1
|
Kosior-Jarecka E, Grzybowski A. Retinal Ganglion Cell Replacement in Glaucoma Therapy: A Narrative Review. J Clin Med 2024; 13:7204. [PMID: 39685661 DOI: 10.3390/jcm13237204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide. It leads to the progressive degeneration of retinal ganglion cells (RGCs), the axons of which form the optic nerve. Enormous RGC apoptosis causes a lack of transfer of visual information to the brain. The RGC loss typical of the central nervous system is irreversible, and when glaucoma progresses, the total amount of RGCs in the retina enormously diminishes. The successful treatment in glaucoma patients is a direct neuroprotection by decreasing the intraocular pressure, which enables RGC protection but does not revive the lost ones. The intriguing new therapy for advanced glaucoma is the possibility of RGC replacement with new healthy cells. In this review article, the strategies regarding RGC replacement therapy are presented with the latest advances in the technique and the obstacles that it meets.
Collapse
Affiliation(s)
- Ewa Kosior-Jarecka
- Department of Diagnostics and Microsurgery of Glaucoma, Medical University of Lublin, 20-079 Lublin, Poland
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, 60-836 Poznan, Poland
| |
Collapse
|
2
|
Jong ED, Hacibekiroglu S, Guo L, Sawula E, Li B, Li C, Ho MT, Shoichet MS, Wallace VA, Nagy A. Soluble CX3CL1-expressing retinal pigment epithelium cells protect rod photoreceptors in a mouse model of retinitis pigmentosa. Stem Cell Res Ther 2023; 14:212. [PMID: 37605279 PMCID: PMC10441732 DOI: 10.1186/s13287-023-03434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Retinitis pigmentosa (RP) is an inherited retinal disease that results in photoreceptor degeneration, leading to severe vision loss or blindness. Due to its genetic heterogeneity, developing a new gene therapy to correct every genetic mutation contributing to its progression is infeasible. Photoreceptor transplantation can be harnessed to restore vision; however, this approach is limited by poor cell survival and synaptic integration into the neural retina. Thus, we developed a combined cell and gene therapy that is expected to protect photoreceptors in most, if not all, cases of RP. METHODS Human embryonic stem cells (hESCs) modified with our FailSafe™ system were genetically engineered to overexpress sCX3CL1, an inhibitor of microglia activation that has been shown to preserve photoreceptor survival and function in mouse models of RP, independent of the genetic cause. These cells were differentiated into human retinal pigment epithelium (hRPE) cells and used as therapeutic cells due to their longevity and safety, both of which have been demonstrated in preclinical and clinical studies. Transgenic hRPE were delivered into the subretinal space of immunodeficient mice and the rd10 mouse model of RP to evaluate donor cell survival and retention of transgene expression. The outer nuclear layer was quantified to assess photoreceptor protection. RESULTS Transgenic FailSafe™ hRPE (FS-hRPE) cells can survive for at least four months in the retina of immunodeficient mice and retain transgene expression. However, these cells do not persist beyond two weeks post-injection in the retina of immunocompetent rd10 recipients, despite Cyclosporine A treatment. Nevertheless, sCX3CL1-expressing FailSafe™ hRPE cells prevented photoreceptor degeneration in a local acting manner during the duration of their presence in the subretinal space. CONCLUSIONS Transgenic hESCs differentiate into hRPE cells and retain sCX3CL1 transgene expression both in vitro and in vivo. Moreover, hRPE cells delivered to the subretinal space of rd10 mice prevented photoreceptor degeneration in a local-acting manner, suggesting that this approach could have applications for preserving photoreceptors in specific subregions of the retina, such as the macula. Overall, our study not only reveals the potential of a combined cell and gene therapy for the treatment of RP, but also the possibility of using hRPE cells to deliver therapeutic biologics in situ to treat diseases over long-term.
Collapse
Affiliation(s)
- Eric D Jong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sabiha Hacibekiroglu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Lily Guo
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Evan Sawula
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Biao Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Chengjin Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Margaret T Ho
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Canada
| | - Molly S Shoichet
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
- Department of Chemistry, University of Toronto, Toronto, Canada
| | - Valerie A Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada.
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Maidana DE, Gonzalez-Buendia L, Miller JW, Vavvas DG. RIPK necrotic cell death pathway in both donor photoreceptor and host immune cells synergize to affect photoreceptor graft survival. FASEB J 2023; 37:e22847. [PMID: 36862516 PMCID: PMC10590064 DOI: 10.1096/fj.202201137r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/29/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
Photoreceptor transplant has been put forward as a repair strategy to tackle degenerated retinas. Nonetheless, cell death and immune rejection seriously limit the success of this strategy, with only a small fraction of transplanted cells surviving. Improving the survival of transplanted cells is of critical importance. Recent evidence has identified receptor-interacting protein kinase 3 (RIPK3) as a molecular trigger controlling necroptotic cell death and inflammation. However, its role in photoreceptor transplantation and regenerative medicine has not been studied. We hypothesized that modulation of RIPK3 to address both cell death and immunity could have advantageous effects on photoreceptor survival. In a model of inherited retinal degeneration, deletion of RIPK3 in donor photoreceptor precursors significantly increases the survival of transplanted cells. Simultaneous RIPK3 deletion in donor photoreceptors and recipients maximizes graft survival. Lastly, to discern the role of RIPK3 in the host immune response, bone marrow transplant experiments demonstrated that peripheral immune cell RIPK3 deficiency is protective for both donor and host photoreceptor survival. Interestingly, this finding is independent of photoreceptor transplantation, as the peripheral protective effect is also observed in an additional retinal detachment photoreceptor degeneration model. Altogether, these results indicate that immunomodulatory and neuroprotective strategies targeting the RIPK3 pathway can aid regenerative therapies of photoreceptor transplantation.
Collapse
Affiliation(s)
- Daniel E. Maidana
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lucia Gonzalez-Buendia
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Joan W. Miller
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
4
|
He XY, Zhao CJ, Xu H, Chen K, Bian BSJ, Gong Y, Weng CH, Zeng YX, Fu Y, Liu Y, Yin ZQ. Synaptic repair and vision restoration in advanced degenerating eyes by transplantation of retinal progenitor cells. Stem Cell Reports 2021; 16:1805-1817. [PMID: 34214489 PMCID: PMC8282465 DOI: 10.1016/j.stemcr.2021.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022] Open
Abstract
Stem cell transplantation shows enormous potential for treatment of incurable retinal degeneration (RD). To determine if and how grafts connect with the neural circuits of the advanced degenerative retina (ADR) and improve vision, we perform calcium imaging of GCaMP5-positive grafts in retinal slices. The organoid-derived C-Kit+/SSEA1- (C-Kit+) retinal progenitor cells (RPCs) become synaptically organized and build spontaneously active synaptic networks in three major layers of ADR. Light stimulation of the host photoreceptors elicits distinct neuronal responses throughout the graft RPCs. The graft RPCs and their differentiated offspring cells in inner nuclear layer synchronize their activities with the host cells and exhibit presynaptic calcium flux patterns that resemble intact retinal neurons. Once graft-to-host network is established, progressive vision loss is stabilized while control eyes continually lose vision. Therefore, transplantation of organoid-derived C-Kit+ RPCs can form functional synaptic networks within ADR and it holds promising avenue for advanced RD treatment.
Collapse
Affiliation(s)
- Xiang-Yu He
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Department of Ophthalmology, the 958th Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China; Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China
| | - Cong-Jian Zhao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Kang Chen
- Department of Ophthalmology, the 958th Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Bai-Shi-Jiao Bian
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yu Gong
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Chuan-Huang Weng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yu-Xiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yan Fu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yong Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China.
| | - Zheng-Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China; Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China.
| |
Collapse
|
5
|
Ma J, Chen M, Ai J, Young MJ, Ge J. Enhanced migration of engrafted retinal progenitor cells into the host retina via disruption of glial barriers. Mol Vis 2021; 27:300-308. [PMID: 34035644 PMCID: PMC8131176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/08/2021] [Indexed: 11/18/2022] Open
Abstract
Purpose Migration and integration remain critical challenges for stem cell replacement therapy. Glial barriers play an important role in preventing cell migration and integration. The purpose of this study was to investigate the effect and mechanisms of chondroitinase ABC on the migration of murine retinal progenitor cells (mRPCs) transplanted into the subretinal space of B6 mice. Methods mRPCs were harvested from the neural retinas of P1 enhanced green fluorescent protein (GFP) B6 mice. Two μl containing 2 × 105 expanded RPCs alone or combined with chondroitinase ABC in suspension were injected into the subretinal space of the recipient B6 mice. Immunohistochemistry was performed on the recipient B6 retinas to evaluate the glial barrier formation and migration of the mRPCs. Western blotting was also used to check the expression of the glial barriers. Results Glial fibrillary acidic protein (GFAP) and vimentin could be seen around the transplanted mRPCs in the B6 mice. Formation of glial barriers prevented the migration of donor cells into the retinal layers. Chondroitinase ABC promoted the migration and survival rates of the engrafted retinal progenitor cells in the retinal layers of recipient B6 mice. Injection induced upregulation of GFAP, chondroitin, and CD44 expression. Chondroitinase ABC disrupted the glial barriers. The CD44 around the mRPCs was much lower in the chondroitinase group. However, the CD44 in the retinal layers was considerably higher in the chondroitinase group. With the employment of chondroitinase ABC, more cells migrated into the outer nuclear layer or inner nuclear layer. The chondroitin and CD44 expression decreased 3 weeks after transplantation in the chondroitinase ABC group. Conclusions Chondroitinase ABC degraded glial barriers and enhanced the migration of transplanted mouse retinal progenitor cells. Chondroitinase ABC may also have induced activation of the CD44 signaling pathway to exert the effect.
Collapse
Affiliation(s)
- Jian Ma
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Min Chen
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Jing Ai
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Michael J. Young
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Fathi M, Ross CT, Hosseinzadeh Z. Functional 3-Dimensional Retinal Organoids: Technological Progress and Existing Challenges. Front Neurosci 2021; 15:668857. [PMID: 33958988 PMCID: PMC8095320 DOI: 10.3389/fnins.2021.668857] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
Stem cell scientists have developed methods for the self-formation of artificial organs, often referred to as organoids. Organoids can be used as model systems for research in multiple biological disciplines. Yoshiki Sasai’s innovation for deriving mammalian retinal tissue from in vitro stem cells has had a large impact on the study of the biology of vision. New developments in retinal organoid technology provide avenues for in vitro models of human retinal diseases, studies of pathological mechanisms, and development of therapies for retinal degeneration, including electronic retinal implants and gene therapy. Moreover, these innovations have played key roles in establishing models for large-scale drug screening, studying the stages of retinal development, and providing a human model for personalized therapeutic approaches, like cell transplants to replace degenerated retinal cells. Here, we first discuss the importance of human retinal organoids to the biomedical sciences. Then, we review various functional features of retinal organoids that have been developed. Finally, we highlight the current limitations of retinal organoid technologies.
Collapse
Affiliation(s)
- Meimanat Fathi
- Department of Cell Techniques and Applied Stem Cell Biology, Faculty of Medicine, Center for Biotechnology and Biomedicine (BBZ), University of Leipzig, Leipzig, Germany.,Physiology and Pathophysiology of the Retina Group, Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Cody T Ross
- Department of Human Behavior, Ecology and Culture, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Zohreh Hosseinzadeh
- Physiology and Pathophysiology of the Retina Group, Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
7
|
Nair DSR, Seiler MJ, Patel KH, Thomas V, Camarillo JCM, Humayun MS, Thomas BB. Tissue Engineering Strategies for Retina Regeneration. APPLIED SCIENCES-BASEL 2021; 11. [PMID: 35251703 PMCID: PMC8896578 DOI: 10.3390/app11052154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The retina is a complex and fragile photosensitive part of the central nervous system which is prone to degenerative diseases leading to permanent vision loss. No proven treatment strategies exist to treat or reverse the degenerative conditions. Recent investigations demonstrate that cell transplantation therapies to replace the dysfunctional retinal pigment epithelial (RPE) cells and or the degenerating photoreceptors (PRs) are viable options to restore vision. Pluripotent stem cells, retinal progenitor cells, and somatic stem cells are the main cell sources used for cell transplantation therapies. The success of retinal transplantation based on cell suspension injection is hindered by limited cell survival and lack of cellular integration. Recent advances in material science helped to develop strategies to grow cells as intact monolayers or as sheets on biomaterial scaffolds for transplantation into the eyes. Such implants are found to be more promising than the bolus injection approach. Tissue engineering techniques are specifically designed to construct biodegradable or non-degradable polymer scaffolds to grow cells as a monolayer and construct implantable grafts. The engineered cell construct along with the extracellular matrix formed, can hold the cells in place to enable easy survival, better integration, and improved visual function. This article reviews the advances in the use of scaffolds for transplantation studies in animal models and their application in current clinical trials.
Collapse
Affiliation(s)
- Deepthi S. Rajendran Nair
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Magdalene J. Seiler
- Departments of Physical Medicine & Rehabilitation, Ophthalmology, Anatomy & Neurobiology, Sue and Bill Gross Stem Cell Research Centre, University of California, Irvine, CA 92697-1705, USA
| | - Kahini H. Patel
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Vinoy Thomas
- Department of Physics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Juan Carlos Martinez Camarillo
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA
| | - Mark S. Humayun
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA
| | - Biju B. Thomas
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA
- Correspondence:
| |
Collapse
|
8
|
Jemni-Damer N, Guedan-Duran A, Fuentes-Andion M, Serrano-Bengoechea N, Alfageme-Lopez N, Armada-Maresca F, Guinea GV, Perez-Rigueiro J, Rojo F, Gonzalez-Nieto D, Kaplan DL, Panetsos F. Biotechnology and Biomaterial-Based Therapeutic Strategies for Age-Related Macular Degeneration. Part II: Cell and Tissue Engineering Therapies. Front Bioeng Biotechnol 2020; 8:588014. [PMID: 33363125 PMCID: PMC7758210 DOI: 10.3389/fbioe.2020.588014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Age-related Macular Degeneration (AMD) is an up-to-date untreatable chronic neurodegenerative eye disease of multifactorial origin, and the main causes of blindness in over 65 y.o. people. It is characterized by a slow progression and the presence of a multitude of factors, highlighting those related to diet, genetic heritage and environmental conditions, present throughout each of the stages of the illness. Current therapeutic approaches, mainly consisting on intraocular drug delivery, are only used for symptoms relief and/or to decelerate the progression of the disease. Furthermore, they are overly simplistic and ignore the complexity of the disease and the enormous differences in the symptomatology between patients. Due to the wide impact of the AMD and the up-to-date absence of clinical solutions, Due to the wide impact of the AMD and the up-to-date absence of clinical solutions, different treatment options have to be considered. Cell therapy is a very promising alternative to drug-based approaches for AMD treatment. Cells delivered to the affected tissue as a suspension have shown poor retention and low survival rate. A solution to these inconveniences has been the encapsulation of these cells on biomaterials, which contrive to their protection, gives them support, and favor their retention of the desired area. We offer a two-papers critical review of the available and under development AMD therapeutic approaches, from a biomaterials and biotechnological point of view. We highlight benefits and limitations and we forecast forthcoming alternatives based on novel biomaterials and biotechnology methods. In this second part we review the preclinical and clinical cell-replacement approaches aiming at the development of efficient AMD-therapies, the employed cell types, as well as the cell-encapsulation and cell-implant systems. We discuss their advantages and disadvantages and how they could improve the survival and integration of the implanted cells.
Collapse
Affiliation(s)
- Nahla Jemni-Damer
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
| | - Atocha Guedan-Duran
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - María Fuentes-Andion
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
| | - Nora Serrano-Bengoechea
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | - Nuria Alfageme-Lopez
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | | | - Gustavo V. Guinea
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - José Perez-Rigueiro
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Francisco Rojo
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Daniel Gonzalez-Nieto
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Fivos Panetsos
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| |
Collapse
|
9
|
West EL, Ribeiro J, Ali RR. Development of Stem Cell Therapies for Retinal Degeneration. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035683. [PMID: 31818854 DOI: 10.1101/cshperspect.a035683] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Degenerative retinal disease is the major cause of sight loss in the developed world and currently there is a lack of effective treatments. As the loss of vision is directly the result of the loss of retinal cells, effective cell replacement through stem-cell-based therapies may have the potential to treat a great number of retinal diseases whatever their underlying etiology. The eye is an ideal organ to develop cell therapies as it is immune privileged, and modern surgical techniques enable precise delivery of cells to the retina. Furthermore, a range of noninvasive diagnostic tests and high-resolution imaging techniques facilitate the evaluation of any therapeutic intervention. In this review, we evaluate the progress to date of current cell therapy strategies for retinal repair, focusing on transplantation of pluripotent stem-cell-derived retinal pigment epithelium (RPE) and photoreceptor cells.
Collapse
Affiliation(s)
- Emma L West
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Joana Ribeiro
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Robin R Ali
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom.,Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan 48105, USA
| |
Collapse
|
10
|
Behtaj S, Öchsner A, Anissimov YG, Rybachuk M. Retinal Tissue Bioengineering, Materials and Methods for the Treatment of Glaucoma. Tissue Eng Regen Med 2020; 17:253-269. [PMID: 32390117 PMCID: PMC7260329 DOI: 10.1007/s13770-020-00254-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Glaucoma, a characteristic type of optic nerve degeneration in the posterior pole of the eye, is a common cause of irreversible vision loss and the second leading cause of blindness worldwide. As an optic neuropathy, glaucoma is identified by increasing degeneration of retinal ganglion cells (RGCs), with consequential vision loss. Current treatments only postpone the development of retinal degeneration, and there are as yet no treatments available for this disability. Recent studies have shown that replacing lost or damaged RGCs with healthy RGCs or RGC precursors, supported by appropriately designed bio-material scaffolds, could facilitate the development and enhancement of connections to ganglion cells and optic nerve axons. The consequence may be an improved retinal regeneration. This technique could also offer the possibility for retinal regeneration in treating other forms of optic nerve ailments through RGC replacement. METHODS In this brief review, we describe the innovations and recent developments in retinal regenerative medicine such as retinal organoids and gene therapy which are specific to glaucoma treatment and focus on the selection of appropriate bio-engineering principles, biomaterials and cell therapies that are presently employed in this growing research area. RESULTS Identification of optimal sources of cells, improving cell survival, functional integration upon transplantation, and developing techniques to deliver cells into the retinal space without provoking immune responses are the main challenges in retinal cell replacement therapies. CONCLUSION The restoration of visual function in glaucoma patients by the RGC replacement therapies requires appropriate protocols and biotechnology methods. Tissue-engineered scaffolds, the generation of retinal organoids, and gene therapy may help to overcome some of the challenges in the generation of clinically safe RGCs.
Collapse
Affiliation(s)
- Sanaz Behtaj
- School of Engineering and Built Environment, Griffith University, Engineering Drive, Southport, QLD, 4222, Australia
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia
- Department of Cell and Molecular Biology, Cell Science Research Centre, Royan Institute for Biotechnology, Isfahan, Iran
| | - Andreas Öchsner
- Faculty of Mechanical Engineering, Esslingen University of Applied Sciences, Kanalstrasse 33, 73728, Esslingen, Germany
| | - Yuri G Anissimov
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia
- School of Environment and Science, Griffith University, Parklands Drive, Southport, QLD, 4222, Australia
- Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia
| | - Maksym Rybachuk
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia.
- School of Engineering and Built Environment, Griffith University, 170 Kessels Road, Nathan, QLD, 4111, Australia.
| |
Collapse
|
11
|
Tavakoli Z, Yazdian F, Tabandeh F, Sheikhpour M. Regenerative medicine as a novel strategy for AMD treatment: a review. Biomed Phys Eng Express 2019; 6:012001. [PMID: 33438587 DOI: 10.1088/2057-1976/ab269a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Age-related macular degeneration (AMD) is known as a major cause of irreversible blindness in elderly adults. The segment of the retina responsible for central vision damages in the disease process. Degeneration of retinal pigmented epithelium (RPE) cells, photoreceptors, and choriocapillaris associated with aging participate for visual loss. In 2010, AMD involved 6.6% of all blindness cases around the world. Some of the researches have evaluated the replacing of damaged RPE in AMD patients by using the cells from various sources. Today, the advancement of RPE differentiation or generation from stem cells has been gained, and currently, clinical trials are testing the efficiency and safety of replacing degenerated RPE with healthy RPE. However, the therapeutic success of RPE transplantation may be restricted unless the transplanted cells can be adhered, distributed and survive for long-term in the transplanted site without any infections. In recent years a variety of scaffold types were used as a carrier for RPE transplantation and AMD treatment. In this review, we have discussed types of scaffolds; natural or synthetic, solid or hydrogel and their results in RPE replacement. Eventually, our aim is highlighting the novel and best scaffold carriers that may have potentially promoting the efficacy of RPE transplantation.
Collapse
Affiliation(s)
- Zahra Tavakoli
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | | | | | | |
Collapse
|
12
|
Garita-Hernandez M, Lampič M, Chaffiol A, Guibbal L, Routet F, Santos-Ferreira T, Gasparini S, Borsch O, Gagliardi G, Reichman S, Picaud S, Sahel JA, Goureau O, Ader M, Dalkara D, Duebel J. Restoration of visual function by transplantation of optogenetically engineered photoreceptors. Nat Commun 2019; 10:4524. [PMID: 31586094 PMCID: PMC6778196 DOI: 10.1038/s41467-019-12330-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 08/28/2019] [Indexed: 12/22/2022] Open
Abstract
A major challenge in the treatment of retinal degenerative diseases, with the transplantation of replacement photoreceptors, is the difficulty in inducing the grafted cells to grow and maintain light sensitive outer segments in the host retina, which depends on proper interaction with the underlying retinal pigment epithelium (RPE). Here, for an RPE-independent treatment approach, we introduce a hyperpolarizing microbial opsin into photoreceptor precursors from newborn mice, and transplant them into blind mice lacking the photoreceptor layer. These optogenetically-transformed photoreceptors are light responsive and their transplantation leads to the recovery of visual function, as shown by ganglion cell recordings and behavioral tests. Subsequently, we generate cone photoreceptors from human induced pluripotent stem cells, expressing the chloride pump Jaws. After transplantation into blind mice, we observe light-driven responses at the photoreceptor and ganglion cell levels. These results demonstrate that structural and functional retinal repair is possible by combining stem cell therapy and optogenetics.
Collapse
Affiliation(s)
| | - Maruša Lampič
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | - Antoine Chaffiol
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | - Laure Guibbal
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | - Fiona Routet
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | | | - Sylvia Gasparini
- CRTD/Center for Regenerative Therapies Dresden, CMCB, TU Dresden, Dresden, Germany
| | - Oliver Borsch
- CRTD/Center for Regenerative Therapies Dresden, CMCB, TU Dresden, Dresden, Germany
| | - Giuliana Gagliardi
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | - Sacha Reichman
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | - Serge Picaud
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | - José-Alain Sahel
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Olivier Goureau
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France
| | - Marius Ader
- CRTD/Center for Regenerative Therapies Dresden, CMCB, TU Dresden, Dresden, Germany
| | - Deniz Dalkara
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France.
| | - Jens Duebel
- Sorbonne Université, Institut de la Vision, INSERM, CNRS, 75012, Paris, France.
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
13
|
Lakowski J, Welby E, Budinger D, Di Marco F, Di Foggia V, Bainbridge JW, Wallace K, Gamm DM, Ali RR, Sowden JC. Isolation of Human Photoreceptor Precursors via a Cell Surface Marker Panel from Stem Cell-Derived Retinal Organoids and Fetal Retinae. Stem Cells 2018; 36:709-722. [PMID: 29327488 PMCID: PMC5947711 DOI: 10.1002/stem.2775] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 12/01/2017] [Accepted: 12/14/2017] [Indexed: 12/16/2022]
Abstract
Loss of photoreceptor cells due to retinal degeneration is one of the main causes of blindness in the developed world. Although there is currently no effective treatment, cell replacement therapy using stem-cell-derived photoreceptor cells may be a feasible future treatment option. In order to ensure safety and efficacy of this approach, robust cell isolation and purification protocols must be developed. To this end, we previously developed a biomarker panel for the isolation of mouse photoreceptor precursors from the developing mouse retina and mouse embryonic stem cell cultures. In the current study we applied this approach to the human pluripotent stem cell (hPSC) system, and identified novel biomarker combinations that can be leveraged for the isolation of human photoreceptors. Human retinal samples and hPSC-derived retinal organoid cultures were screened against 242 human monoclonal antibodies using a high through-put flow cytometry approach. We identified 46 biomarkers with significant expression levels in the human retina and hPSC differentiation cultures. Human retinal cell samples, either from fetal tissue or derived from embryonic and induced pluripotent stem cell cultures, were fluorescence-activated cell sorted (FACS) using selected candidate biomarkers that showed expression in discrete cell populations. Enrichment for photoreceptors and exclusion of mitotically active cells was demonstrated by immunocytochemical analysis with photoreceptor-specific antibodies and Ki-67. We established a biomarker combination, which enables the robust purification of viable human photoreceptors from both human retinae and hPSC-derived organoid cultures. Stem Cells 2018;36:709-722.
Collapse
Affiliation(s)
- Jörn Lakowski
- Stem Cells and Regenerative Medicine Section, University College LondonLondonUnited Kingdom
| | - Emily Welby
- Stem Cells and Regenerative Medicine Section, University College LondonLondonUnited Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, University College LondonLondonUnited Kingdom
| | - Dimitri Budinger
- Stem Cells and Regenerative Medicine Section, University College LondonLondonUnited Kingdom
| | - Fabiana Di Marco
- Stem Cells and Regenerative Medicine Section, University College LondonLondonUnited Kingdom
| | - Valentina Di Foggia
- Stem Cells and Regenerative Medicine Section, University College LondonLondonUnited Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, University College LondonLondonUnited Kingdom
| | | | - Kyle Wallace
- Waisman Center, University of Wisconsin‐Madison School of Medicine and Public Health, Waisman Center Rm T609MadisonWisconsinUSA
| | - David M. Gamm
- Waisman Center, University of Wisconsin‐Madison School of Medicine and Public Health, Waisman Center Rm T609MadisonWisconsinUSA
- Department of Ophthalmology and Visual Sciences and McPherson Eye Research InstituteUniversity of Wisconsin‐Madison School of Medicine and Public Health, Waisman Center Rm T609MadisonWisconsinUSA
| | - Robin R. Ali
- Department of GeneticsUCL Institute of OphthalmologyLondonUnited Kingdom
| | - Jane C. Sowden
- Stem Cells and Regenerative Medicine Section, University College LondonLondonUnited Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, University College LondonLondonUnited Kingdom
| |
Collapse
|
14
|
Maleki B, Tabandeh F, Soheili ZS, Morshedi D. Application of proteinous nanofibrils to culture retinal pigmented epithelium cells: A versatile biomaterial. REACT FUNCT POLYM 2017. [DOI: 10.1016/j.reactfunctpolym.2017.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
15
|
Abstract
The recent advances in cell-based therapies for the repair of the pigmented epithelium is providing additional impetus for the translation of photoreceptor transplantation to eventual clinical trials. The prospects for transplantation of photoreceptors as a potential therapy for the treatment of photoreceptor degeneration will depend on successfully addressing many critical issues in preclinical studies. Although most of the studies that have carried out transplants of photoreceptors have primarily used normal mice, there have been recent reports that have also shown some success following transplantation to mouse models of retinitis pigmentosa. However, while these results are promising, there are several key issues that require further investigation in order to better understand the optimum timing for transplantation, given the extensive remodeling of the retina that occurs in late stage disease.
Collapse
|
16
|
Novel Regulatory Mechanisms for the SoxC Transcriptional Network Required for Visual Pathway Development. J Neurosci 2017; 37:4967-4981. [PMID: 28411269 DOI: 10.1523/jneurosci.3430-13.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 01/01/2023] Open
Abstract
What pathways specify retinal ganglion cell (RGC) fate in the developing retina? Here we report on mechanisms by which a molecular pathway involving Sox4/Sox11 is required for RGC differentiation and for optic nerve formation in mice in vivo, and is sufficient to differentiate human induced pluripotent stem cells into electrophysiologically active RGCs. These data place Sox4 downstream of RE1 silencing transcription factor in regulating RGC fate, and further describe a newly identified, Sox4-regulated site for post-translational modification with small ubiquitin-related modifier (SUMOylation) in Sox11, which suppresses Sox11's nuclear localization and its ability to promote RGC differentiation, providing a mechanism for the SoxC familial compensation observed here and elsewhere in the nervous system. These data define novel regulatory mechanisms for this SoxC molecular network, and suggest pro-RGC molecular approaches for cell replacement-based therapies for glaucoma and other optic neuropathies.SIGNIFICANCE STATEMENT Glaucoma is the most common cause of blindness worldwide and, along with other optic neuropathies, is characterized by loss of retinal ganglion cells (RGCs). Unfortunately, vision and RGC loss are irreversible, and lead to bilateral blindness in ∼14% of all diagnosed patients. Differentiated and transplanted RGC-like cells derived from stem cells have the potential to replace neurons that have already been lost and thereby to restore visual function. These data uncover new mechanisms of retinal progenitor cell (RPC)-to-RGC and human stem cell-to-RGC fate specification, and take a significant step toward understanding neuronal and retinal development and ultimately cell-transplant therapy.
Collapse
|
17
|
Experimental Study of the Biological Properties of Human Embryonic Stem Cell-Derived Retinal Progenitor Cells. Sci Rep 2017; 7:42363. [PMID: 28205557 PMCID: PMC5304228 DOI: 10.1038/srep42363] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 01/09/2017] [Indexed: 01/16/2023] Open
Abstract
Retinal degenerative diseases are among the leading causes of blindness worldwide, and cell replacement is considered as a promising therapeutic. However, the resources of seed cells are scarce. To further explore this type of therapy, we adopted a culture system that could harvest a substantial quantity of retinal progenitor cells (RPCs) from human embryonic stem cells (hESCs) within a relatively short period of time. Furthermore, we transplanted these RPCs into the subretinal spaces of Royal College of Surgeons (RCS) rats. We quantified the thickness of the treated rats' outer nuclear layers (ONLs) and explored the visual function via electroretinography (ERG). It was found that the differentiated cells expressed RPC markers and photoreceptor progenitor markers. The transplanted RPCs survived for at least 12 weeks, resulting in beneficial effects on the morphology of the host retina, and led to a significant improvement in the visual function of the treated animals. These therapeutic effects suggest that the hESCs-derived RPCs could delay degeneration of the retina and partially restore visual function.
Collapse
|
18
|
Zalis MC, Johansson S, Englund-Johansson U. Immunocytochemical Profiling of Cultured Mouse Primary Retinal Cells. J Histochem Cytochem 2017; 65:223-239. [PMID: 28151698 PMCID: PMC5407564 DOI: 10.1369/0022155416689675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Primary retinal cell cultures and immunocytochemistry are important experimental platforms in ophthalmic research. Translation of retinal cells from their native environment to the in vitro milieu leads to cellular stress, jeopardizing their in vivo phenotype features. Moreover, the specificity and stability of many retinal immunochemical markers are poorly evaluated in retinal cell cultures. Hence, we here evaluated the expression profile of 17 retinal markers, that is, recoverin, rhodopsin, arrestin, Chx10, PKC, DCX, CRALBP, GS, vimentin, TPRV4, RBPMS, Brn3a, β-tubulin III, NeuN, MAP2, GFAP, and synaptophysin. At 7 and 18 days of culture, the marker expression profiles of mouse postnatal retinal cells were compared with their age-matched in vivo retinas. We demonstrate stable in vitro expression of all markers, except for arrestin and CRALBP. Differences in cellular expression and location of some markers were observed, both over time in culture and compared with the age-matched retina. We hypothesize that these differences are likely culture condition dependent. Taken together, we suggest a thorough evaluation of the antibodies in specific culture settings, before extrapolating the in vitro results to an in vivo setting. Moreover, the identification of specific cell types may require a combination of different genes expressed or markers with structural information.
Collapse
Affiliation(s)
- Marina C Zalis
- Division of Ophthalmology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden (MCZ, SJ, UEJ)
| | - Sebastian Johansson
- Division of Ophthalmology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden (MCZ, SJ, UEJ)
| | - Ulrica Englund-Johansson
- Division of Ophthalmology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden (MCZ, SJ, UEJ)
| |
Collapse
|
19
|
Guadagni V, Cerri C, Piano I, Novelli E, Gargini C, Fiorentini C, Caleo M, Strettoi E. The bacterial toxin CNF1 as a tool to induce retinal degeneration reminiscent of retinitis pigmentosa. Sci Rep 2016; 6:35919. [PMID: 27775019 PMCID: PMC5075935 DOI: 10.1038/srep35919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/04/2016] [Indexed: 12/02/2022] Open
Abstract
Retinitis pigmentosa (RP) comprises a group of inherited pathologies characterized by progressive photoreceptor degeneration. In rodent models of RP, expression of defective genes and retinal degeneration usually manifest during the first weeks of postnatal life, making it difficult to distinguish consequences of primary genetic defects from abnormalities in retinal development. Moreover, mouse eyes are small and not always adequate to test pharmacological and surgical treatments. An inducible paradigm of retinal degeneration potentially extensible to large animals is therefore desirable. Starting from the serendipitous observation that intraocular injections of a Rho GTPase activator, the bacterial toxin Cytotoxic Necrotizing Factor 1 (CNF1), lead to retinal degeneration, we implemented an inducible model recapitulating most of the key features of Retinitis Pigmentosa. The model also unmasks an intrinsic vulnerability of photoreceptors to the mechanism of CNF1 action, indicating still unexplored molecular pathways potentially leading to the death of these cells in inherited forms of retinal degeneration.
Collapse
Affiliation(s)
- Viviana Guadagni
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Chiara Cerri
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy.,Accademia dei Lincei, Rome, 00165, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, 56126, Italy
| | - Elena Novelli
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa, Pisa, 56126, Italy
| | | | - Matteo Caleo
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Enrica Strettoi
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| |
Collapse
|
20
|
Unachukwu UJ, Warren A, Li Z, Mishra S, Zhou J, Sauane M, Lim H, Vazquez M, Redenti S. Predicted molecular signaling guiding photoreceptor cell migration following transplantation into damaged retina. Sci Rep 2016; 6:22392. [PMID: 26935401 PMCID: PMC4776098 DOI: 10.1038/srep22392] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/11/2016] [Indexed: 12/18/2022] Open
Abstract
To replace photoreceptors lost to disease or trauma and restore vision, laboratories around the world are investigating photoreceptor replacement strategies using subretinal transplantation of photoreceptor precursor cells (PPCs) and retinal progenitor cells (RPCs). Significant obstacles to advancement of photoreceptor cell-replacement include low migration rates of transplanted cells into host retina and an absence of data describing chemotactic signaling guiding migration of transplanted cells in the damaged retinal microenvironment. To elucidate chemotactic signaling guiding transplanted cell migration, bioinformatics modeling of PPC transplantation into light-damaged retina was performed. The bioinformatics modeling analyzed whole-genome expression data and matched PPC chemotactic cell-surface receptors to cognate ligands expressed in the light-damaged retinal microenvironment. A library of significantly predicted chemotactic ligand-receptor pairs, as well as downstream signaling networks was generated. PPC and RPC migration in microfluidic ligand gradients were analyzed using a highly predicted ligand-receptor pair, SDF-1α – CXCR4, and both PPCs and RPCs exhibited significant chemotaxis. This work present a systems level model and begins to elucidate molecular mechanisms involved in PPC and RPC migration within the damaged retinal microenvironment.
Collapse
Affiliation(s)
- Uchenna John Unachukwu
- Biochemistry Doctoral Program, The Graduate School, City University of New York, New York, NY, USA.,Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY, USA
| | - Alice Warren
- Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY, USA
| | - Ze Li
- Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY, USA
| | - Shawn Mishra
- Department of Biomedical Engineering, City College of New York, City University of New York, NY, USA
| | - Jing Zhou
- Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY, USA.,Neuroscience Doctoral Program, The Graduate School, City University of New York, New York, NY, USA
| | - Moira Sauane
- Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY, USA
| | - Hyungsik Lim
- Departments of Physics and Biology, Hunter College of the City University of New York, New York, NY USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, City College of New York, City University of New York, NY, USA
| | - Stephen Redenti
- Biochemistry Doctoral Program, The Graduate School, City University of New York, New York, NY, USA.,Department of Biological Sciences, Lehman College, City University of New York, Bronx, NY, USA
| |
Collapse
|
21
|
Hamon A, Roger JE, Yang XJ, Perron M. Müller glial cell-dependent regeneration of the neural retina: An overview across vertebrate model systems. Dev Dyn 2016; 245:727-38. [PMID: 26661417 PMCID: PMC4900950 DOI: 10.1002/dvdy.24375] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/12/2015] [Accepted: 11/22/2015] [Indexed: 12/21/2022] Open
Abstract
Retinal dystrophies are a major cause of blindness for which there are currently no curative treatments. Transplantation of stem cell‐derived neuronal progenitors to replace lost cells has been widely investigated as a therapeutic option. Another promising strategy would be to trigger self‐repair mechanisms in patients, through the recruitment of endogenous cells with stemness properties. Accumulating evidence in the past 15 year0s has revealed that several retinal cell types possess neurogenic potential, thus opening new avenues for regenerative medicine. Among them, Müller glial cells have been shown to be able to undergo a reprogramming process to re‐acquire a stem/progenitor state, allowing them to proliferate and generate new neurons for repair following retinal damages. Although Müller cell–dependent spontaneous regeneration is remarkable in some species such as the fish, it is extremely limited and ineffective in mammals. Understanding the cellular events and molecular mechanisms underlying Müller cell activities in species endowed with regenerative capacities could provide knowledge to unlock the restricted potential of their mammalian counterparts. In this context, the present review provides an overview of Müller cell responses to injury across vertebrate model systems and summarizes recent advances in this rapidly evolving field. Developmental Dynamics 245:727–738, 2016. © 2015 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc. The present review provides an overview of Müller cell responses to injury across vertebrate model systems and summarizes recent advances in this rapidly evolving field.
Collapse
Affiliation(s)
- Annaïg Hamon
- Paris-Saclay Institute of Neuroscience, CNRS, Univ Paris Sud, Université Paris-Saclay, Orsay, France.,Centre d'Etude et de Recherche Thérapeutique en Ophtalmologie, Retina France, Orsay, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neuroscience, CNRS, Univ Paris Sud, Université Paris-Saclay, Orsay, France.,Centre d'Etude et de Recherche Thérapeutique en Ophtalmologie, Retina France, Orsay, France
| | - Xian-Jie Yang
- Stein Eye Institute, University of California Los Angeles, Los Angeles, California
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CNRS, Univ Paris Sud, Université Paris-Saclay, Orsay, France.,Centre d'Etude et de Recherche Thérapeutique en Ophtalmologie, Retina France, Orsay, France.,Stein Eye Institute, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
22
|
Abstract
Severe loss of photoreceptor cells in inherited or acquired retinal degenerative diseases can result in partial loss of sight or complete blindness. The optogenetic strategy for restoration of vision utilizes optogenetic tools to convert surviving inner retinal neurons into photosensitive cells; thus, light sensitivity is imparted to the retina after the death of photoreceptor cells. Proof-of-concept studies, especially those using microbial rhodopsins, have demonstrated restoration of light responses in surviving retinal neurons and visually guided behaviors in animal models. Significant progress has also been made in improving microbial rhodopsin-based optogenetic tools, developing virus-mediated gene delivery, and targeting specific retinal neurons and subcellular compartments of retinal ganglion cells. In this article, we review the current status of the field and outline further directions and challenges to the advancement of this strategy toward clinical application and improvement in the outcomes of restored vision.
Collapse
Affiliation(s)
- Zhuo-Hua Pan
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; , , .,Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201;
| | - Qi Lu
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201;
| | - Anding Bi
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; , ,
| | | | - Gary W Abrams
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; , ,
| |
Collapse
|
23
|
Qu Z, Guan Y, Cui L, Song J, Gu J, Zhao H, Xu L, Lu L, Jin Y, Xu GT. Transplantation of rat embryonic stem cell-derived retinal progenitor cells preserves the retinal structure and function in rat retinal degeneration. Stem Cell Res Ther 2015; 6:219. [PMID: 26553210 PMCID: PMC4640237 DOI: 10.1186/s13287-015-0207-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Degenerative retinal diseases like age-related macular degeneration (AMD) are the leading cause of blindness. Cell transplantation showed promising therapeutic effect for such diseases, and embryonic stem cell (ESC) is one of the sources of such donor cells. Here, we aimed to generate retinal progenitor cells (RPCs) from rat ESCs (rESCs) and to test their therapeutic effects in rat model. METHODS The rESCs (DA8-16) were cultured in N2B27 medium with 2i, and differentiated to two types of RPCs following the SFEBq method with modifications. For rESC-RPC1, the cells were switched to adherent culture at D10, while for rESC-RPC2, the suspension culture was maintained to D14. Both RPCs were harvested at D16. Primary RPCs were obtained from P1 SD rats, and some of them were labeled with EGFP by infection with lentivirus. To generate Rax::EGFP knock-in rESC lines, TALENs were engineered to facilitate homologous recombination in rESCs, which were cotransfected with the targeting vector and TALEN vectors. The differentiated cells were analyzed with live image, immunofluorescence staining, flow cytometric analysis, gene expression microarray, etc. RCS rats were used to mimic the degeneration of retina and test the therapeutic effects of subretinally transplanted donor cells. The structure and function of retina were examined. RESULTS We established two protocols through which two types of rESC-derived RPCs were obtained and both contained committed retina lineage cells and some neural progenitor cells (NPCs). These rESC-derived RPCs survived in the host retinas of RCS rats and protected the retinal structure and function in early stage following the transplantation. However, the glia enriched rESC-RPC1 obtained through early and longer adherent culture only increased the b-wave amplitude at 4 weeks, while the longer suspension culture gave rise to evidently neuronal differentiation in rESC-RPC2 which significantly improved the visual function of RCS rats. CONCLUSIONS We have successfully differentiated rESCs to glia enriched RPCs and retinal neuron enriched RPCs in vitro. The retinal neuron enriched rESC-RPC2 protected the structure and function of retina in rats with genetic retinal degeneration and could be a candidate cell source for treating some degenerative retinal diseases in human trials.
Collapse
Affiliation(s)
- Zepeng Qu
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Room 208, Building 5, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Yuan Guan
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Room 208, Building 5, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Lu Cui
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Room 208, Building 5, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Jian Song
- ShanghaiTech University School of Life Science and Technology, Shanghai, 201210, China.
| | - Junjie Gu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China.
| | - Hanzhi Zhao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China.
| | - Lei Xu
- Department of Regenerative Medicine, Stem Cell Research Center, and Institute for Nutritional Sciences, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Lixia Lu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, Tongji University School of Medicine, 1239 Siping Road, Medical Building, Room 521, Shanghai, 200092, China.
- Department of Regenerative Medicine, Stem Cell Research Center, and Institute for Nutritional Sciences, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Ying Jin
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Room 208, Building 5, 280 South Chongqing Road, Shanghai, 200025, China.
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China.
- ShanghaiTech University School of Life Science and Technology, Shanghai, 201210, China.
| | - Guo-Tong Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, Tongji University School of Medicine, 1239 Siping Road, Medical Building, Room 521, Shanghai, 200092, China.
- Department of Regenerative Medicine, Stem Cell Research Center, and Institute for Nutritional Sciences, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
24
|
|
25
|
Fields MA, Bowrey HE, Gong J, Ablonczy Z, Del Priore LV. Retinoid Processing in Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium Cultures. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 134:477-90. [PMID: 26310172 DOI: 10.1016/bs.pmbts.2015.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stem cell therapy for retinal degenerative diseases such as age-related macular degeneration is a promising clinical option for the replacement of photoreceptors and retinal pigment epithelium (RPE). Induced pluripotent stem cell technology has emerged as a viable potential source of cells for transplantation in retinal degenerative disorders. Induced pluripotent stem cells have been used to derive RPE and have been tested for their functional behavior. These cells have the ability to express RPE-specific proteins and morphologically resemble native RPE. Induced pluripotent stem cell-derived RPE are also able to contribute to the visual cycle by their ability to metabolize all-trans retinol, a critical function of RPE in maintaining visual function. Advances in induced pluripotent stem cell technology will contribute to the development of clinical therapies for retinal degenerative diseases as well as provide a tool to understand the pathology of these disorders.
Collapse
Affiliation(s)
- Mark A Fields
- Department of Ophthalmology, Albert Florens Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, USA.
| | - Hannah E Bowrey
- Department of Ophthalmology, Albert Florens Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jie Gong
- Department of Ophthalmology, Albert Florens Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Zsolt Ablonczy
- Department of Ophthalmology, Albert Florens Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lucian V Del Priore
- Department of Ophthalmology, Albert Florens Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
26
|
Lakowski J, Gonzalez-Cordero A, West EL, Han YT, Welby E, Naeem A, Blackford SJI, Bainbridge JWB, Pearson RA, Ali RR, Sowden JC. Transplantation of Photoreceptor Precursors Isolated via a Cell Surface Biomarker Panel From Embryonic Stem Cell-Derived Self-Forming Retina. Stem Cells 2015; 33:2469-82. [PMID: 25982268 PMCID: PMC4862023 DOI: 10.1002/stem.2051] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 03/21/2015] [Indexed: 10/25/2022]
Abstract
Loss of photoreceptors due to retinal degeneration is a major cause of untreatable blindness. Cell replacement therapy, using pluripotent stem cell-derived photoreceptor cells, may be a feasible future treatment. Achieving safe and effective cell replacement is critically dependent on the stringent selection and purification of optimal cells for transplantation. Previously, we demonstrated effective transplantation of post-mitotic photoreceptor precursor cells labelled by fluorescent reporter genes. As genetically labelled cells are not desirable for therapy, here we developed a surface biomarker cell selection strategy for application to complex pluripotent stem cell differentiation cultures. We show that a five cell surface biomarker panel CD73(+)CD24(+)CD133(+)CD47(+)CD15(-) facilitates the isolation of photoreceptor precursors from three-dimensional self-forming retina differentiated from mouse embryonic stem cells. Importantly, stem cell-derived cells isolated using the biomarker panel successfully integrate and mature into new rod photoreceptors in the adult mouse retinae after subretinal transplantation. Conversely, unsorted or negatively selected cells do not give rise to newly integrated rods after transplantation. The biomarker panel also removes detrimental proliferating cells prior to transplantation. Notably, we demonstrate how expression of the biomarker panel is conserved in the human retina and propose that a similar selection strategy will facilitate isolation of human transplantation-competent cells for therapeutic application.
Collapse
Affiliation(s)
- Jorn Lakowski
- Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health, University College London, London, United Kingdom
| | | | - Emma L West
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Ya-Ting Han
- Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health, University College London, London, United Kingdom
| | - Emily Welby
- Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health, University College London, London, United Kingdom
| | - Arifa Naeem
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | | | | | - Rachael A Pearson
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Robin R Ali
- Department of Genetics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Jane C Sowden
- Stem Cells and Regenerative Medicine Section, UCL Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
27
|
Xu Y, Balasubramaniam B, Copland DA, Liu J, Armitage MJ, Dick AD. Activated adult microglia influence retinal progenitor cell proliferation and differentiation toward recoverin-expressing neuron-like cells in a co-culture model. Graefes Arch Clin Exp Ophthalmol 2015; 253:1085-96. [PMID: 25680876 DOI: 10.1007/s00417-015-2961-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/15/2014] [Accepted: 12/22/2014] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Microglia contribute to immune homeostasis of the retina, and thus act as a potential regulator determining successful repair or retinal stem cell transplantation. We investigated the interaction between human microglia and retinal progenitor cells in cell co-culture to further our exploration on developing a new therapeutic strategy for retinal degeneration. METHODS Microglia and retinal progenitor cultures were developed using CD11b(+) and CD133(+), respectively, from adult donor retina. Microglia activation was developed using interferon-gamma and lipopolysaccharide. Retinal progenitor differentiation was analysed in co-culture with or without microglial activation. Retinal progenitor proliferation was analysed in presence of conditioned medium from activated microglia. Phenotype and function of adult human retinal cell cultures were examined using cell morphology, immunohistochemistry and real-time PCR. RESULTS By morphology, neuron-like cells generated in co-culture expressed photoreceptor marker recoverin. Neurospheres derived from retinal progenitor cells showed reduced growth in the presence of conditioned medium from activated microglia. Delayed retinal progenitor cell migration and reduced cellular differentiation was observed in co-cultures with activated microglia. In independent experiments, activated microglia showed enhanced mRNA expression of CXCL10, IL-27, IL-6, and TNF-alpha compared to controls. CONCLUSION Adult human retina retains retinal progenitors or potential to reprogram cells to then proliferate and differentiate into neuron-like cells in vitro. Human microglia support retinal progenitor differentiation into neuron-like cells, but such capacity is altered following microglial activation. Modulating microglia activity is a potential approach to promote retinal repair and facilitate success of stem-cell transplantation.
Collapse
Affiliation(s)
- Yunhe Xu
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom,
| | | | | | | | | | | |
Collapse
|
28
|
Mansergh FC, Carrigan M, Hokamp K, Farrar GJ. Gene expression changes during retinal development and rod specification. Mol Vis 2015; 21:61-87. [PMID: 25678762 PMCID: PMC4301594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/16/2015] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Retinitis pigmentosa (RP) typically results from individual mutations in any one of >70 genes that cause rod photoreceptor cells to degenerate prematurely, eventually resulting in blindness. Gene therapies targeting individual RP genes have shown efficacy at clinical trial; however, these therapies require the surviving photoreceptor cells to be viable and functional, and may be economically feasible for only the more commonly mutated genes. An alternative potential treatment strategy, particularly for late stage disease, may involve stem cell transplants into the photoreceptor layer of the retina. Rod progenitors from postnatal mouse retinas can be transplanted and can form photoreceptors in recipient adult retinas; optimal numbers of transplantable cells are obtained from postnatal day 3-5 (P3-5) retinas. These cells can also be expanded in culture; however, this results in the loss of photoreceptor potential. Gene expression differences between postnatal retinas, cultured retinal progenitor cells (RPCs), and rod photoreceptor precursors were investigated to identify gene expression patterns involved in the specification of rod photoreceptors. METHODS Microarrays were used to investigate differences in gene expression between cultured RPCs that have lost photoreceptor potential, P1 retinas, and fresh P5 retinas that contain significant numbers of transplantable photoreceptors. Additionally, fluorescence-activated cell sorting (FACS) sorted Rho-eGFP-expressing rod photoreceptor precursors were compared with Rho-eGFP-negative cells from the same P5 retinas. Differential expression was confirmed with quantitative polymerase chain reaction (q-PCR). RESULTS Analysis of the microarray data sets, including the use of t-distributed stochastic neighbor embedding (t-SNE) to identify expression pattern neighbors of key photoreceptor specific genes, resulted in the identification of 636 genes differentially regulated during rod specification. Forty-four of these genes when mutated have previously been found to cause retinal disease. Although gene function in other tissues may be known, the retinal function of approximately 61% of the gene list is as yet undetermined. Many of these genes' promoters contain binding sites for the key photoreceptor transcription factors Crx and Nr2e3; moreover, the genomic clustering of differentially regulated genes appears to be non-random. CONCLUSIONS This study aids in understanding gene expression differences between rod photoreceptor progenitors versus cultured RPCs that have lost photoreceptor potential. The results provide insights into rod photoreceptor development and should expedite the development of cell-based treatments for RP. Furthermore, the data set includes a large number of retinopathy genes; less-well-characterized genes within this data set are a resource for those seeking to identify novel retinopathy genes in patients with RP (GEO accession: GSE59201).
Collapse
Affiliation(s)
- Fiona C Mansergh
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Matthew Carrigan
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Karsten Hokamp
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - G Jane Farrar
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
29
|
Huang R, Baranov P, Lai K, Zhang X, Ge, J, Young MJ. Functional and morphological analysis of the subretinal injection of human retinal progenitor cells under Cyclosporin A treatment. Mol Vis 2014; 20:1271-80. [PMID: 25352736 PMCID: PMC4168833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 09/17/2014] [Indexed: 11/17/2022] Open
Abstract
PURPOSE The purpose of this study is to evaluate the functional and morphological changes in subretinal xenografts of human retinal progenitor cells (hRPCs) in B6 mice treated with Cyclosporin A (CsA; 210 mg/l in drinking water). METHODS The hRPCs from human fetal eyes were isolated and expanded for transplantation. These cells, with green fluorescent protein (GFP) at 11 passages, were transplanted into the subretinal space in B6 mice. A combination of invasive and noninvasive approaches was used to analyze the structural and functional consequences of the subretinal injection of the hRPCs. The process of change was monitored using spectral domain optical coherence tomography (SDOCT), histology, and electroretinography (ERG) at 3 days, 1 week, and 3 weeks after transplantation. Cell counts were used to evaluate the survival rate with a confocal microscope. ERGs were performed to evaluate the physiologic changes, and the structural changes were evaluated using SDOCT and histological examination. RESULTS The results of the histological examination showed that the hRPCs gained a better survival rate in the mice treated with CsA. The SDOCT showed that the bleb size of the retinal detachment was significantly decreased, and the retinal reattachment was nearly complete by 3 weeks. The ERG response amplitudes in the CsA group were less decreased after the injection, when compared with the control group, in the dark-adapted and light-adapted conditions. However, the cone-mediated function in both groups was less affected by the transplantation after 3 weeks than the rod-mediated function. CONCLUSIONS Although significant functional and structural recovery was observed after the subretinal injection of the hRPCs, the effectiveness of CsA in xenotransplantation may be a novel and potential approach for increasing retinal progenitor cell survival.
Collapse
Affiliation(s)
- Rui Huang
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China,Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| | - Petr Baranov
- Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| | - Kunbei Lai
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China,Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| | - Xinmei Zhang
- Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| | - Jian Ge,
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Michael J. Young
- Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Yu H, Vu THK, Cho KS, Guo C, Chen DF. Mobilizing endogenous stem cells for retinal repair. Transl Res 2014; 163:387-98. [PMID: 24333552 PMCID: PMC3976683 DOI: 10.1016/j.trsl.2013.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 02/06/2023]
Abstract
Irreversible vision loss is most often caused by the loss of function and subsequent death of retinal neurons, such as photoreceptor cells-the cells that initiate vision by capturing and transducing signals of light. One reason why retinal degenerative diseases are devastating is that, once retinal neurons are lost, they don't grow back. Stem cell-based cell replacement strategy for retinal degenerative diseases are leading the way in clinical trials of transplantation therapy, and the exciting findings in both human and animal models point to the possibility of restoring vision through a cell replacement regenerative approach. A less invasive method of retinal regeneration by mobilizing endogenous stem cells is, thus, highly desirable and promising for restoring vision. Although many obstacles remain to be overcome, the field of endogenous retinal repair is progressing at a rapid pace, with encouraging results in recent years.
Collapse
Affiliation(s)
- Honghua Yu
- Department of Ophthalmology, Liuhuaqiao Hospital, Guangzhou, PR China; Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass
| | - Thi Hong Khanh Vu
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass; Department of Ophthalmology, Leiden University Medical Center, Leiden, Netherlands
| | - Kin-Sang Cho
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass
| | - Chenying Guo
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass
| | - Dong Feng Chen
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass; VA Boston Healthcare System, Boston, Mass.
| |
Collapse
|
33
|
Unachukwu UJ, Sauane M, Vazquez M, Redenti S. Microfluidic generated EGF-gradients induce chemokinesis of transplantable retinal progenitor cells via the JAK/STAT and PI3kinase signaling pathways. PLoS One 2013; 8:e83906. [PMID: 24376770 PMCID: PMC3871684 DOI: 10.1371/journal.pone.0083906] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/10/2013] [Indexed: 11/18/2022] Open
Abstract
A growing number of studies are evaluating retinal progenitor cell (RPC) transplantation as an approach to repair retinal degeneration and restore visual function. To advance cell-replacement strategies for a practical retinal therapy, it is important to define the molecular and biochemical mechanisms guiding RPC motility. We have analyzed RPC expression of the epidermal growth factor receptor (EGFR) and evaluated whether exposure to epidermal growth factor (EGF) can coordinate motogenic activity in vitro. Using Boyden chamber analysis as an initial high-throughput screen, we determined that RPC motility was optimally stimulated by EGF concentrations in the range of 20-400 ng/ml, with decreased stimulation at higher concentrations, suggesting concentration-dependence of EGF-induced motility. Using bioinformatics analysis of the EGF ligand in a retina-specific gene network pathway, we predicted a chemotactic function for EGF involving the MAPK and JAK-STAT intracellular signaling pathways. Based on targeted inhibition studies, we show that ligand binding, phosphorylation of EGFR and activation of the intracellular STAT3 and PI3kinase signaling pathways are necessary to drive RPC motility. Using engineered microfluidic devices to generate quantifiable steady-state gradients of EGF coupled with live-cell tracking, we analyzed the dynamics of individual RPC motility. Microfluidic analysis, including center of mass and maximum accumulated distance, revealed that EGF induced motility is chemokinetic with optimal activity observed in response to low concentration gradients. Our combined results show that EGFR expressing RPCs exhibit enhanced chemokinetic motility in the presence of low nanomole levels of EGF. These findings may serve to inform further studies evaluating the extent to which EGFR activity, in response to endogenous ligand, drives motility and migration of RPCs in retinal transplantation paradigms.
Collapse
Affiliation(s)
- Uchenna J. Unachukwu
- Biochemistry Doctoral Program, The Graduate School and University Center, City University of New York, New York, New York, United States of America
| | - Moira Sauane
- Department of Biological Sciences, Herbert Lehman College, City University of New York, Bronx, New York, United States of America
| | - Maribel Vazquez
- Department of Biomedical Engineering, City College of New York, City University of New York, New York, New York, United States of America
| | - Stephen Redenti
- Biochemistry Doctoral Program, The Graduate School and University Center, City University of New York, Department of Biological Sciences, Herbert Lehman College, City University of New York, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
34
|
Al-Ubaidi MR, Naash MI, Conley SM. A perspective on the role of the extracellular matrix in progressive retinal degenerative disorders. Invest Ophthalmol Vis Sci 2013; 54:8119-24. [PMID: 24346621 DOI: 10.1167/iovs.13-13536] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Progressive inherited retinal degenerative disorders (PIRDDs) are the leading cause of blindness in developed countries, with AMD and RP constituting the majority of PIRDDs. Currently, over 8 million Americans have PIRDDs, and that number is estimated to drastically increase by the end of this decade. Although a mutant protein is expressed starting early during retinal development in patients with PIRDDs, symptoms of retinal degeneration do not manifest until much later. Historically, research has focused on understanding the role a mutation has in the function of a protein and what role the mutant protein has in the disease process. However, it remains unknown why the disease, irrespective of the mutation, manifests clinically much later in life, while cellular indicators of disease (e.g., accumulation of toxic protein products and cell death) occur throughout early and middle life. Herein, we propose that there exists a time point at which the degenerative process is accelerated, leading to the appearance of clinical symptoms. This point is defined by structural disruptions of the extracellular matrix (ECM). Death of a critical number of ECM-maintaining mutant protein-expressing retinal cells contributes to that break point in the degenerative process. Therefore, it is important to understand the changes occurring at the ECM during PIRDDs and to take that into account when therapeutic approaches are designed.
Collapse
Affiliation(s)
- Muayyad R Al-Ubaidi
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | | | | |
Collapse
|
35
|
Tzameret A, Sher I, Belkin M, Treves AJ, Meir A, Nagler A, Levkovitch-Verbin H, Barshack I, Rosner M, Rotenstreich Y. Transplantation of human bone marrow mesenchymal stem cells as a thin subretinal layer ameliorates retinal degeneration in a rat model of retinal dystrophy. Exp Eye Res 2013; 118:135-44. [PMID: 24239509 DOI: 10.1016/j.exer.2013.10.023] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/24/2013] [Accepted: 10/31/2013] [Indexed: 02/06/2023]
Abstract
Vision incapacitation and blindness associated with retinal degeneration affect millions of people worldwide. Cell based therapy and specifically transplantation of human adult bone marrow-derived stem cells (hBM-MSCs) present possible treatment strategy. Subretinal transplantation of human or rat BM-MSCs was shown previously to improve retinal function in Royal College Surgeons (RCS) rats. In those studies cells were transplanted via a transscleral-transchoroidal approach, creating a localized subretinal bleb. Limited number of cells could be injected and photoreceptor rescue was restricted to areas in proximity to the injection site. Here we describe a new surgical method for subretinal transplantation that facilitates uniform distribution of transplanted cells as a thin layer along most of the subretinal space. We assessed the therapeutic effect of hBM-MSCs on RCS rats when transplanted either subretinally or intravitreally. We also examined whether a second transplantation can prolong the therapeutic effect. A cell suspension of 2.5 × 10(6) cells in 5 μl was injected subretinally or intravitreally in RCS rats at 28 days postnatal. In the subretinal group, hBM-MSCs were transplanted posterior to the limbus in the superotemporal part of the eye through a longitudinal triangular scleral tunnel reaching the choroid. In the intravitreal group, the cells were injected into the superotemporal part of the vitreous cavity. In cross sections of subretinally transplanted eyes, removed 2 h following transplantation, hBM-MSCs were distributed as a near-homogenous thin layer along most of the subretinal space. In some animals the cells were also detected in the choroid. In the intravitreal injection group, hBM-MSCs were clustered in the vitreous cavity. Transplanted cells could be detected up to 2 weeks after transplantation but not at later time points. Retinal function and structure were assessed by electroretinogram (ERG) and histology analysis, respectively. Six weeks post transplantation, the mean maximal scotopic ERG b-wave amplitude response recorded in RCS control eyes was 1.2 μV. By contrast, in transplanted eyes mean responses of 56.4 μV and 66.2 μV were recorded in the intravitreally and subretinally transplanted eyes, respectively. In the subretinal group, retinal function was significantly higher in transplanted compared with control eyes up to 20 weeks following transplantation. By contrast, in the intravitreal group, rescue of retinal function persisted only up to 12 weeks following transplantation. Histological analysis revealed that 8 weeks following subretinal transplantation, the retinas of control eyes were dystrophic, with outer nuclear layer (ONL) containing a single cell layer. An extensive photoreceptor rescue was demonstrated in transplanted eyes at this time point, with 3-4 cell layers in the ONL along the entire retina. A second subretinal transplantation at 70 days postnatal did not enhance or prolong the therapeutic effect of hBM-MSCs. No immunosuppressants were used and long-term safety analysis demonstrated no gross or microscopic adverse effects. Taken together our findings suggest that transplantation of hBM-MSCs as a thin subretinal layer enhances the therapeutic effect and the safety of cell transplantation.
Collapse
Affiliation(s)
- Adi Tzameret
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Ifat Sher
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Michael Belkin
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Avraham J Treves
- Center for Stem Cells and Regenerative Medicine, Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amilia Meir
- Center for Stem Cells and Regenerative Medicine, Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Arnon Nagler
- Hematology Division, Sheba Medical Center, Tel-Hashomer, Israel
| | - Hani Levkovitch-Verbin
- Rothberg Ophthalmic Molecular Biology Laboratory, Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer, Israel
| | - Iris Barshack
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer, Israel
| | - Mordechai Rosner
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Ygal Rotenstreich
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel.
| |
Collapse
|
36
|
Jung G, Sun J, Petrowitz B, Riecken K, Kruszewski K, Jankowiak W, Kunst F, Skevas C, Richard G, Fehse B, Bartsch U. Genetically modified neural stem cells for a local and sustained delivery of neuroprotective factors to the dystrophic mouse retina. Stem Cells Transl Med 2013; 2:1001-10. [PMID: 24167317 DOI: 10.5966/sctm.2013-0013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A continuous intraocular delivery of neurotrophic factors (NFs) is being explored as a strategy to rescue photoreceptor cells and visual functions in degenerative retinal disorders that are currently untreatable. To establish a cell-based intraocular delivery system for a sustained administration of NFs to the dystrophic mouse retina, we used a polycistronic lentiviral vector to genetically modify adherently cultivated murine neural stem (NS) cells. The vector concurrently encoded a gene of interest, a reporter gene, and a resistance gene and thus facilitated the selection, cloning, and in vivo tracking of the modified cells. To evaluate whether modified NS cells permit delivery of functionally relevant quantities of NFs to the dystrophic mouse retina, we expressed a secretable variant of ciliary neurotrophic factor (CNTF) in NS cells and grafted the cells into the vitreous space of Pde6b(rd1) and Pde6b(rd10) mice, two animal models of retinitis pigmentosa. In both mouse lines, grafted cells attached to the retina and lens, where they differentiated into astrocytes and some neurons. Adverse effects of the transplanted cells on the morphology of host retinas were not observed. Importantly, the CNTF-secreting NS cells significantly attenuated photoreceptor degeneration in both mutant mouse lines. The neuroprotective effect was significantly more pronounced when clonally derived NS cell lines selected for high expression levels of CNTF were grafted into Pde6b(rd1) mice. Intravitreal transplantations of modified NS cells may thus represent a useful method for preclinical studies aimed at evaluating the therapeutic potential of a cell-based intraocular delivery of NFs in mouse models of photoreceptor degeneration.
Collapse
|
37
|
Chen G, Ma J, Shatos MA, Chen H, Cyr D, Lashkari K. Application of human persistent fetal vasculature neural progenitors for transplantation in the inner retina. Cell Transplant 2013; 21:2621-34. [PMID: 23317920 DOI: 10.3727/096368912x647153] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Persistent fetal vasculature (PFV) is a potentially serious developmental anomaly in human eyes, which results from a failure of the primary vitreous and the hyaloid vascular systems to regress during development. Recent findings from our laboratory indicate that fibrovascular membranes harvested from subjects with PFV contain neural progenitor cells (herein called NPPFV cells). Our studies on successful isolation, culture, and characterization of NPPFV cells have shown that they highly express neuronal progenitor markers (nestin, Pax6, and Ki67) as well as retinal neuronal markers (β-III-tubulin and Brn3a). In the presence of retinoic acid and neurotrophins, these cells acquire a neural morphological appearance in vitro, including a round soma and extensive neurites, and express mature neuronal markers (β-III-tubulin and NF200). Further experiments, including real-time qRT-PCR to quantify characteristic gene expression profiles of these cells and Ca(2+) imaging to evaluate the response to stimulation with different neurotransmitters, indicate that NPPFV cells may resemble a more advanced stage of retinal development and show more differentiation toward inner retinal neurons rather than photoreceptors. To explore the potential of inner retinal transplantation, NPPFV cells were transplanted intravitreally into the eyes of adult C57BL/6 mice. Engrafted NPPFV cells survived well in the intraocular environment in presence of rapamycin and some cells migrated into the inner nuclear layer of the retina 1 week posttransplantation. Three weeks after transplantation, NPPFV cells were observed to migrate and integrate in the inner retina. In response to daily intraperitoneal injections of retinoic acid, a portion of transplanted NPPFV cells exhibited retinal ganglion cell-like morphology and expressed mature neuronal markers (β-III-tubulin and synaptophysin). These findings indicate that fibrovascular membranes from human PFV harbor a population of neuronal progenitors that may be potential candidates for cell-based therapy for degenerative diseases of the inner retina.
Collapse
Affiliation(s)
- Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | | | | | | | | | | |
Collapse
|
38
|
Karl MO. The potential of stem cell research for the treatment of neuronal damage in glaucoma. Cell Tissue Res 2013; 353:311-25. [PMID: 23708526 DOI: 10.1007/s00441-013-1646-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/23/2013] [Indexed: 01/29/2023]
Abstract
Stem cell research offers a wide variety of approaches for the advancement of our understanding of basic mechanisms of neurodegeneration and tissue regeneration and for the discovery and development of new therapeutic strategies to prevent and restore neuronal cell loss. Similar to most other regions of our central nervous system, degenerative diseases of the retina lead to the loss of neurons, which are not replaced. Recent work in animals has provided proof-of-concept evidence for the restoration of photoreceptor cells by cell transplantation and neuronal cell replacement by regeneration from endogenous cell sources. However, efficient therapeutic prevention of neuronal cell loss has not been achieved. Moreover, successful cell replacement of retinal neurons in humans, including that of ganglion cells, remains a major challenge. Future successes in the discovery and translation of neuroprotective drug and gene therapies and of cell-based regenerative therapies will depend on a better understanding of the underlying disease pathomechanisms. Existing stem cell and cell-reprogramming technologies offer the potential to generate human retina cells, to develop specific human-cell-based retina disease models, and to open up novel therapeutic strategies. Further, we might glean substantial knowledge from species that can or cannot regenerate their neuronal retina, in the search for new therapeutic approaches. Thus, stem cell research will pave the way toward clinical translation. In this review, I address some of the major possibilities presently on offer and speculate about the power of stem cell research to gain further insights into the pathomechanisms of retinal neurodegeneration (with special emphasis on glaucoma) and to advance our therapeutic options.
Collapse
Affiliation(s)
- Mike O Karl
- German Center for Neurodegenerative Diseases e.V. (DZNE), Arnoldstrasse 18/18b, 01307, Dresden, Germany.
| |
Collapse
|
39
|
Melville H, Carpiniello M, Hollis K, Staffaroni A, Golestaneh N. Stem cells: a new paradigm for disease modeling and developing therapies for age-related macular degeneration. J Transl Med 2013; 11:53. [PMID: 23452406 PMCID: PMC3599723 DOI: 10.1186/1479-5876-11-53] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 02/19/2013] [Indexed: 02/06/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in people over age 55 in the U.S. and the developed world. This condition leads to the progressive impairment of central visual acuity. There are significant limitations in the understanding of disease progression in AMD as well as a lack of effective methods of treatment. Lately, there has been considerable enthusiasm for application of stem cell biology for both disease modeling and therapeutic application. Human embryonic stem cells and induced pluripotent stem cells (iPSCs) have been used in cell culture assays and in vivo animal models. Recently a clinical trial was approved by FDA to investigate the safety and efficacy of the human embryonic stem cell-derived retinal pigment epithelium (RPE) transplantation in sub-retinal space of patients with dry AMD These studies suggest that stem cell research may provide both insight regarding disease development and progression, as well as direction for therapeutic innovation for the millions of patients afflicted with AMD.
Collapse
Affiliation(s)
- Heather Melville
- Georgetown University School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
| | - Matthew Carpiniello
- Georgetown University School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
| | - Kia Hollis
- Georgetown University School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
| | - Andrew Staffaroni
- Georgetown University School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
| | - Nady Golestaneh
- Georgetown University School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
- Department of Ophthalmology, Georgetown University, School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
- Department of Neurology, Georgetown University, School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, School of Medicine, 3900 Reservoir Rd, Washington, DC 20057, USA
| |
Collapse
|
40
|
Striem-Amit E, Bubic R, Amedi A. Neurophysiological Mechanisms Underlying Plastic Changes and Rehabilitation following Sensory Loss in Blindness and Deafness. Front Neurosci 2013. [DOI: 10.1201/9781439812174-27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
|
41
|
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
|
42
|
Forrester JV, Xu H. Good news-bad news: the Yin and Yang of immune privilege in the eye. Front Immunol 2012; 3:338. [PMID: 23230433 PMCID: PMC3515883 DOI: 10.3389/fimmu.2012.00338] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/23/2012] [Indexed: 12/27/2022] Open
Abstract
The eye and the brain are prototypical tissues manifesting immune privilege (IP) in which immune responses to foreign antigens, particularly alloantigens are suppressed, and even completely inhibited. Explanations for this phenomenon are numerous and mostly reflect our evolving understanding of the molecular and cellular processes underpinning immunological responses generally. IP is now viewed as a property of many tissues and the level of expression of IP varies not only with the tissue but with the nature of the foreign antigen and changes in the limited conditions under which privilege can operate as a mechanism of immunological tolerance. As a result, IP functions normally as a homeostatic mechanism preserving normal function in tissues, particularly those with highly specialized function and limited capacity for renewal such as the eye and brain. However, IP is relatively easily bypassed in the face of a sufficiently strong immunological response, and the privileged tissues may be at greater risk of collateral damage because its natural defenses are more easily breached than in a fully immunocompetent tissue which rapidly rejects foreign antigen and restores integrity. This two-edged sword cuts its swathe through the eye: under most circumstances, IP mechanisms such as blood-ocular barriers, intraocular immune modulators, induction of T regulatory cells, lack of lymphatics, and other properties maintain tissue integrity; however, when these are breached, various degrees of tissue damage occur from severe tissue destruction in retinal viral infections and other forms of uveoretinal inflammation, to less severe inflammatory responses in conditions such as macular degeneration. Conversely, ocular IP and tumor-related IP can combine to permit extensive tumor growth and increased risk of metastasis thus threatening the survival of the host.
Collapse
Affiliation(s)
- John V. Forrester
- Laboratory of Immunology, Lion’s Eye Institute, University of Western AustraliaPerth, WA, Australia
- Ocular Immunology Laboratory, Section of Immunology and Infection, Institute of Medical Sciences, University of AberdeenAberdeen, UK
| | - Heping Xu
- Laboratory of Immunology, Lion’s Eye Institute, University of Western AustraliaPerth, WA, Australia
| |
Collapse
|
43
|
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: 2.7] [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
|
44
|
Sahni JN, Angi M, Irigoyen C, Semeraro F, Romano MR, Parmeggiani F. Therapeutic challenges to retinitis pigmentosa: from neuroprotection to gene therapy. Curr Genomics 2012; 12:276-84. [PMID: 22131873 PMCID: PMC3131735 DOI: 10.2174/138920211795860062] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 04/08/2011] [Accepted: 04/18/2011] [Indexed: 12/12/2022] Open
Abstract
Syndromic retinitis pigmentosa (RP) is the result of several mutations expressed in rod photoreceptors, over 40 of which have so far been identified. Enormous efforts are being made to relate the advances in unraveling the patho-physiological mechanisms to therapeutic approaches in animal models, and eventually in clinical trials on humans. This review summarizes briefly the current clinical management of RP and focuses on the new exciting treatment possibilities. To date, there is no approved therapy able to stop the evolution of RP or restore vision. The current management includes an attempt at slowing down the degenerative process by vitamin supplementation, trying to treat ocular complications and to provide psychological support to blind patients. Novel therapeutic may be tailored dependant on the stage of the disease and can be divided in three groups. In the early stages, when there are surviving photoreceptors, the first approach would be to try to halt the degeneration by correction of the underlying biochemical abnormality in the visual cycle using gene therapy or pharmacological treatment. A second approach aims to cope with photoreceptor cell death using neurotrophic growth factors or anti-apoptotic factors, reducing the production of retino-toxic molecules, and limiting oxidative damage. In advanced stages, when there are few or no functional photoreceptors, strategies that may benefit include retinal transplantation, electronic retinal implants or a newly described optogenetic technique using a light-activated channel to genetically resensitize remnant cone-photoreceptor cells.
Collapse
Affiliation(s)
- Jayashree N Sahni
- St. Paul's Eye Unit, Royal Liverpool University Hospital, Liverpool, UK
| | | | | | | | | | | |
Collapse
|
45
|
Nita M, Strzałka-Mrozik B, Grzybowski A, Romaniuk W, Mazurek U. Ophthalmic transplantology: posterior segment of the eye--part II. Med Sci Monit 2012; 18:RA97-103. [PMID: 22648265 PMCID: PMC3560715 DOI: 10.12659/msm.882868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Transplants of the retina are among the new strategies being used in the treatment of genetic and degenerative macular diseases. Moreover, various cell cultures are being tested to treat retinal disorders. Material/Methods Literature dated from 2004 to 2011 was comprehensively examined via Medline and PubMed searches for the following terms: auto-, homo-, heterologous transplantation, retina, stem cells, cultivated cells. Results Tissue and cell therapy of retinal diseases are reviewed, including full-thickness retina/retinal pigment epithelium (RPE)/choroid graft; full and partial thickness RPE/choroid complex grafts; RPE/Bruch membrane complex graft; and RPE, iris pigment epithelium and stem cell grafts. Recommendations for transplants, as well as the benefits and weaknesses of specific techniques in retina transplants, are discussed. Conclusions Auto- and allogenic transplants of a full or partial thickness retina/RPE/Bruch membrane/choroid complex represent an alternative treatment offered to patients with some macular diseases. Stem cell transplantation to reconstruct and regenerate the macula requires further biomolecular and animal research studies.
Collapse
Affiliation(s)
- Małgorzata Nita
- Domestic and Specialized Medicine Centre Dilmed, Katowice, Poland
| | | | | | | | | |
Collapse
|
46
|
Sundaram V, Moore AT, Ali RR, Bainbridge JW. Retinal dystrophies and gene therapy. Eur J Pediatr 2012; 171:757-65. [PMID: 22080959 DOI: 10.1007/s00431-011-1615-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 10/18/2011] [Indexed: 12/11/2022]
Abstract
Retinal dystrophies are inherited disorders of photoreceptor and retinal pigment epithelial function that may result in severe visual impairment. Advances in molecular genetics have helped identify many of the gene defects responsible, and progress in gene transfer technology has enabled therapeutic strategies to be developed and applied. The first human clinical trials of gene therapy for RPE65 associated retinal dystrophy have shown promising initial results and have helped prepare the way for further trials of gene therapy for inherited retinal disorders. The results of these trials will provide further insight into the safety and efficacy of gene therapy for a range of currently untreatable and debilitating eye disorders.
Collapse
Affiliation(s)
- Venki Sundaram
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | | | | | | |
Collapse
|
47
|
Mazumder MAJ, Fitzpatrick SD, Muirhead B, Sheardown H. Cell-adhesive thermogelling PNIPAAm/hyaluronic acid cell delivery hydrogels for potential application as minimally invasive retinal therapeutics. J Biomed Mater Res A 2012; 100:1877-87. [DOI: 10.1002/jbm.a.34021] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Revised: 10/28/2011] [Accepted: 11/07/2011] [Indexed: 01/28/2023]
|
48
|
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: 1.5] [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
|
49
|
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: 0.6] [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
|
50
|
Amirpour N, Karamali F, Rabiee F, Rezaei L, Esfandiari E, Razavi S, Dehghani A, Razmju H, Nasr-Esfahani MH, Baharvand H. Differentiation of Human Embryonic Stem Cell–Derived Retinal Progenitors into Retinal Cells by Sonic Hedgehog and/or Retinal Pigmented Epithelium and Transplantation into the Subretinal Space of Sodium Iodate–Injected Rabbits. Stem Cells Dev 2012; 21:42-53. [DOI: 10.1089/scd.2011.0073] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Noushin Amirpour
- Department of Cell and Molecular Biology, Royan Institute for Animal Biotechnology, Isfahan, Iran
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Fereshteh Karamali
- Department of Cell and Molecular Biology, Royan Institute for Animal Biotechnology, Isfahan, Iran
| | - Farzaneh Rabiee
- Department of Cell and Molecular Biology, Royan Institute for Animal Biotechnology, Isfahan, Iran
| | - Leila Rezaei
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Shahnaz Razavi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Alireza Dehghani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Hassan Razmju
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | | | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|