974-9233.92115] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Treatment of exudative age-related macular degeneration has been revolutionized within the last 6 years with the introduction of vascular endothelial growth factor neutralizing agents. Previously popular “destructive treatments,” such as laser photocoagulation and photodynamic treatment have either been abandoned or used as an adjunct to pharmacotherapy. Despite the increase in vision after antivascular endothelial growth factor (VEGF) agents, they require repetitive and costly intravitreal injections that also carry the inherit risks of infection, retinal tears, and detachment. Several new and more potent VEGF inhibitors are at different stages of development. The goal of evolving pharmacotherapy is to preserve the therapeutic effect while reducing or eliminating the discomfort of intravitreal drug delivery, as well as identify new therapeutic targets. Complement inhibitors, immunomodulators, integrin inhibitors are a few of the new class of drugs that are expected to be in our armamentarium soon. Current medications act to decrease leakage through abnormal subretinal choroidal vasculature and promote involution. However, these medications are only effective in treating the active stage of the choroidal neovascular membrane. Restoration of vision of a large number of patients with involuted choroidal neovascular membranes is warranted. For this purpose, tissue engineering techniques have been employed to reconstruct the subretinal anatomy. Discovery of biomarkers, pharmacogenetics, and very specific targeting holds the promise of increased potency and safety in the future.
Collapse
Affiliation(s)
- Yoreh Barak
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | |
Collapse
|
324
|
Eberle D, Kurth T, Santos-Ferreira T, Wilson J, Corbeil D, Ader M. Outer segment formation of transplanted photoreceptor precursor cells. PLoS One 2012; 7:e46305. [PMID: 23029471 PMCID: PMC3460822 DOI: 10.1371/journal.pone.0046305] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 08/30/2012] [Indexed: 12/30/2022] Open
Abstract
Transplantation of photoreceptor precursor cells (PPCs) into the retina represents a promising treatment for cell replacement in blinding diseases characterized by photoreceptor loss. In preclinical studies, we and others demonstrated that grafted PPCs integrate into the host outer nuclear layer (ONL) and develop into mature photoreceptors. However, a key feature of light detecting photoreceptors, the outer segment (OS) with natively aligned disc membrane staples, has not been studied in detail following transplantation. Therefore, we used as donor cells PPCs isolated from neonatal double transgenic reporter mice in which OSs are selectively labeled by green fluorescent protein while cell bodies are highlighted by red fluorescent protein. PPCs were enriched using CD73-based magnetic associated cell sorting and subsequently transplanted into either adult wild-type or a model of autosomal-dominant retinal degeneration mice. Three weeks post-transplantation, donor photoreceptors were identified based on fluorescent-reporter expression and OS formation was monitored at light and electron microscopy levels. Donor cells that properly integrated into the host wild-type retina developed OSs with the formation of a connecting cilium and well-aligned disc membrane staples similar to the surrounding native cells of the host. Surprisingly, the majority of not-integrated PPCs that remained in the sub-retinal space also generated native-like OSs in wild-type mice and those affected by retinal degeneration. Moreover, they showed an improved photoreceptor maturation and OS formation by comparison to donor cells located on the vitreous side suggesting that environmental cues influence the PPC differentiation and maturation. We conclude that transplanted PPCs, whether integrated or not into the host ONL, are able to generate the cellular structure for effective light detection, a phenomenon observed in wild-type as well as in degenerated retinas. Given that patients suffering from retinitis pigmentosa lose almost all photoreceptors, our findings are of utmost importance for the development of cell-based therapies.
Collapse
Affiliation(s)
- Dominic Eberle
- DFG-Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence/TU-Dresden, Dresden, Germany
| | - Thomas Kurth
- DFG-Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence/TU-Dresden, Dresden, Germany
| | - Tiago Santos-Ferreira
- DFG-Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence/TU-Dresden, Dresden, Germany
| | - John Wilson
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Denis Corbeil
- DFG-Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence/TU-Dresden, Dresden, Germany
- Biotechnology Center, TU-Dresden, Dresden, Germany
| | - Marius Ader
- DFG-Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence/TU-Dresden, Dresden, Germany
| |
Collapse
|
325
|
Megaw R, Dhillon B. Towards photoreceptor transplantation for visual recovery. Regen Med 2012; 7:627-9. [PMID: 22954431 DOI: 10.2217/rme.12.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
326
|
The emerging role of stem cells in ocular neurodegeneration: hype or hope? Mol Cell Biochem 2012; 365:65-76. [PMID: 22290231 DOI: 10.1007/s11010-012-1244-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 01/14/2012] [Indexed: 01/14/2023]
Abstract
Affecting over a hundred million individuals worldwide, retinal diseases are among the leading causes of irreversible visual impairment and blindness. Thus, an appropriate study models, especially animal models, are essential to furthering our understanding of the etiology, pathology, and progression of these diseases. In this review, we provide an overview of retinal disorders in the context of biotherapeutic approaches in these disorders.
Collapse
|
327
|
Thumann G. Prospectives for gene therapy of retinal degenerations. Curr Genomics 2012; 13:350-62. [PMID: 23372421 PMCID: PMC3401892 DOI: 10.2174/138920212801619214] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/08/2012] [Accepted: 05/22/2012] [Indexed: 12/16/2022] Open
Abstract
Retinal degenerations encompass a large number of diseases in which the retina and associated retinal pigment epithelial (RPE) cells progressively degenerate leading to severe visual disorders or blindness. Retinal degenerations can be divided into two groups, a group in which the defect has been linked to a specific gene and a second group that has a complex etiology that includes environmental and genetic influences. The first group encompasses a number of relatively rare diseases with the most prevalent being Retinitis pigmentosa that affects approximately 1 million individuals worldwide. Attempts have been made to correct the defective gene by transfecting the appropriate cells with the wild-type gene and while these attempts have been successful in animal models, human gene therapy for these inherited retinal degenerations has only begun recently and the results are promising. To the second group belong glaucoma, age-related macular degeneration (AMD) and diabetic retinopathy (DR). These retinal degenerations have a genetic component since they occur more often in families with affected probands but they are also linked to environmental factors, specifically elevated intraocular pressure, age and high blood sugar levels respectively. The economic and medical impact of these three diseases can be assessed by the number of individuals affected; AMD affects over 30 million, DR over 40 million and glaucoma over 65 million individuals worldwide. The basic defect in these diseases appears to be the relative lack of a neurogenic environment; the neovascularization that often accompanies these diseases has suggested that a decrease in pigment epithelium-derived factor (PEDF), at least in part, may be responsible for the neurodegeneration since PEDF is not only an effective neurogenic and neuroprotective agent but also a potent inhibitor of neovascularization. In the last few years inhibitors of vascularization, especially antibodies against vascular endothelial cell growth factors (VEGF), have been used to prevent the neovascularization that accompanies AMD and DR resulting in the amelioration of vision in a significant number of patients. In animal models it has been shown that transfection of RPE cells with the gene for PEDF and other growth factors can prevent or slow degeneration. A limited number of studies in humans have also shown that transfection of RPE cells in vivo with the gene for PEDF is effective in preventing degeneration and restore vision. Most of these studies have used virally mediated gene delivery with all its accompanying side effects and have not been widely used. New techniques using non-viral protocols that allow efficient delivery and permanent integration of the transgene into the host cell genome offer novel opportunities for effective treatment of retinal degenerations.
Collapse
Affiliation(s)
- Gabriele Thumann
- Universitätsaugenklinik / IZKF Aachen, RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany
| |
Collapse
|
328
|
In vitro expanded stem cells from the developing retina fail to generate photoreceptors but differentiate into myelinating oligodendrocytes. PLoS One 2012; 7:e41798. [PMID: 22848612 PMCID: PMC3405018 DOI: 10.1371/journal.pone.0041798] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/25/2012] [Indexed: 01/06/2023] Open
Abstract
Cell transplantation to treat retinal degenerative diseases represents an option for the replacement of lost photoreceptor cells. In vitro expandable cells isolated from the developing mammalian retina have been suggested as a potential source for the generation of high numbers of donor photoreceptors. In this study we used standardized culture conditions based on the presence of the mitogens FGF-2 and EGF to generate high numbers of cells in vitro from the developing mouse retina. These presumptive ‘retinal stem cells’ (‘RSCs’) can be propagated as monolayer cultures over multiple passages, express markers of undifferentiated neural cells, and generate neuronal and glial cell types upon withdrawal of mitogens in vitro or following transplantation into the adult mouse retina. The proportion of neuronal differentiation can be significantly increased by stepwise removal of mitogens and inhibition of the notch signaling pathway. However, ‘RSCs’, by contrast to their primary counterparts in vivo, i.e. retinal progenitor cells, loose the expression of retina-specific progenitor markers like Rax and Chx10 after passaging and fail to differentiate into photoreceptors both in vitro or after intraretinal transplantation. Notably, ‘RSCs’ can be induced to differentiate into myelinating oligodendrocytes, a cell type not generated by primary retinal progenitor cells. Based on these findings we conclude that ‘RSCs’ expanded in high concentrations of FGF-2 and EGF loose their retinal identity and acquire features of in vitro expandable neural stem-like cells making them an inappropriate cell source for strategies aimed at replacing photoreceptor cells in the degenerated retina.
Collapse
|
329
|
Mellough CB, Sernagor E, Moreno-Gimeno I, Steel DHW, Lako M. Efficient stage-specific differentiation of human pluripotent stem cells toward retinal photoreceptor cells. Stem Cells 2012; 30:673-86. [PMID: 22267304 DOI: 10.1002/stem.1037] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recent successes in the stem cell field have identified some of the key chemical and biological cues which drive photoreceptor derivation from human embryonic stem cells (hESC) and human induced pluripotent stem cells (hiPSC); however, the efficiency of this process is variable. We have designed a three-step photoreceptor differentiation protocol combining previously published methods that direct the differentiation of hESC and hiPSC toward a retinal lineage, which we further modified with additional supplements selected on the basis of reports from the eye field and retinal development. We report that hESC and hiPSC differentiating under our regimen over a 60 day period sequentially acquire markers associated with neural, retinal field, retinal pigmented epithelium and photoreceptor cells, including mature photoreceptor markers OPN1SW and RHODOPSIN with a higher efficiency than previously reported. In addition, we report the ability of hESC and hiPSC cultures to generate neural and retinal phenotypes under minimal culture conditions, which may be linked to their ability to endogenously upregulate the expression of a range of factors important for retinal cell type specification. However, cultures that were differentiated with full supplementation under our photoreceptor-induction regimen achieve this within a significantly shorter time frame and show a substantial increase in the expression of photoreceptor-specific markers in comparison to cultures differentiated under minimal conditions. Interestingly, cultures supplemented only with B27 and/or N2 displayed comparable differentiation efficiency to those under full supplementation, indicating a key role for B27 and N2 during the differentiation process. Furthermore, our data highlight an important role for Dkk1 and Noggin in enhancing the differentiation of hESC and hiPSC toward retinal progenitor cells and photoreceptor precursors during the early stages of differentiation, while suggesting that further maturation of these cells into photoreceptors may not require additional factors and can ensue under minimal culture conditions.
Collapse
Affiliation(s)
- Carla B Mellough
- Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | | | | | | | | |
Collapse
|
330
|
Kim KY, Jung YW, Sullivan GJ, Chung L, Park IH. Cellular reprogramming: a novel tool for investigating autism spectrum disorders. Trends Mol Med 2012; 18:463-71. [PMID: 22771169 DOI: 10.1016/j.molmed.2012.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 06/05/2012] [Accepted: 06/11/2012] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairment in reciprocal social interaction and communication, as well as the manifestation of stereotyped behaviors. Despite much effort, ASDs are not yet fully understood. Advanced genetics and genomics technologies have recently identified novel ASD genes, and approaches using genetically engineered murine models or postmortem human brain have facilitated understanding ASD. Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) provides unprecedented opportunities in generating human disease models. Here, we present an overview of applying iPSCs in developing cellular models for understanding ASD. We also discuss future perspectives in the use of iPSCs as a source of cell therapy and as a screening platform for identifying small molecules with efficacy for alleviating ASD.
Collapse
Affiliation(s)
- Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, 10 Amistad, 201B, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
331
|
West EL, Gonzalez-Cordero A, Hippert C, Osakada F, Martinez-Barbera JP, Pearson RA, Sowden JC, Takahashi M, Ali RR. Defining the integration capacity of embryonic stem cell-derived photoreceptor precursors. Stem Cells 2012; 30:1424-35. [PMID: 22570183 PMCID: PMC3580313 DOI: 10.1002/stem.1123] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Retinal degeneration is a leading cause of irreversible blindness in the developed world. Differentiation of retinal cells, including photoreceptors, from both mouse and human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), potentially provide a renewable source of cells for retinal transplantation. Previously, we have shown both the functional integration of transplanted rod photoreceptor precursors, isolated from the postnatal retina, in the adult murine retina, and photoreceptor cell generation by stepwise treatment of ESCs with defined factors. In this study, we assessed the extent to which this protocol recapitulates retinal development and also evaluated differentiation and integration of ESC-derived retinal cells following transplantation using our established procedures. Optimized retinal differentiation via isolation of Rax.GFP retinal progenitors recreated a retinal niche and increased the yield of Crx(+) and Rhodopsin(+) photoreceptors. Rod birth peaked at day 20 of culture and expression of the early photoreceptor markers Crx and Nrl increased until day 28. Nrl levels were low in ESC-derived populations compared with developing retinae. Transplantation of early stage retinal cultures produced large tumors, which were avoided by prolonged retinal differentiation (up to day 28) prior to transplantation. Integrated mature photoreceptors were not observed in the adult retina, even when more than 60% of transplanted ESC-derived cells expressed Crx. We conclude that exclusion of proliferative cells from ESC-derived cultures is essential for effective transplantation. Despite showing expression profiles characteristic of immature photoreceptors, the ESC-derived precursors generated using this protocol did not display transplantation competence equivalent to precursors from the postnatal retina.
Collapse
Affiliation(s)
- Emma L West
- Department of Genetics, UCL Institute of Ophthalmology, University College London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
332
|
Jasty S, Srinivasan P, Pasricha G, Chatterjee N, Subramanian K. Gene Expression Profiles and Retinal Potential of Stem/Progenitor Cells Derived from Human Iris and Ciliary Pigment Epithelium. Stem Cell Rev Rep 2012; 8:1163-77. [DOI: 10.1007/s12015-012-9394-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
333
|
Ong JM, da Cruz L. A review and update on the current status of stem cell therapy and the retina. Br Med Bull 2012; 102:133-46. [PMID: 22577179 DOI: 10.1093/bmb/lds013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION OR BACKGROUND Many diseases of the retina result in irreversible visual loss. Stem cell (SC) therapy is a rapidly developing field and represents a novel approach to replace non-functioning neuro-retinal cells. SOURCES OF DATA A systematic computerized literature search was conducted on PubMed (http://www.ncbi.nlm.nih.gov/pubmed/). AREAS OF AGREEMENT The use of stem cells (SCs) in animal models of retinal diseases has resulted in improvement in visual function and performance. SC therapy represents an exciting prospect in restoring vision. Areas of controversy The use of human embryonic SCs raises ethical concerns. GROWING POINTS Human trials using SCs in retinal diseases have recently been approved. AREAS TIMELY FOR DEVELOPING RESEARCH The success of SCs in retinal therapy depends not only on implanted cell survival, but also on how well SCs migrate, integrate and form synapses. Further research will be needed to overcome these hurdles.
Collapse
Affiliation(s)
- J M Ong
- National Institute of Health Research, Biomedical Research Centre, Moorfields Eye Hospital, London, UK.
| | | |
Collapse
|
334
|
|
335
|
Bae D, Mondragon-Teran P, Hernandez D, Ruban L, Mason C, Bhattacharya SS, Veraitch FS. Hypoxia Enhances the Generation of Retinal Progenitor Cells from Human Induced Pluripotent and Embryonic Stem Cells. Stem Cells Dev 2012; 21:1344-55. [DOI: 10.1089/scd.2011.0225] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Daekyeong Bae
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Paul Mondragon-Teran
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Diana Hernandez
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Ludmila Ruban
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Chris Mason
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Shomi S. Bhattacharya
- CABIMER, Isla de la Cartuja, Sevilla, Spain
- UCL-Institute of Ophthalmology, London, United Kingdom
| | - Farlan S. Veraitch
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
336
|
Clarke L, Ballios BG, van der Kooy D. Generation and clonal isolation of retinal stem cells from human embryonic stem cells. Eur J Neurosci 2012; 36:1951-9. [PMID: 22591375 DOI: 10.1111/j.1460-9568.2012.08123.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Retinal stem cells (RSCs) are present within the pigmented ciliary epithelium (CE) of the adult human eye and produce progeny that differentiate in vitro into all neural retinal subtypes and retinal pigmented epithelium (RPE). We hypothesized that a RSC population, similar to the adult CE-derived RSC, is contained within pigmented colonies that arise in long-term cultures of human embryonic stem cells (hESCs) suggested to recapitulate retinal development in vitro. Single pigmented hESC-derived cells were isolated and plated in serum-free media containing growth factors and, after 2 weeks, clonal sphere colonies containing both pigmented and non-pigmented cells were observed. These colonies expressed the early retinal transcription factors Rx, Chx10 and Pax6, and could be dissociated and replated as single cells to form secondary clonal colonies. When allowed to differentiate, expression of markers for both RPE and neurons was observed. Rhodopsin expression was detected after explant co-culture and transplantation into the developing mouse eye as well as following treatment with soluble factors in vitro. We show that RSCs emerge in an in vitro model of retinal development and are a potential source of human photoreceptors for use in transplantation.
Collapse
Affiliation(s)
- Laura Clarke
- Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, ON, Canada.
| | | | | |
Collapse
|
337
|
Cai S, Smith ME, Redenti SM, Wnek GE, Young MJ. Mouse retinal progenitor cell dynamics on electrospun poly (ϵ-caprolactone). JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 23:1451-65. [PMID: 21781383 DOI: 10.1163/092050611x584388] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Age-related macular degeneration, retinitis pigmentosa and glaucoma are among the many retinal degenerative diseases where retinal cell death leads to irreversible vision loss and blindness. Working toward a cell-replacement-based therapy for such diseases, a number of research groups have recently evaluated the feasibility of using retinal progenitor cells (RPCs) cultured and transplanted on biodegradable polymer substrates to replace damaged retinal tissue. Appropriate polymer substrate design is essential to providing a three-dimensional environment that can facilitate cell adhesion, proliferation and post-transplantation migration into the host environment. In this study, we have designed and fabricated a novel, ultra-thin electrospun poly(ϵ-caprolactone) (PCL) scaffold with microscale fiber diameters, appropriate porosity for infiltration by RPCs, and biologically compatible mechanical characteristics. We have verified that our electrospun PCL scaffold supports robust mouse RPC proliferation, adhesion, and differentiation in vitro, as well as migration into mouse retinal explants. These promising results make PCL a strong candidate for further development as a cell transplantation substrate in retinal regenerative research.
Collapse
Affiliation(s)
- Sophie Cai
- a Department of Ophthalmology , Schepens Eye Research Institute, Harvard Medical School , 20 Staniford Street , Boston , MA , 02114 , USA
| | | | | | | | | |
Collapse
|
338
|
|
339
|
Derivation of human differential photoreceptor-like cells from the iris by defined combinations of CRX, RX and NEUROD. PLoS One 2012; 7:e35611. [PMID: 22558175 PMCID: PMC3338414 DOI: 10.1371/journal.pone.0035611] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 03/19/2012] [Indexed: 12/14/2022] Open
Abstract
Examples of direct differentiation by defined transcription factors have been provided for beta-cells, cardiomyocytes and neurons. In the human visual system, there are four kinds of photoreceptors in the retina. Neural retina and iris-pigmented epithelium (IPE) share a common developmental origin, leading us to test whether human iris cells could differentiate to retinal neurons. We here define the transcription factor combinations that can determine human photoreceptor cell fate. Expression of rhodopsin, blue opsin and green/red opsin in induced photoreceptor cells were dependent on combinations of transcription factors: A combination of CRX and NEUROD induced rhodopsin and blue opsin, but did not induce green opsin; a combination of CRX and RX induced blue opsin and green/red opsin, but did not induce rhodopsin. Phototransduction-related genes as well as opsin genes were up-regulated in those cells. Functional analysis; i.e. patch clamp recordings, clearly revealed that generated photoreceptor cells, induced by CRX, RX and NEUROD, responded to light. The response was an inward current instead of the typical outward current. These data suggest that photosensitive photoreceptor cells can be generated by combinations of transcription factors. The combination of CRX and RX generate immature photoreceptors: and additional NEUROD promotes maturation. These findings contribute substantially to a major advance toward eventual cell-based therapy for retinal degenerative diseases.
Collapse
|
340
|
Mandai M, Homma K, Okamoto S, Yamada C, Nomori A, Takahashi M. Adequate Time Window and Environmental Factors Supporting Retinal Graft Cell Survival in rd Mice. CELL MEDICINE 2012; 4:45-54. [PMID: 26858854 DOI: 10.3727/215517912x639315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Postnatal photoreceptor cells can be integrated into the wild-type adult retina in mice, and retinal transplantation is now one therapeutic option for retinal degenerative diseases when photoreceptor degeneration is the primary cause of the disease. The aim of this study was to specify the optimal time window during the course of retinal degeneration and to modulate the host and/or graft environment for a successful transplantation. We first studied the background features of the mice with phosphodiesterase 6b (PDE6b) gene mutation (rd; C3H/Hej) and found that the infiltration of microglia and glial fibrillary acidic protein (GFAP) expression once increased at the peak of rod death (∼2-3 weeks of age) but then reduced for a following period until gliosis began to take place with enhanced GFAP expression (∼8 weeks of age). The postnatal retinal cells (p4-p7) were successfully transplanted during this period with neurite extension into the host retina. In later transplantations (6 or 8 weeks of age), graft cells survived better in the presence of chondroitinase ABC (ChABC), which digests chondroitin sulfate proteoglycan (CSPG), an essential component of gliosis. In contrast, in earlier transplantations (4 weeks of age), graft cells survived better in the presence of valproic acid (VPA), a neural differentiating reagent, or glatiramer acetate, an immune modulator. These suggest that, immediately after the outer nuclear layer (ONL) degeneration, an inflammatory reaction may be easily induced but the host neurons may be more able to accept donor cells in the presence of neural differentiating factor. These results will help optimize transplantation conditions when we consider clinical application.
Collapse
Affiliation(s)
- Michiko Mandai
- Laboratory for Retinal Regeneration , Riken Kobe CDB, Kobe, Hyogo , Japan
| | - Kohei Homma
- Laboratory for Retinal Regeneration , Riken Kobe CDB, Kobe, Hyogo , Japan
| | - Satoshi Okamoto
- Laboratory for Retinal Regeneration , Riken Kobe CDB, Kobe, Hyogo , Japan
| | - Chikako Yamada
- Laboratory for Retinal Regeneration , Riken Kobe CDB, Kobe, Hyogo , Japan
| | - Akane Nomori
- Laboratory for Retinal Regeneration , Riken Kobe CDB, Kobe, Hyogo , Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration , Riken Kobe CDB, Kobe, Hyogo , Japan
| |
Collapse
|
341
|
Phillips MJ, Wallace KA, Dickerson SJ, Miller MJ, Verhoeven AD, Martin JM, Wright LS, Shen W, Capowski EE, Percin EF, Perez ET, Zhong X, Canto-Soler MV, Gamm DM. Blood-derived human iPS cells generate optic vesicle-like structures with the capacity to form retinal laminae and develop synapses. Invest Ophthalmol Vis Sci 2012; 53:2007-19. [PMID: 22410558 DOI: 10.1167/iovs.11-9313] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE We sought to determine if human induced pluripotent stem cells (iPSCs) derived from blood could produce optic vesicle-like structures (OVs) with the capacity to stratify and express markers of intercellular communication. METHODS Activated T-lymphocytes from a routine peripheral blood sample were reprogrammed by retroviral transduction to iPSCs. The T-lymphocyte-derived iPSCs (TiPSCs) were characterized for pluripotency and differentiated to OVs using our previously published protocol. TiPSC-OVs were then manually isolated, pooled, and cultured en masse to more mature stages of retinogenesis. Throughout this stepwise differentiation process, changes in anterior neural, retinal, and synaptic marker expression were monitored by PCR, immunocytochemistry, and/or flow cytometry. RESULTS TiPSCs generated abundant OVs, which contained a near homogeneous population of proliferating neuroretinal progenitor cells (NRPCs). These NRPCs differentiated into multiple neuroretinal cell types, similar to OV cultures from human embryonic stem cells and fibroblast-derived iPSCs. In addition, portions of some TiPSC-OVs maintained their distinctive neuroepithelial appearance and spontaneously formed primitive laminae, reminiscent of the developing retina. Retinal progeny from TiPSC-OV cultures expressed numerous genes and proteins critical for synaptogenesis and gap junction formation, concomitant with the emergence of glia and the upregulation of thrombospondins in culture. CONCLUSIONS We demonstrate for the first time that human blood-derived iPSCs can generate retinal cell types, providing a highly convenient donor cell source for iPSC-based retinal studies. We also show that cultured TiPSC-OVs have the capacity to self-assemble into rudimentary neuroretinal structures and express markers indicative of chemical and electrical synapses.
Collapse
Affiliation(s)
- M Joseph Phillips
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
342
|
Hambright D, Park KY, Brooks M, McKay R, Swaroop A, Nasonkin IO. Long-term survival and differentiation of retinal neurons derived from human embryonic stem cell lines in un-immunosuppressed mouse retina. Mol Vis 2012; 18:920-36. [PMID: 22539871 PMCID: PMC3335781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 04/08/2012] [Indexed: 10/28/2022] Open
Abstract
PURPOSE To examine the potential of NIH-maintained human embryonic stem cell (hESC) lines TE03 and UC06 to differentiate into retinal progenitor cells (hESC-RPCs) using the noggin/Dkk-1/IGF-1/FGF9 protocol. An additional goal is to examine the in vivo dynamics of maturation and retinal integration of subretinal and epiretinal (vitreous space) hESC-RPC grafts without immunosuppression. METHODS hESCs were neuralized in vitro with noggin for 2 weeks and expanded to derive neuroepithelial cells (hESC-neural precursors, NPs). Wnt (Integration 1 and wingless) blocking morphogens Dickkopf-1 (Dkk-1) and Insulin-like growth factor 1 (IGF-1) were used to direct NPs to a rostral neural fate, and fibroblast growth factor 9 (FGF9)/fibroblast growth factor-basic (bFGF) were added to bias the differentiation of developing anterior neuroectoderm cells to neural retina (NR) rather than retinal pigment epithelium (RPE). Cells were dissociated and grafted into the subretinal and epiretinal space of young adult (4-6-week-old) mice (C57BL/6J x129/Sv mixed background). Remaining cells were replated for (i) immunocytochemical analysis and (ii) used for quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis. Mice were sacrificed 3 weeks or 3 months after grafting, and the grafts were examined by histology and immunohistochemistry for survival of hESC-RPCs, presence of mature neuronal and retinal markers, and the dynamics of in vivo maturation and integration into the host retina. RESULTS At the time of grafting, hESC-RPCs exhibited immature neural/neuronal immunophenotypes represented by nestin and neuronal class III β-tubulin, with about half of the cells positive for cell proliferation marker Kiel University -raised antibody number 67 (Ki67), and no recoverin-positive (recoverin [+]) cells. The grafted cells expressed eye field markers paired box 6 (PAX6), retina and anterior neural fold homeobox (RAX), sine oculis homeobox homolog 6 (SIX6), LIM homeobox 2 (LHX2), early NR markers (Ceh-10 homeodomain containing homolog [CHX10], achaete-scute complex homolog 1 [MASH1], mouse atonal homolog 5 [MATH5], neurogenic differentiation 1 [NEUROD1]), and some retinal cell fate markers (brain-specific homeobox/POU domain transcription factor 3B [BRN3B], prospero homeobox 1 [PROX1], and recoverin). The cells in the subretinal grafts matured to predominantly recoverin [+] phenotype by 3 months and survived in a xenogenic environment without immunosuppression as long as the blood-retinal barrier was not breached by the transplantation procedure. The epiretinal grafts survived but did not express markers of mature retinal cells. Retinal integration into the retinal ganglion cell (RGC) layer and the inner nuclear layer (INL) was efficient from the epiretinal but not subretinal grafts. The subretinal grafts showed limited ability to structurally integrate into the host retina and only in cases when NR was damaged during grafting. Only limited synaptogenesis and no tumorigenicity was observed in grafts. CONCLUSIONS Our studies show that (i) immunosuppression is not mandatory to xenogenic graft survival in the retina, (ii) the subretinal but not the epiretinal niche can promote maturation of hESC-RPCs to photoreceptors, and (iii) the hESC-RPCs from epiretinal but not subretinal grafts can efficiently integrate into the RGC layer and INL. The latter could be of value for long-lasting neuroprotection of retina in some degenerative conditions and glaucoma. Overall, our results provide new insights into the technical aspects associated with cell-based therapy in the retina.
Collapse
Affiliation(s)
- Dustin Hambright
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Kye-Yoon Park
- Laboratory of Molecular Biology and NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Matthew Brooks
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Ron McKay
- Laboratory of Molecular Biology and NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Igor O. Nasonkin
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
343
|
Sanie-Jahromi F, Ahmadieh H, Soheili ZS, Davari M, Ghaderi S, Kanavi MR, Samiei S, Deezagi A, Pakravesh J, Bagheri A. Enhanced generation of retinal progenitor cells from human retinal pigment epithelial cells induced by amniotic fluid. BMC Res Notes 2012; 5:182. [PMID: 22490806 PMCID: PMC3428660 DOI: 10.1186/1756-0500-5-182] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 04/04/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Retinal progenitor cells are a convenient source of cell replacement therapy in retinal degenerative disorders. The purpose of this study was to evaluate the expression patterns of the homeobox genes PAX6 and CHX10 (retinal progenitor markers) during treatment of human retinal pigment epithelium (RPE) cells with amniotic fluid (AF), RPE cells harvested from neonatal cadaver globes were cultured in a mixture of DMEM and Ham's F12 supplemented with 10% FBS. At different passages, cells were trypsinized and co-cultured with 30% AF obtained from normal fetuses of 1416 weeks gestational age. RESULTS Compared to FBS-treated controls, AF-treated cultures exhibited special morphological changes in culture, including appearance of spheroid colonies, improved initial cell adhesion and ordered cell alignment. Cell proliferation assays indicated a remarkable increase in the proliferation rate of RPE cells cultivated in 30% AF-supplemented medium, compared with those grown in the absence of AF. Immunocytochemical analyses exhibited nuclear localization of retinal progenitor markers at a ratio of 33% and 27% for CHX10 and PAX6, respectively. This indicated a 3-fold increase in retinal progenitor markers in AF-treated cultures compared to FBS-treated controls. Real-time PCR data of retinal progenitor genes (PAX6, CHX10 and VSX-1) confirmed these results and demonstrated AF's capacity for promoting retinal progenitor cell generation. CONCLUSION Taken together, the results suggest that AF significantly promotes the rate of retinal progenitor cell generation, indicating that AF can be used as an enriched supplement for serum-free media used for the in vitro propagation of human progenitor cells.
Collapse
|
344
|
Jarrett SG, Boulton ME. Consequences of oxidative stress in age-related macular degeneration. Mol Aspects Med 2012; 33:399-417. [PMID: 22510306 DOI: 10.1016/j.mam.2012.03.009] [Citation(s) in RCA: 379] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 03/31/2012] [Indexed: 12/24/2022]
Abstract
The retina resides in an environment that is primed for the generation of reactive oxygen species (ROS) and resultant oxidative damage. The retina is one of the highest oxygen-consuming tissues in the human body. The highest oxygen levels are found in the choroid, but this falls dramatically across the outermost retina, creating a large gradient of oxygen towards the retina and inner segments of the photoreceptors which contain high levels of polyunsaturated fatty acids. This micro-environment together with abundant photosensitizers, visible light exposure and a high energy demand supports a highly oxidative milieu. However, oxidative damage is normally minimized by the presence of a range of antioxidant and efficient repair systems. Unfortunately, as we age oxidative damage increases, antioxidant capacity decreases and the efficiency of reparative systems become impaired. The result is retinal dysfunction and cell loss leading to visual impairment. It appears that these age-related oxidative changes are a hallmark of early age-related macular degeneration (AMD) which, in combination with hereditary susceptibility and other retinal modifiers, can progress to the pathology and visual morbidity associated with advanced AMD. This review reassesses the consequences of oxidative stress in AMD and strategies for preventing or reversing oxidative damage in retinal tissues.
Collapse
Affiliation(s)
- Stuart G Jarrett
- Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
345
|
Photoreceptor Differentiation following Transplantation of Allogeneic Retinal Progenitor Cells to the Dystrophic Rhodopsin Pro347Leu Transgenic Pig. Stem Cells Int 2012; 2012:939801. [PMID: 22567027 PMCID: PMC3337587 DOI: 10.1155/2012/939801] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 01/26/2012] [Indexed: 11/24/2022] Open
Abstract
Purpose. Transplantation of stem, progenitor, or precursor cells has resulted in photoreceptor replacement and evidence of functional efficacy in rodent models of retinal degeneration. Ongoing work has been directed toward the replication of these results in a large animal model, namely, the pig. Methods. Retinal progenitor cells were derived from the neural retina of GFP-transgenic pigs and transplanted to the subretinal space of rhodopsin Pro347Leu-transgenic allorecipients, in the early stage of the degeneration and the absence of immune suppression. Results. Results confirm the survival of allogeneic porcine RPCs without immune suppression in the setting of photoreceptor dystrophy. The expression of multiple photoreceptor markers by grafted cells included the rod outer segment-specific marker ROM-1. Further evidence of photoreceptor differentiation included the presence of numerous photoreceptor rosettes within GFP-positive grafts, indicative of the development of cellular polarity and self-assembly into rudiments of outer retinal tissue. Conclusion. Together, these data support the tolerance of RPCs as allografts and demonstrate the high level of rod photoreceptor development that can be obtained from cultured RPCs following transplantation. Strategies for further progress in this area, together with possible functional implications, are discussed.
Collapse
|
346
|
Doonan F, Cotter TG. Norgestrel may be a potential therapy for retinal degenerations. Expert Opin Investig Drugs 2012; 21:579-81. [DOI: 10.1517/13543784.2012.667400] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Francesca Doonan
- University College Cork, Biosciences Research Institute, Biochemistry Department, Cell Development and Disease Laboratory,
Cork, Ireland ;
| | - Thomas G Cotter
- University College Cork, Biosciences Research Institute, Biochemistry Department, Cell Development and Disease Laboratory,
Cork, Ireland ;
| |
Collapse
|
347
|
Schmeer CW, Wohl SG, Isenmann S. Cell-replacement therapy and neural repair in the retina. Cell Tissue Res 2012; 349:363-74. [PMID: 22354517 DOI: 10.1007/s00441-012-1335-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 01/18/2012] [Indexed: 01/12/2023]
Abstract
Visual impairment severely affects the quality of life of patients and their families and is also associated with a deep economic impact. The most common pathologies responsible for visual impairment and legally defined blindness in developed countries include age-related macular degeneration, glaucoma and diabetic retinopathy. These conditions share common pathophysiological features: dysfunction and loss of retinal neurons. To date, two main approaches are being taken to develop putative therapeutic strategies: neuroprotection and cell replacement. Cell replacement is a novel therapeutic approach to restore visual capabilities to the degenerated adult neural retina and represents an emerging field of regenerative neurotherapy. The discovery of a population of proliferative cells in the mammalian retina has raised the possibility of harnessing endogenous retinal stem cells to elicit retinal repair. Furthermore, the development of suitable protocols for the reprogramming of differentiated somatic cells to a pluripotent state further increases the therapeutic potential of stem-cell-based technologies for the treatment of major retinal diseases. Stem-cell transplantation in animal models has been most effectively used for the replacement of photoreceptors, although this therapeutic approach is also being used for inner retinal pathologies. In this review, we discuss recent advances in the development of cell-replacement approaches for the treatment of currently incurable degenerative retinal diseases.
Collapse
Affiliation(s)
- Christian W Schmeer
- Hans Berger Clinic of Neurology, University Hospital Jena, Erlanger Allee 101, 07747 Jena, Germany.
| | | | | |
Collapse
|
348
|
Rowland TJ, Buchholz DE, Clegg DO. Pluripotent human stem cells for the treatment of retinal disease. J Cell Physiol 2012; 227:457-66. [PMID: 21520078 DOI: 10.1002/jcp.22814] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Despite advancements made in our understanding of ocular biology, therapeutic options for many debilitating retinal diseases remain limited. Stem cell-based therapies are a potential avenue for treatment of retinal disease, and this mini-review will focus on current research in this area. Cellular therapies to replace retinal pigmented epithelium (RPE) and/or photoreceptors to treat age-related macular degeneration (AMD), Stargardt's macular dystrophy, and retinitis pigmentosa are currently being developed. Over the past decade, significant advancements have been made using different types of human stem cells with varying capacities to differentiate into these target retinal cell types. We review and evaluate pluripotent stem cells, both human embryonic stem cells and human induced pluripotent stem cells, as well as protocols for differentiation of ocular cells, and culture and transplant techniques that might be used to deliver cells to patients.
Collapse
Affiliation(s)
- Teisha J Rowland
- Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, California, USA
| | | | | |
Collapse
|
349
|
RPE-secreted factors: influence differentiation in human retinal cell line in dose- and density-dependent manner. J Ocul Biol Dis Infor 2012; 3:144-60. [PMID: 23316262 DOI: 10.1007/s12177-011-9076-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 12/29/2011] [Indexed: 12/28/2022] Open
Abstract
Retinal pigment epithelial (RPE) cells play an important role in normal functioning of retina and photoreceptors, and some retinal degenerations arise due to malfunctioning RPE. Retinal pigment epithelium transplantation is being explored as a strategy to rescue degenerating photoreceptors in diseases such as age-related macular degeneration (AMD) and retinitis pigmentosa (RP). Additionally, RPE-secreted factors could rescue degenerating photoreceptors by prolonging survival or by their ability to differentiate and give rise to photoreceptors by transdifferentiation. In this study, we have explored what role cell density could play in differentiation induced in a human retinal progenitor cell line, in response to RPE-secreted growth factors. Retinal progenitors plated at low (1 × 10(4) cells/cm(2)), medium (2-4 × 10(4) cells/cm(2)), and high (1 × 10(5) cells/cm(2)) cell density were exposed to various dilutions of RPE-conditioned medium (secreted factors) under conditions of defined medium culture. Progenitor cell differentiation was monitored phenotypically (morphological, biochemical analysis, and immunophenotyping, and western blot analysis were performed). Our data show that differentiation in response to RPE-secreted factors is modulated by cell density and dilutions of conditioned medium. We conclude that before embarking on RPE transplantation as a modality for treatment of RP and AMD, one will have to determine the role that cell density and inhibitory and stimulatory neurotrophins secreted by RPE could play in the efficacy of survival of transplants. We report that RPE-conditioned medium enhances neuronal phenotype (photoreceptors, bipolars) at the lowest cell density in the absence of cell-cell contact. Eighty percent to 90% of progenitor cells differentiate into photoreceptors and bipolars at 50% concentration of conditioned medium, while exposure to 100% conditioned medium might increase multipolar neurons (ganglionic and amacrine phenotypes) to a small degree. However, no clear-cut pattern of differentiation in response to RPE-secreted factors is noted at higher cell densities.
Collapse
|
350
|
La Torre A, Lamba DA, Jayabalu A, Reh TA. Production and transplantation of retinal cells from human and mouse embryonic stem cells. Methods Mol Biol 2012; 884:229-46. [PMID: 22688710 DOI: 10.1007/978-1-61779-848-1_16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Over the last few years, numerous studies have introduced strategies for the generation of neuronal populations from embryonic stem cells. These techniques are valuable both in the study of early neurogenesis and in the generation of an unlimited source of donor cells for replacement therapies. We have developed a protocol to direct mouse and human embryonic stem cells to retinal fates by using the current model of eye specification. Our method is a multistep protocol in which the cultures are treated with IGF1 and a combination of BMP and Wnt inhibitors to promote the expression of key retinal progenitor genes, as assayed by RT-PCR and immunofluorescence microscopy. The retinal progenitor population spontaneously undergoes differentiation towards various types of retinal neurons, including photoreceptors.
Collapse
Affiliation(s)
- Anna La Torre
- Department of Biological Structure, Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|