1
|
Sokolova N, Zilova L, Wittbrodt J. Unravelling the link between embryogenesis and adult stem cell potential in the ciliary marginal zone: A comparative study between mammals and teleost fish. Cells Dev 2023; 174:203848. [PMID: 37172718 DOI: 10.1016/j.cdev.2023.203848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
The discovery and study of adult stem cells have revolutionized regenerative medicine by offering new opportunities for treating various medical conditions. Anamniote stem cells, which retain their full proliferative capacity and full differentiation range throughout their lifetime, harbour a greater potential compared to mammalian adult stem cells, which only exhibit limited stem cell potential. Therefore, understanding the mechanisms underlying these differences is of significant interest. In this review, we examine the similarities and differences of adult retinal stem cells in anamniotes and mammals, from their embryonic stages in the optic vesicle to their residence in the postembryonic retinal stem cell niche, the ciliary marginal zone located in the retinal periphery. In anamniotes, developing precursors of retinal stem cells are exposed to various environmental cues during their migration in the complex morphogenetic remodelling of the optic vesicle to the optic cup. In contrast, their mammalian counterparts in the retinal periphery are primarily instructed by neighbouring tissues once they are in place. We explore the distinct modes of optic cup morphogenesis in mammals and teleost fish and highlight molecular mechanisms governing morphogenesis and stem cells instruction. The review concludes with the molecular mechanisms of ciliary marginal zone formation and offers a perspective on the impact of comparative single cell transcriptomic studies to reveal the evolutionary similarities and differences.
Collapse
Affiliation(s)
- Natalia Sokolova
- Centre for Organismal Studies Heidelberg, Germany; Heidelberg Biosciences International Graduate School, Germany
| | - Lucie Zilova
- Centre for Organismal Studies Heidelberg, Germany.
| | | |
Collapse
|
2
|
Olmos-Carreño CL, Figueres-Oñate M, Scicolone GE, López-Mascaraque L. Cell Fate of Retinal Progenitor Cells: In Ovo UbC-StarTrack Analysis. Int J Mol Sci 2022; 23:ijms232012388. [PMID: 36293245 PMCID: PMC9604099 DOI: 10.3390/ijms232012388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/28/2022] Open
Abstract
Clonal cell analysis outlines the ontogenic potential of single progenitor cells, allowing the elucidation of the neural heterogeneity among different cell types and their lineages. In this work, we analyze the potency of retinal stem/progenitor cells through development using the chick embryo as a model. We implemented in ovo the clonal genetic tracing strategy UbC-StarTrack for tracking retinal cell lineages derived from individual progenitors of the ciliary margin at E3.5 (HH21-22). The clonal assignment of the derived-cell progeny was performed in the neural retina at E11.5-12 (HH38) through the identification of sibling cells as cells expressing the same combination of fluorophores. Moreover, cell types were assessed based on their cellular morphology and laminar location. Ciliary margin derived-cell progenies are organized in columnar associations distributed along the peripheral retina with a limited tangential dispersion. The analysis revealed that, at the early stages of development, this region harbors multipotent and committed progenitor cells.
Collapse
Affiliation(s)
- Cindy L. Olmos-Carreño
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN), CONICET and Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, 28002 Madrid, Spain
| | - María Figueres-Oñate
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, 28002 Madrid, Spain
- Correspondence: (M.F.-O.); (L.L.-M.)
| | - Gabriel E. Scicolone
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN), CONICET and Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
| | - Laura López-Mascaraque
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, 28002 Madrid, Spain
- Correspondence: (M.F.-O.); (L.L.-M.)
| |
Collapse
|
3
|
Todd L, Reh TA. Comparative Biology of Vertebrate Retinal Regeneration: Restoration of Vision through Cellular Reprogramming. Cold Spring Harb Perspect Biol 2022; 14:a040816. [PMID: 34580118 PMCID: PMC9248829 DOI: 10.1101/cshperspect.a040816] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The regenerative capacity of the vertebrate retina varies substantially across species. Whereas fish and amphibians can regenerate functional retina, mammals do not. In this perspective piece, we outline the various strategies nonmammalian vertebrates use to achieve functional regeneration of vision. We review key differences underlying the regenerative potential across species including the cellular source of postnatal progenitors, the diversity of cell fates regenerated, and the level of functional vision that can be achieved. Finally, we provide an outlook on the field of engineering the mammalian retina to replace neurons lost to injury or disease.
Collapse
Affiliation(s)
- Levi Todd
- Department of Biological Structure, University of Washington, Seattle, Washington 98195, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
4
|
Zibetti C. Deciphering the Retinal Epigenome during Development, Disease and Reprogramming: Advancements, Challenges and Perspectives. Cells 2022; 11:cells11050806. [PMID: 35269428 PMCID: PMC8908986 DOI: 10.3390/cells11050806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Retinal neurogenesis is driven by concerted actions of transcription factors, some of which are expressed in a continuum and across several cell subtypes throughout development. While seemingly redundant, many factors diversify their regulatory outcome on gene expression, by coordinating variations in chromatin landscapes to drive divergent retinal specification programs. Recent studies have furthered the understanding of the epigenetic contribution to the progression of age-related macular degeneration, a leading cause of blindness in the elderly. The knowledge of the epigenomic mechanisms that control the acquisition and stabilization of retinal cell fates and are evoked upon damage, holds the potential for the treatment of retinal degeneration. Herein, this review presents the state-of-the-art approaches to investigate the retinal epigenome during development, disease, and reprogramming. A pipeline is then reviewed to functionally interrogate the epigenetic and transcriptional networks underlying cell fate specification, relying on a truly unbiased screening of open chromatin states. The related work proposes an inferential model to identify gene regulatory networks, features the first footprinting analysis and the first tentative, systematic query of candidate pioneer factors in the retina ever conducted in any model organism, leading to the identification of previously uncharacterized master regulators of retinal cell identity, such as the nuclear factor I, NFI. This pipeline is virtually applicable to the study of genetic programs and candidate pioneer factors in any developmental context. Finally, challenges and limitations intrinsic to the current next-generation sequencing techniques are discussed, as well as recent advances in super-resolution imaging, enabling spatio-temporal resolution of the genome.
Collapse
Affiliation(s)
- Cristina Zibetti
- Department of Ophthalmology, Institute of Clinical Medicine, University of Oslo, Kirkeveien 166, Building 36, 0455 Oslo, Norway
| |
Collapse
|
5
|
Sherpa RD, Hui SP. An insight on established retinal injury mechanisms and prevalent retinal stem cell activation pathways in vertebrate models. Animal Model Exp Med 2021; 4:189-203. [PMID: 34557646 PMCID: PMC8446703 DOI: 10.1002/ame2.12177] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/09/2021] [Indexed: 12/22/2022] Open
Abstract
Implementing different tools and injury mechanisms in multiple animal models of retina regeneration, researchers have discovered the existence of retinal stem/progenitor cells. Although they appear to be distributed uniformly across the vertebrate lineage, the reparative potential of the retina is mainly restricted to lower vertebrates. Regenerative repair post-injury requires the creation of a proliferative niche, vital for proper stem cell activation, propagation, and lineage differentiation. This seems to be lacking in mammals. Hence, in this review, we first discuss the many forms of retinal injuries that have been generated using animal models. Next, we discuss how they are utilized to stimulate regeneration and mimic eye disease pathologies. The key to driving stem cell activation in mammals relies on the information we can gather from these models. Lastly, we present a brief update about the genes, growth factors, and signaling pathways that have been brought to light using these models.
Collapse
Affiliation(s)
| | - Subhra Prakash Hui
- S. N. Pradhan Centre for NeurosciencesUniversity of CalcuttaKolkataIndia
| |
Collapse
|
6
|
Regulation of the Brain Neural Niche by Soluble Molecule Akhirin. J Dev Biol 2021; 9:jdb9030029. [PMID: 34449638 PMCID: PMC8395899 DOI: 10.3390/jdb9030029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 01/22/2023] Open
Abstract
In the central nervous system (CNS), which comprises the eyes, spinal cord, and brain, neural cells are produced by the repeated division of neural stem cells (NSCs) during the development of the CNS. Contrary to the notion that the CNS is relatively static with a limited cell turnover, cells with stem cell-like properties have been isolated from most neural tissues. The microenvironment, also known as the NSC niche, consists of NSCs/neural progenitor cells, other neurons, glial cells, and blood vessels; this niche is thought to regulate neurogenesis and the differentiation of NSCs into neurons and glia. Although it has been established that neurons, glia, and blood vessels interact with each other in a complex manner to generate neural tissues in the NSC niche, the underlying molecular mechanisms in the CNS niche are unclear. Herein, we would like to introduce the extracellular secreted protein, Akhirin (AKH; Akhi is the Bengali translation for eye). AKH is specifically expressed in the CNS niche-the ciliary body epithelium in the retina, the central canal of the spinal cord, the subventricular zone, and the subgranular zone of the dentate gyrus of the hippocampus-and is supposedly involved in NSC niche regulation. In this review, we discuss the role of AKH as a niche molecule during mouse brain formation.
Collapse
|
7
|
Prospects for the application of Müller glia and their derivatives in retinal regenerative therapies. Prog Retin Eye Res 2021; 85:100970. [PMID: 33930561 DOI: 10.1016/j.preteyeres.2021.100970] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023]
Abstract
Neural cell death is the main feature of all retinal degenerative disorders that lead to blindness. Despite therapeutic advances, progression of retinal disease cannot always be prevented, and once neuronal cell damage occurs, visual loss cannot be reversed. Recent research in the stem cell field, and the identification of Müller glia with stem cell characteristics in the human eye, have provided hope for the use of these cells in retinal therapies to restore vision. Müller glial cells, which are the major structural cells of the retina, play a very important role in retinal homeostasis during health and disease. They are responsible for the spontaneous retinal regeneration observed in zebrafish and lower vertebrates during early postnatal life, and despite the presence of Müller glia with stem cell characteristics in the adult mammalian retina, there is no evidence that they promote regeneration in humans. Like many other stem cells and neurons derived from pluripotent stem cells, Müller glia with stem cell potential do not differentiate into retinal neurons or integrate into the retina when transplanted into the vitreous of experimental animals with retinal degeneration. However, despite their lack of integration, grafted Müller glia have been shown to induce partial restoration of visual function in spontaneous or induced experimental models of photoreceptor or retinal ganglion cell damage. This improvement in visual function observed after Müller cell transplantation has been ascribed to the release of neuroprotective factors that promote the repair and survival of damaged neurons. Due to the development and availability of pluripotent stem cell lines for therapeutic uses, derivation of Müller cells from retinal organoids formed by iPSC and ESC has provided more realistic prospects for the application of these cells to retinal therapies. Several opportunities for research in the regenerative field have also been unlocked in recent years due to a better understanding of the genomic and proteomic profiles of the developing and regenerating retina in zebrafish, providing the basis for further studies of the human retina. In addition, the increased interest on the nature and function of cellular organelle release and the characterization of molecular components of exosomes released by Müller glia, may help us to design new approaches that could be applied to the development of more effective treatments for retinal degenerative diseases.
Collapse
|
8
|
Völkner M, Kurth T, Schor J, Ebner LJA, Bardtke L, Kavak C, Hackermüller J, Karl MO. Mouse Retinal Organoid Growth and Maintenance in Longer-Term Culture. Front Cell Dev Biol 2021; 9:645704. [PMID: 33996806 PMCID: PMC8114082 DOI: 10.3389/fcell.2021.645704] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
Using retinal organoid systems, organ-like 3D tissues, relies implicitly on their robustness. However, essential key parameters, particularly retinal growth and longer-term culture, are still insufficiently defined. Here, we hypothesize that a previously optimized protocol for high yield of evenly-sized mouse retinal organoids with low variability facilitates assessment of such parameters. We demonstrate that these organoids reliably complete retinogenesis, and can be maintained at least up to 60 days in culture. During this time, the organoids continue to mature on a molecular and (ultra)structural level: They develop photoreceptor outer segments and synapses, transiently maintain its cell composition for about 5-10 days after completing retinogenesis, and subsequently develop pathologic changes - mainly of the inner but also outer retina and reactive gliosis. To test whether this organoid system provides experimental access to the retina during and upon completion of development, we defined and stimulated organoid growth by activating sonic hedgehog signaling, which in patients and mice in vivo with a congenital defect leads to enlarged eyes. Here, a sonic hedgehog signaling activator increased retinal epithelia length in the organoid system when applied during but not after completion of development. This experimentally supports organoid maturation, stability, and experimental reproducibility in this organoid system, and provides a potential enlarged retina pathology model, as well as a protocol for producing larger organoids. Together, our study advances the understanding of retinal growth, maturation, and maintenance, and further optimizes the organoid system for future utilization.
Collapse
Affiliation(s)
- Manuela Völkner
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering, Technology Platform, Electron Microscopy and Histology Facility, Technische Universität Dresden, Dresden, Germany
| | - Jana Schor
- Young Investigators Group Bioinformatics and Transcriptomics, Department Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Lynn J A Ebner
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lara Bardtke
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Cagri Kavak
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Jörg Hackermüller
- Young Investigators Group Bioinformatics and Transcriptomics, Department Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Mike O Karl
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.,CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Grisé KN, Bautista NX, Jacques K, Coles BLK, van der Kooy D. Glucocorticoid agonists enhance retinal stem cell self-renewal and proliferation. Stem Cell Res Ther 2021; 12:83. [PMID: 33494791 PMCID: PMC7831262 DOI: 10.1186/s13287-021-02136-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/01/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Adult mammalian retinal stem cells (RSCs) readily proliferate, self-renew, and generate progeny that differentiate into all retinal cell types in vitro. RSC-derived progeny can be induced to differentiate into photoreceptors, making them a potential source for retinal cell transplant therapies. Despite their proliferative propensity in vitro, RSCs in the adult mammalian eye do not proliferate and do not have a regenerative response to injury. Thus, identifying and modulating the mechanisms that regulate RSC proliferation may enhance the capacity to produce RSC-derived progeny in vitro and enable RSC activation in vivo. METHODS Here, we used medium-throughput screening to identify small molecules that can expand the number of RSCs and their progeny in culture. In vitro differentiation assays were used to assess the effects of synthetic glucocorticoid agonist dexamethasone on RSC-derived progenitor cell fate. Intravitreal injections of dexamethasone into adult mouse eyes were used to investigate the effects on endogenous RSCs. RESULTS We discovered that high-affinity synthetic glucocorticoid agonists increase RSC self-renewal and increase retinal progenitor proliferation up to 6-fold without influencing their differentiation in vitro. Intravitreal injection of synthetic glucocorticoid agonist dexamethasone induced in vivo proliferation in the ciliary epithelium-the niche in which adult RSCs reside. CONCLUSIONS Together, our results identify glucocorticoids as novel regulators of retinal stem and progenitor cell proliferation in culture and provide evidence that GCs may activate endogenous RSCs.
Collapse
Affiliation(s)
- Kenneth N Grisé
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada.
| | - Nelson X Bautista
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Krystal Jacques
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Brenda L K Coles
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|
10
|
Eymann J, Di-Poï N. Glia-Mediated Regenerative Response Following Acute Excitotoxic Damage in the Postnatal Squamate Retina. Front Cell Dev Biol 2020; 8:406. [PMID: 32548121 PMCID: PMC7270358 DOI: 10.3389/fcell.2020.00406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/04/2020] [Indexed: 01/13/2023] Open
Abstract
The retina is a complex tissue responsible for both detection and primary processing of visual stimuli. Although all vertebrate retinas share a similar, multi-layered organization, the ability to regenerate individual retinal cells varies tremendously, being extremely limited in mammals and birds when compared to anamniotes such as fish and amphibians. However, little is yet known about damage response and regeneration of retinal tissues in "non-classical" squamate reptiles (lizards, snakes), which occupy a key phylogenetic position within amniotes and exhibit unique regenerative features in many tissues. Here, we address this gap by establishing and characterizing a model of excitotoxic retinal damage in bearded dragon lizard (Pogona vitticeps). We particularly focus on identifying, at the cellular and molecular level, a putative endogenous cellular source for retinal regeneration, as diverse self-repair strategies have been characterized in vertebrates using a variety of retinal injury and transgenic models. Our findings reveal for the first time that squamates hold the potential for postnatal retinal regeneration following acute injury. Although no changes occur in the activity of physiologically active progenitors recently identified at the peripheral retinal margin of bearded dragon, two distinct successive populations of proliferating cells at central retina respond to neurotoxin treatment. Following an initial microglia response, a second source of proliferating cells exhibit common hallmarks of vertebrate Müller glia (MG) activation, including cell cycle re-entry, dedifferentiation into a progenitor-like phenotype, and re-expression of proneural markers. The observed lizard glial responses, although not as substantial as in anamniotes, appear more robust than the absent or neonatal-limited regeneration reported without exogenous stimulation in other amniotes. Altogether, these results help to complete our evolutionary understanding of regenerative potential of the vertebrate retina, and further highlight the major importance of glial cells in retinal regeneration. Furthermore, our work offers a new powerful vertebrate model to elucidate the developmental and evolutionary bases of retinal regeneration within amniotes. Such new understanding of self-repair mechanisms in non-classical species endowed with regenerative properties may help designing therapeutic strategies for vertebrate retinal diseases.
Collapse
Affiliation(s)
- Julia Eymann
- Research Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nicolas Di-Poï
- Research Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Abstract
Retinal degeneration is a leading cause of untreatable blindness in the industrialised world. It is typically irreversible and there are few curative treatments available. The use of stem cells to generate new retinal neurons for transplantation purposes has received significant interest in recent years and is beginning to move towards clinical trials. However, such approaches are likely to be most effective for relatively focal areas of repair. An intriguing complementary approach is endogenous self-repair. Retinal cells from the ciliary marginal zone (CMZ), retinal pigment epithelium (RPE) and Müller glial cells (MG) have all been shown to play a role in retinal repair, typically in lower vertebrates. Among them, MG have received renewed interest, due to their distribution throughout (centre to periphery) the neural retina and their potential to re-acquire a progenitor-like state following retinal injury with the ability to proliferate and generate new neurons. Triggering these innate self-repair mechanisms represents an exciting therapeutic option in treating retinal degeneration. However, these cells behave differently in mammalian and non-mammalian species, with a considerably restricted potential in mammals. In this short review, we look at some of the recent progress made in our understanding of the signalling pathways that underlie MG-mediated regeneration in lower vertebrates, and some of the challenges that have been revealed in our attempts to reactivate this process in the mammalian retina.
Collapse
Affiliation(s)
- Rahul Langhe
- Institute of Ophthalmology, University College London, London, UK
| | | |
Collapse
|
12
|
Eymann J, Salomies L, Macrì S, Di-Poï N. Variations in the proliferative activity of the peripheral retina correlate with postnatal ocular growth in squamate reptiles. J Comp Neurol 2019; 527:2356-2370. [PMID: 30860599 PMCID: PMC6766921 DOI: 10.1002/cne.24677] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/26/2022]
Abstract
The retina is a complex, multilayered tissue responsible for the perception of visual stimuli from the environment. Contrary to mammals, the capacity for postnatal eye growth in fish and amphibians, and to a lower extent in birds, is coordinated with a progenitor population residing in the ciliary marginal zone (CMZ) at the retinal peripheral margin. However, little is known about embryonic retinogenesis and postnatal retinal growth in squamates (lizards, snakes), despite their exceptional array of ecologies and ocular morphologies. Here, we address this gap by performing the first large‐scale study assessing both ontogenetic and adult changes in the stem/progenitor activity of the squamate peripheral retina. Our study reveals for the first time that squamates exhibit a source of proliferating progenitors persisting post embryogenesis in a newly identified retinociliary junction anteriorly adjacent to the retina. This region is strikingly similar to the vertebrate CMZ by its peripheral location and pseudostratified nature, and shares a common pattern of slow‐cycling cells, spatial differentiation gradient, and response to postnatal ocular growth. Additionally, its proliferative activity varies considerably among squamate species, in correlation with embryonic and postnatal differences in eye size and growth. Together our data indicate that squamates possess a proliferative peripheral retina that acts as a source of progenitors to compensate, at least in part, for postnatal ocular growth. Our findings also highlight the remarkable variation in activity and location of vertebrate retinal progenitors, indicating that the currently accepted scenario of reduced CMZ activity over the course of evolution is too simplistic.
Collapse
Affiliation(s)
- Julia Eymann
- Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Lotta Salomies
- Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Simone Macrì
- Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nicolas Di-Poï
- Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Abstract
Purpose Retinal degenerative diseases lead to the death of retinal neurons causing visual impairment and blindness. In lower order vertebrates, the retina and its surrounding tissue contain stem cell niches capable of regenerating damaged tissue. Here we examine these niches and review their capacity to be used as retinal stem/progenitor cells (RSC/RPCs) for retinal repair. Recent Findings Exogenous factors can control the in vitro activation of RSCs/PCs found in several niches within the adult eye including cells in the ciliary margin, the retinal pigment epithelium, iris pigment epithelium as well as the inducement of Müller and amacrine cells within the neural retina itself. Recently, factors have been identified for the activation of adult mammalian Müller cells to a RPC state in vivo. Summary Whereas cell transplantation still holds potential for retinal repair, activation of the dormant native regeneration process may lead to a more successful process including greater integration efficiency and proper synaptic targeting.
Collapse
|
14
|
da Silva BR, Santos LE, de Melo Reis RA, de Mello FG, Ribeiro-Resende VT. Müller Cells Derived from Adult Chicken and Mouse Retina Neurospheres Acquire the Dopaminergic Phenotype. Cell Mol Neurobiol 2019; 39:99-109. [PMID: 30430378 DOI: 10.1007/s10571-018-0636-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/08/2018] [Indexed: 10/27/2022]
Abstract
Neurospheres prepared from multipotent progenitors in the retina obtained from postnatal mice differentiate into neurons and Müller glia (De Melo Reis et al., in Cell Mol Neurobiol 31:835-846, 2011). Here, we investigated whether neurospheres prepared from adult chickens (ciliary marginal zone, CMZ) or (ciliary body) retina could also lead to differentiated neurons and glia. Neurospheres were prepared from post-hatched chickens or from adult mice after 7 days in the presence of mitogenic factors (FGFb, insulin, and EGF), generating neurons and glial cells. In addition, Müller (2M6 or glutamine synthetase positive cells) derived from post-hatch chicken CMZ neurospheres displayed the dopaminergic phenotype. Furthermore, we observed that Müller cells derived from adult chickens and mice retina neurospheres released significant amounts of dopamine as well as of its metabolites. Taken together, our data lead us to conclude that as for embryonic (chick) or newborn (mouse), the dopaminergic phenotype is a default condition of Müller glial cells obtained from neurospheres prepared from mature retina. Our data raise the possibility that Müller cells from differentiated tissue could be used to ameliorate neurodegenerative diseases involving dopaminergic dysfunction as in Parkinson's disease as shown previously (Stutz et al., in J Neurochem 128:829-840, 2014).
Collapse
Affiliation(s)
- Bárbara Rangel da Silva
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Luis Eduardo Santos
- Laboratório de Doenças Neurodegenerativas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, CCS, Sala C1-31, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Ricardo A de Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Fernando Garcia de Mello
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Victor T Ribeiro-Resende
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil.
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ, 25255-030, Brazil.
| |
Collapse
|
15
|
Marcucci F, Murcia-Belmonte V, Wang Q, Coca Y, Ferreiro-Galve S, Kuwajima T, Khalid S, Ross ME, Mason C, Herrera E. The Ciliary Margin Zone of the Mammalian Retina Generates Retinal Ganglion Cells. Cell Rep 2017; 17:3153-3164. [PMID: 28009286 DOI: 10.1016/j.celrep.2016.11.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/23/2016] [Accepted: 11/01/2016] [Indexed: 10/20/2022] Open
Abstract
The retina of lower vertebrates grows continuously by integrating new neurons generated from progenitors in the ciliary margin zone (CMZ). Whether the mammalian CMZ provides the neural retina with retinal cells is controversial. Live imaging of embryonic retina expressing eGFP in the CMZ shows that cells migrate laterally from the CMZ to the neural retina where differentiated retinal ganglion cells (RGCs) reside. Because Cyclin D2, a cell-cycle regulator, is enriched in ventral CMZ, we analyzed Cyclin D2-/- mice to test whether the CMZ is a source of retinal cells. Neurogenesis is diminished in Cyclin D2 mutants, leading to a reduction of RGCs in the ventral retina. In line with these findings, in the albino retina, the decreased production of ipsilateral RGCs is correlated with fewer Cyclin D2+ cells. Together, these results implicate the mammalian CMZ as a neurogenic site that produces RGCs and whose proper generation depends on Cyclin D2 activity.
Collapse
Affiliation(s)
- Florencia Marcucci
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Veronica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain
| | - Qing Wang
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yaiza Coca
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain
| | - Susana Ferreiro-Galve
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain
| | - Takaaki Kuwajima
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sania Khalid
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain & Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, USA
| | - Carol Mason
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | - Eloisa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain.
| |
Collapse
|
16
|
Gu D, Wang S, Zhang S, Zhang P, Zhou G. Directed transdifferentiation of Müller glial cells to photoreceptors using the sonic hedgehog signaling pathway agonist purmorphamine. Mol Med Rep 2017; 16:7993-8002. [PMID: 28983586 PMCID: PMC5779882 DOI: 10.3892/mmr.2017.7652] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 07/28/2017] [Indexed: 01/08/2023] Open
Abstract
Specification of distinct cell types from Müller glial cells is key to the potential application of endogenous repair in retinal regeneration. Sonic hedgehog (SHH) has been established as a potent mitogen for rat Müller glial cells, which also induces Müller glial cells to dedifferentiate and adopt the phenotype of rod photoreceptors. The present study investigated the effects of purmorphamine, a small molecule that activates the SHH‑pathway, in the proliferation, dedifferentiation and transdifferentiation of Müller glial cells, as determined by several methods including immunofluorescence, polymerase chain reaction and western blotting. It was demonstrated that it may be able to replace SHH for the regeneration of retinal neurons. Purmorphamine was revealed to stimulate the proliferation of Müller glial cells by increasing the expression of cyclin D1 and cyclin D3. In addition, purmorphamine‑treated Müller glial cells were induced to dedifferentiate by inducing the expression of progenitor‑specific markers; subsequently differentiating into rod‑like photoreceptors. Intraocular injection of purmorphamine promoted the activation of Müller glial cells, and in turn, the production of rod‑like photoreceptors in acute damaged retina. These results suggested that the endogenous neurogenic capacity of retinal Müller glial cells may be enhanced by this small molecular agonist of the SHH signaling pathway.
Collapse
Affiliation(s)
- Dandan Gu
- Department of Anatomy, Histology and Embryology, Institute of Acupuncture Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Songtao Wang
- Department of Integrative Medicine and Neurobiology, Institute of Acupuncture Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Shuai Zhang
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Fudan University, Shanghai 200032, P.R. China
| | - Peng Zhang
- Department of Integrative Medicine and Neurobiology, Institute of Acupuncture Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Guomin Zhou
- Department of Anatomy, Histology and Embryology, Institute of Acupuncture Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
17
|
Tang X, Gao J, Jia X, Zhao W, Zhang Y, Pan W, He J. Bipotent progenitors as embryonic origin of retinal stem cells. J Cell Biol 2017; 216:1833-1847. [PMID: 28465291 PMCID: PMC5461025 DOI: 10.1083/jcb.201611057] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/01/2017] [Accepted: 04/03/2017] [Indexed: 01/24/2023] Open
Abstract
In lower vertebrates, retinal stem cells (RSCs) capable of producing all retinal cell types are a resource for retinal tissue growth throughout life. However, the embryonic origin of RSCs remains largely elusive. Using a Zebrabow-based clonal analysis, we characterized the RSC niche in the ciliary marginal zone of zebrafish retina and illustrate that blood vessels associated with RSCs are required for the maintenance of actively proliferating RSCs. Full lineage analysis of RSC progenitors reveals lineage patterns of RSC production. Moreover, in vivo lineage analysis demonstrates that these RSC progenitors are the direct descendants of a set of bipotent progenitors in the medial epithelial layer of developing optic vesicles, suggesting the involvement of the mixed-lineage states in the RSC lineage specification.
Collapse
Affiliation(s)
- Xia Tang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianan Gao
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinling Jia
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wencao Zhao
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Yijie Zhang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weijun Pan
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Jie He
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
18
|
Chen X, Chen Z, Li Z, Zhao C, Zeng Y, Zou T, Fu C, Liu X, Xu H, Yin ZQ. Grafted c-kit +/SSEA1 - eye-wall progenitor cells delay retinal degeneration in mice by regulating neural plasticity and forming new graft-to-host synapses. Stem Cell Res Ther 2016; 7:191. [PMID: 28038685 PMCID: PMC5203726 DOI: 10.1186/s13287-016-0451-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/25/2016] [Accepted: 12/06/2016] [Indexed: 12/12/2022] Open
Abstract
Background Despite diverse pathogenesis, the common pathological change observed in age-related macular degeneration and in most hereditary retinal degeneration (RD) diseases is photoreceptor loss. Photoreceptor replacement by cell transplantation may be a feasible treatment for RD. The major obstacles to clinical translation of stem cell-based cell therapy in RD remain the difficulty of obtaining sufficient quantities of appropriate and safe donor cells and the poor integration of grafted stem cell-derived photoreceptors into the remaining retinal circuitry. Methods Eye-wall c-kit+/stage-specific embryonic antigen 1 (SSEA1)− cells were isolated via fluorescence-activated cell sorting, and their self-renewal and differentiation potential were detected by immunochemistry and flow cytometry in vitro. After labeling with quantum nanocrystal dots and transplantation into the subretinal space of rd1 RD mice, differentiation and synapse formation by daughter cells of the eye-wall c-kit+/SSEA1− cells were evaluated by immunochemistry and western blotting. Morphological changes of the inner retina of rd1 mice after cell transplantation were demonstrated by immunochemistry. Retinal function of rd1 mice that received cell grafts was tested via flash electroretinograms and the light/dark transition test. Results Eye-wall c-kit+/SSEA1− cells were self-renewing and clonogenic, and they retained their proliferative potential through more than 20 passages. Additionally, eye-wall c-kit+/SSEA1− cells were capable of differentiating into multiple retinal cell types including photoreceptors, bipolar cells, horizontal cells, amacrine cells, Müller cells, and retinal pigment epithelium cells and of transdifferentiating into smooth muscle cells and endothelial cells in vitro. The levels of synaptophysin and postsynaptic density-95 in the retinas of eye-wall c-kit+/SSEA1− cell-transplanted rd1 mice were significantly increased at 4 weeks post transplantation. The c-kit+/SSEA1− cells were capable of differentiating into functional photoreceptors that formed new synaptic connections with recipient retinas in rd1 mice. Transplantation also partially corrected the abnormalities of inner retina of rd1 mice. At 4 and 8 weeks post transplantation, the rd1 mice that received c-kit+/SSEA1− cells showed significant increases in a-wave and b-wave amplitude and the percentage of time spent in the dark area. Conclusions Grafted c-kit+/SSEA1− cells restored the retinal function of rd1 mice via regulating neural plasticity and forming new graft-to-host synapses. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0451-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xi Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.,School of Medicine, Nankai University, Tianjin, 300071, China.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zehua Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Zhengya Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Chen Zhao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Yuxiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Caiyun Fu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| |
Collapse
|
19
|
Bélanger MC, Robert B, Cayouette M. Msx1-Positive Progenitors in the Retinal Ciliary Margin Give Rise to Both Neural and Non-neural Progenies in Mammals. Dev Cell 2016; 40:137-150. [PMID: 28011038 DOI: 10.1016/j.devcel.2016.11.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 11/04/2016] [Accepted: 11/22/2016] [Indexed: 11/18/2022]
Abstract
In lower vertebrates, stem/progenitor cells located in a peripheral domain of the retina, called the ciliary margin zone (CMZ), cooperate with retinal domain progenitors to build the mature neural retina. In mammals, it is believed that the CMZ lacks neurogenic potential and that the retina develops from one pool of multipotent retinal progenitor cells (RPCs). Here we identify a population of Msx1-expressing progenitors in the mouse CMZ that is both molecularly and functionally distinct from RPCs. Using genetic lineage tracing, we report that Msx1 progenitors have unique developmental properties compared with RPCs. Msx1 lineages contain both neural retina and non-neural ciliary epithelial progenies and overall generate fewer photoreceptors than classical RPC lineages. Furthermore, we show that the endocytic adaptor protein Numb regulates the balance between neural and non-neural fates in Msx1 progenitors. These results uncover a population of CMZ progenitors, distinct from classical RPCs, that also contributes to mammalian retinogenesis.
Collapse
Affiliation(s)
- Marie-Claude Bélanger
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montreal, QC H3A 1A3, Canada
| | - Benoit Robert
- Department of Molecular Genetics of Morphogenesis, Institut Pasteur, Paris 75015, France
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montreal, QC H3A 1A3, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
20
|
Charng J, Cideciyan AV, Jacobson SG, Sumaroka A, Schwartz SB, Swider M, Roman AJ, Sheplock R, Anand M, Peden MC, Khanna H, Heon E, Wright AF, Swaroop A. Variegated yet non-random rod and cone photoreceptor disease patterns in RPGR-ORF15-associated retinal degeneration. Hum Mol Genet 2016; 25:5444-5459. [PMID: 27798110 PMCID: PMC6078602 DOI: 10.1093/hmg/ddw361] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/16/2016] [Accepted: 10/18/2016] [Indexed: 12/13/2022] Open
Abstract
Mutations in the ORF15 exon of the RPGR gene cause a common form of X-linked retinitis pigmentosa, which often results in severe loss of vision. In dogs and mice, gene augmentation therapy has been shown to arrest the progressive degeneration of rod and cone photoreceptors. However, the distribution of potentially treatable photoreceptors across the human retinas and the rate of degeneration are not known. Here, we have defined structural and functional features of the disease in 70 individuals with ORF15 mutations. We also correlated the features observed in patients with those of three Rpgr-mutant (Rpgr-ko, Rd9, and Rpgr-cko) mice. In patients, there was pronounced macular disease. Across the retina, rod and cone dysfunction showed a range of patterns and a spectrum of severity between individuals, but a high symmetry was observed between eyes of each individual. Genotype was not related to disease expression. In the Rpgr-ko mice, there were intra-retinal differences in rhodopsin and cone opsin trafficking. In Rd9 and Rpgr-cko mice, retinal degeneration showed inter-ocular symmetry. Longitudinal results in patients revealed localized rod and cone dysfunction with progression rates of 0.8 to 1.3 log per decade in sensitivity loss. Relatively retained rod and cone photoreceptors in mid- and far-peripheral temporal-inferior and nasal-inferior visual field regions should be good targets for future localized gene therapies in patients.
Collapse
Affiliation(s)
- Jason Charng
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Artur V. Cideciyan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Samuel G. Jacobson
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Alexander Sumaroka
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Sharon B. Schwartz
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Malgorzata Swider
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Alejandro J. Roman
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Rebecca Sheplock
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Manisha Anand
- Department of Ophthalmology, University of Massachusetts Medical School, Worcester, Massachusetts, MA, USA
| | - Marc C. Peden
- Retina Associates of Florida, Tampa, Florida, FL, USA
| | - Hemant Khanna
- Department of Ophthalmology, University of Massachusetts Medical School, Worcester, Massachusetts, MA, USA
| | - Elise Heon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Alan F. Wright
- MRC Human Genetics Unit, MRC IGMM, University of Edinburgh, Edinburgh, Scotland, UK
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, MD, USA
| |
Collapse
|
21
|
Richieri-Costa A, Vendramini-Pittoli S, Kokitsu-Nakata NM, Zechi-Ceide RM, Alvarez CW, Ribeiro-Bicudo LA. Multisystem Involvement in a Patient with a PTCH1 Mutation: Clinical and Imaging Findings. J Pediatr Genet 2016; 6:103-106. [PMID: 28496998 DOI: 10.1055/s-0036-1588028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 07/21/2016] [Indexed: 10/21/2022]
Abstract
In this article, we report on a Brazilian female patient born to consanguineous parents and presenting with alobar holoprosencephaly, severe eye involvement, and unusual skin hyperpigmented lesions. She was found to have a mutation (c.2240T > C; p.Val751Gly) in exon 15 of the PTCH1 gene. Mutations in this gene are associated with the nevoid basal cell carcinoma syndrome (NBCCS, OMIM 109400) and, in other instances, with holoprosencephaly (holoprosencephaly-7, OMIM 610828). Severe eye involvement ranging from orbital coloboma to microphthalmia has been seldom reported in patients with NBCCS with PTCH1 mutations. To our knowledge, this is the first report of an individual with central nervous system, skin, and eye manifestations due to a PTCH1 mutation. Mechanisms involved in these multisystem manifestations are discussed.
Collapse
Affiliation(s)
- Antonio Richieri-Costa
- Serviço de Genética, Hospital de Reabilitação de Anomalias Craniofaciais, Universidade de São Paulo, Bauru, Sao Paulo, Brazil
| | - Siulan Vendramini-Pittoli
- Serviço de Genética, Hospital de Reabilitação de Anomalias Craniofaciais, Universidade de São Paulo, Bauru, Sao Paulo, Brazil
| | - Nancy Mizue Kokitsu-Nakata
- Serviço de Genética, Hospital de Reabilitação de Anomalias Craniofaciais, Universidade de São Paulo, Bauru, Sao Paulo, Brazil
| | - Roseli Maria Zechi-Ceide
- Serviço de Genética, Hospital de Reabilitação de Anomalias Craniofaciais, Universidade de São Paulo, Bauru, Sao Paulo, Brazil
| | - Camila Wenceslau Alvarez
- Serviço de Genética, Hospital de Reabilitação de Anomalias Craniofaciais, Universidade de São Paulo, Bauru, Sao Paulo, Brazil
| | | |
Collapse
|
22
|
Chondrogenic cells respond to partial-thickness defects of articular cartilage in adult rats: an in vivo study. J Mol Histol 2016; 47:249-58. [PMID: 26956364 DOI: 10.1007/s10735-016-9668-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/04/2016] [Indexed: 01/11/2023]
Abstract
The purpose of this study was to establish a partial-thickness articular cartilage defects model in adult rats and explore the respond of chondrogenic cells to the cartilage injury. Forty-five adult Sprague-Dawley rats were divided into operated group, sham-operated group and control group. Partial-thickness cartilage defects were created on the weight-bearing region of femoral condyles by a converted ophthalmic knife. Rats were exposed to 5-bromo-2'-deoxyuridine (BrdU) for five consecutive days and were sacrificed 1, 2 and 4 weeks after surgery. Evaluations of macroscopic and histological changes were made. Chondrocyte apoptosis was evaluated by TUNEL assay. Immunofluorescence staining of CD105 and BrdU, double staining of CD105/integrin α5β1 and CD105-positive cells counting were performed for evaluations of cells around the defects. Cartilage softening and fibrillation with chondrocyte apoptosis were observed around the injury site after surgery. Results of histological scores indicated no significant difference between one time point and a successive time point for either group. CD105-positive cells and BrdU-label-retaining cells were observed around the linear injury. And cells counting showed the number of CD105-positive cells increased at later time points (P < 0.05). Immunofluorescence double staining demonstrated co-localization of CD105 and integrin α5β1 in activated cells around the defects. We establish a partial-thickness cartilage defects model in adult rats and demonstrate this injury may lead to activation of putative progenitor cells. In addition, the activated cells express integrin α5β1 specially, which may help in early discovery of osteoarthritis.
Collapse
|
23
|
Christ A, Herzog K, Willnow TE. LRP2, an auxiliary receptor that controls sonic hedgehog signaling in development and disease. Dev Dyn 2016; 245:569-79. [PMID: 26872844 DOI: 10.1002/dvdy.24394] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 12/31/2022] Open
Abstract
To fulfill their multiple roles in organ development and adult tissue homeostasis, hedgehog (HH) morphogens act through their receptor Patched (PTCH) on target cells. However, HH actions also require HH binding proteins, auxiliary cell surface receptors that agonize or antagonize morphogen signaling in a context-dependent manner. Here, we discuss recent findings on the LDL receptor-related protein 2 (LRP2), an exemplary HH binding protein that modulates sonic hedgehog activities in stem and progenitor cell niches in embryonic and adult tissues. LRP2 functions are crucial for developmental processes in a number of tissues, including the brain, the eye, and the heart, and defects in this receptor pathway are the cause of devastating congenital diseases in humans. Developmental Dynamics 245:569-579, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Katja Herzog
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125, Berlin, Germany
| |
Collapse
|
24
|
Christ A, Christa A, Klippert J, Eule JC, Bachmann S, Wallace VA, Hammes A, Willnow TE. LRP2 Acts as SHH Clearance Receptor to Protect the Retinal Margin from Mitogenic Stimuli. Dev Cell 2015; 35:36-48. [PMID: 26439398 DOI: 10.1016/j.devcel.2015.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 08/21/2015] [Accepted: 09/07/2015] [Indexed: 01/17/2023]
Abstract
During forebrain development, LRP2 promotes morphogen signaling as an auxiliary SHH receptor. However, in the developing retina, LRP2 assumes the opposing function, mediating endocytic clearance of SHH and antagonizing morphogen action. LRP2-mediated clearance prevents spread of SHH activity from the central retina into the retinal margin to protect quiescent progenitor cells in this niche from mitogenic stimuli. Loss of LRP2 in mice increases the sensitivity of the retinal margin for SHH, causing expansion of the retinal progenitor cell pool and hyperproliferation of this tissue. Our findings document the ability of LRP2 to act, in a context-dependent manner, as activator or inhibitor of the SHH pathway. Our current findings uncovered LRP2 activity as the molecular mechanism imposing quiescence of the retinal margin in the mammalian eye and suggest SHH-induced proliferation of the retinal margin as cause of the large eye phenotype observed in mouse models and patients with LRP2 defects.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.
| | - Anna Christa
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Julia Klippert
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - J Corinna Eule
- Small Animal Clinic, Free University Berlin, 14163 Berlin, Germany
| | - Sebastian Bachmann
- Institute for Vegetative Anatomy, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Valerie A Wallace
- Toronto Western Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Annette Hammes
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.
| |
Collapse
|
25
|
Zhu J, Lamba DA. Restoring Vision: Where are We with Stem Cells? CURRENT OPHTHALMOLOGY REPORTS 2015. [DOI: 10.1007/s40135-015-0078-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Todd L, Fischer AJ. Hedgehog signaling stimulates the formation of proliferating Müller glia-derived progenitor cells in the chick retina. Development 2015; 142:2610-22. [PMID: 26116667 DOI: 10.1242/dev.121616] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 06/15/2015] [Indexed: 12/29/2022]
Abstract
Müller glia can be stimulated to de-differentiate and become proliferating progenitor cells that regenerate neurons in the retina. The signaling pathways that regulate the formation of proliferating Müller glia-derived progenitor cells (MGPCs) are beginning to be revealed. The purpose of this study was to investigate whether Hedgehog (Hh) signaling influences the formation of MGPCs in the chick retina. We find that Hh signaling is increased in damaged retinas where MGPCs are known to form. Sonic Hedgehog (Shh) is normally present in the axons of ganglion cells, but becomes associated with Müller glia and MGPCs following retinal damage. Activation of Hh signaling with recombinant human SHH (rhShh) or smoothened agonist (SAG) increased levels of Ptch1, Gli1, Gli2, Gli3, Hes1 and Hes5, and stimulated the formation of proliferating MGPCs in damaged retinas. In undamaged retinas, SAG or rhShh had no apparent effect upon the Müller glia. However, SAG combined with FGF2 potentiated the formation of MGPCs, whereas SAG combined with IGF1 stimulated the nuclear migration of Müller glia, but not the formation of MGPCs. Conversely, inhibition of Hh signaling with KAAD-cyclopamine, Gli antagonists or antibody to Shh reduced numbers of proliferating MGPCs in damaged and FGF2-treated retinas. Hh signaling potentiates Pax6, Klf4 and cFos expression in Müller glia during the formation of MGPCs. We find that FGF2/MAPK signaling recruits Hh signaling into the signaling network that drives the formation of proliferating MGPCs. Our findings implicate Hh signaling as a key component of the network of signaling pathways that promote the de-differentiation of Müller glia and proliferation of MGPCs.
Collapse
Affiliation(s)
- Levi Todd
- Department of Neuroscience, College of Medicine, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave, Columbus, OH 43210, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave, Columbus, OH 43210, USA
| |
Collapse
|
27
|
|
28
|
Jayakody SA, Gonzalez-Cordero A, Ali RR, Pearson RA. Cellular strategies for retinal repair by photoreceptor replacement. Prog Retin Eye Res 2015; 46:31-66. [PMID: 25660226 DOI: 10.1016/j.preteyeres.2015.01.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 01/13/2015] [Accepted: 01/19/2015] [Indexed: 02/08/2023]
Abstract
Loss of photoreceptors due to retinal degeneration is a major cause of blindness in the developed world. While no effective treatment is currently available, cell replacement therapy, using pluripotent stem cell-derived photoreceptor precursor cells, may be a feasible future treatment. Recent reports have demonstrated rescue of visual function following the transplantation of immature photoreceptors and we have seen major advances in our ability to generate transplantation-competent donor cells from stem cell sources. Moreover, we are beginning to realise the possibilities of using endogenous populations of cells from within the retina itself to mediate retinal repair. Here, we present a review of our current understanding of endogenous repair mechanisms together with recent progress in the use of both ocular and pluripotent stem cells for the treatment of photoreceptor loss. We consider how our understanding of retinal development has underpinned many of the recent major advances in translation and moved us closer to the goal of restoring vision by cellular means.
Collapse
Affiliation(s)
- Sujatha A Jayakody
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK
| | - Anai Gonzalez-Cordero
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK
| | - Robin R Ali
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK; NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, City Road, London EC1V 2PD, UK
| | - Rachael A Pearson
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK.
| |
Collapse
|
29
|
Wen W, Pillai-Kastoori L, Wilson SG, Morris AC. Sox4 regulates choroid fissure closure by limiting Hedgehog signaling during ocular morphogenesis. Dev Biol 2014; 399:139-153. [PMID: 25557621 DOI: 10.1016/j.ydbio.2014.12.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/15/2014] [Accepted: 12/22/2014] [Indexed: 01/20/2023]
Abstract
SoxC transcription factors play critical roles in many developmental processes, including neurogenesis, cardiac formation, and skeletal differentiation. In vitro and in vivo loss-of-function studies have suggested that SoxC genes are required for oculogenesis; however the mechanism was poorly understood. Here, we have explored the function of the SoxC factor Sox4 during zebrafish eye development. We show that sox4a and sox4b are expressed in the forebrain and periocular mesenchyme adjacent to the optic stalk during early eye development. Knockdown of sox4 in zebrafish resulted in coloboma, a structural malformation of the eye that is a significant cause of pediatric visual impairment in humans, in which the choroid fissure fails to close. Sox4 morphants displayed altered proximo-distal patterning of the optic vesicle, including expanded pax2 expression in the optic stalk, as well as ectopic cell proliferation in the retina. We show that the abnormal ocular morphogenesis observed in Sox4-deficient zebrafish is caused by elevated Hedgehog (Hh) signaling, and this is due to increased expression of the Hh pathway ligand Indian Hedgehog b (ihhb). Consistent with these results, coloboma in sox4 morphants could be rescued by pharmacological treatment with the Hh inhibitor cyclopamine, or by co-knockdown of ihhb. Conversely, overexpression of sox4 reduced Hh signaling and ihhb expression, resulting in cyclopia. Finally, we demonstrate that sox4 and sox11 have overlapping, but not completely redundant, functions in regulating ocular morphogenesis. Taken together, our data demonstrate that Sox4 is required to limit the extent of Hh signaling during eye development, and suggest that mutations in SoxC factors could contribute to the development of coloboma.
Collapse
Affiliation(s)
- Wen Wen
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| | | | - Stephen G Wilson
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| | - Ann C Morris
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA.
| |
Collapse
|
30
|
Heavner WE, Andoniadou CL, Pevny LH. Establishment of the neurogenic boundary of the mouse retina requires cooperation of SOX2 and WNT signaling. Neural Dev 2014; 9:27. [PMID: 25488119 PMCID: PMC4295269 DOI: 10.1186/1749-8104-9-27] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/14/2014] [Indexed: 12/03/2022] Open
Abstract
Background Eye development in vertebrates relies on the critical regulation of SOX2 expression. Humans with mutations in SOX2 often suffer from eye defects including anophthalmia (no eye) and microphthalmia (small eye). In mice, deletion of Sox2 in optic cup progenitor cells results in loss of neural competence and cell fate conversion of the neural retina to a non-neurogenic fate, specifically the acquisition of fate associated with progenitors of the ciliary epithelium. This fate is also promoted with constitutive expression of stabilized β-Catenin in the optic cup, where the WNT pathway is up-regulated. We addressed whether SOX2 co-ordinates the neurogenic boundary of the retina through modulating the WNT/β-Catenin pathway by using a genetic approach in the mouse. Results Upon deletion of Sox2 in the optic cup, response to WNT signaling was expanded, correlating with loss of neural competence, cell fate conversion of the neural retina to ciliary epithelium primordium and, in addition, increased cell cycle time of optic cup progenitors. Removal of Ctnnb1 rescued the cell fate conversion; however, the loss of neural competence and the proliferation defect resulting from lack of SOX2 were not overcome. Lastly, central Sox2-deficient optic cup progenitor cells exhibited WNT-independent up-regulation of D-type Cyclins. Conclusion We propose two distinct roles for SOX2 in the developing retina. Our findings suggest that SOX2 antagonizes the WNT pathway to maintain a neurogenic fate and, in contrast, regulates cycling of optic cup progenitors in a WNT-independent manner. Given that WNT signaling acting upstream of SOX2 has been implicated in the tumorigenicity of embryonic stem cell-derived retinal progenitor cells, our results distinguish the endogenous role of WNT signaling in early optic cup patterning and support a WNT-independent role for SOX2 in maintaining retinal progenitor cell proliferation. Electronic supplementary material The online version of this article (doi:10.1186/1749-8104-9-27) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Whitney E Heavner
- UNC Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | |
Collapse
|
31
|
Balenci L, Wonders C, Coles BLK, Clarke L, van der Kooy D. Bone morphogenetic proteins and secreted frizzled related protein 2 maintain the quiescence of adult mammalian retinal stem cells. Stem Cells 2014; 31:2218-30. [PMID: 23843349 DOI: 10.1002/stem.1470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 06/04/2013] [Accepted: 06/06/2013] [Indexed: 12/22/2022]
Abstract
Rare retinal stem cells (RSCs) within the ciliary epithelium at the retinal margin of the adult mouse and human eyes can divide in vitro in the absence of growth factors to generate clonal, self-renewing spheres which can generate all the retinal cell types. Since no regenerative properties are seen in situ in the adult mammalian eye, we sought to determine the factors that are involved in the repression of endogenous RSCs. We discovered that factors secreted by the adult lens and cornea block the proliferation of adult RSCs in vitro. Bone morphogenetic protein (BMP)2, BMP4, and secreted frizzled related protein 2 were identified as principal effectors of the anti-proliferative effects on RSCs. As a similar induced quiescence was observed in vitro on both mouse and human RSCs, targeting these molecules in vivo may reactivate RSCs directly in situ in the eyes of the blind.
Collapse
Affiliation(s)
- Laurent Balenci
- Department of Molecular Genetics, Terrence Donnelly Centre for Cellular and Biomolecular Research University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
32
|
Activation of Sonic hedgehog signaling in neural progenitor cells promotes glioma development in the zebrafish optic pathway. Oncogenesis 2014; 3:e96. [PMID: 24686726 PMCID: PMC4038393 DOI: 10.1038/oncsis.2014.10] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 02/11/2014] [Accepted: 02/27/2014] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of Sonic hedgehog (Shh) signaling has been implicated in glioma pathogenesis. Yet, the role of this pathway in gliomagenesis remains controversial because of the lack of relevant animal models. Using the cytokeratin 5 promoter, we ectopically expressed a constitutively active zebrafish Smoothened (Smoa1) in neural progenitor cells and analyzed tumorigenic capacity of activated Shh signaling in both transient and stable transgenic fish. Transient transgenic fish overexpressing Smoa1 developed retinal and brain tumors, suggesting smoa1 is oncogenic in the zebrafish central nervous system (CNS). We further established stable transgenic lines that simultaneously developed optic pathway glioma (OPG) and various retinal tumors. In one of these lines, up to 80% of F1 and F2 fish developed tumors within 1 year of age. Microarray analysis of tumor samples showed upregulated expression of genes involved in the cell cycle, cancer signaling and Shh downstream targets ptc1, gli1 and gli2a. Tumors also exhibited specific gene signatures characteristic of radial glia and progenitor cells as transcriptions of radial glia genes cyp19a1b, s100β, blbp, gfap and the stem/progenitor genes nestin and sox2 were significantly upregulated. Overexpression of GFAP, S100β, BLBP and Sox2 was confirmed by immunofluorescence. We also detected overexpression of Mdm2 throughout the optic pathway in fish with OPG, therefore implicating the Mdm2–Tp53 pathway in glioma pathogenesis. In conclusion, we demonstrate that activated Shh signaling initiates tumorigenesis in the zebrafish CNS and provide the first OPG model not associated with neurofibromatosis 1.
Collapse
|
33
|
Abstract
A unique feature of humans is the complexity of our central nervous system. A fully functional brain requires that billions of neurons make specific contacts in a highly coordinated way, an issue that is still not well understood. The neural retina constitutes an excellent system with which to analyze key aspects of neurogenesis and circuit formation in the central nervous system. Here, we provide an overview of retinal neurogenesis in vertebrates and discuss implications of the developmental mechanisms involved for regenerative therapy approaches.
Collapse
Affiliation(s)
- Lázaro Centanin
- Department of Developmental Biology/Physiology, Centre for Organismal Studies (COS) Heidelberg, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | | |
Collapse
|
34
|
Zhao JJ, Ouyang H, Luo J, Patel S, Xue Y, Quach J, Sfeir N, Zhang M, Fu X, Ding S, Chen S, Zhang K. Induction of retinal progenitors and neurons from mammalian Müller glia under defined conditions. J Biol Chem 2014; 289:11945-11951. [PMID: 24523410 DOI: 10.1074/jbc.m113.532671] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Vision impairment caused by loss of retinal neurons affects millions of people worldwide, and currently, there is no effective treatment. Müller glia of mammalian retina may represent an under-recognized and potential source for regeneration of a wide range of retinal cell types, including retinal ganglion cells and photoreceptors. Here, we demonstrated that mouse Müller glia cells have the capacity to be reprogrammed into the retinal neuronal cell fate and are competent to give rise to photoreceptors under a defined culture condition. Inactivation of p53 released proliferation restriction of Müller glia and significantly enhanced the induction of retinal progenitor from Müller glia in culture. Moreover, following the ocular transplantation, the Müller glia-derived progenitors were differentiated toward the fates of photoreceptors and retinal ganglion cells. Together, these results demonstrate the feasibility of using Müller glia as a potential source for retinal repair and regeneration.
Collapse
Affiliation(s)
- Jack Jiagang Zhao
- Department of Ophthalmology and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093.
| | - Hong Ouyang
- Department of Ophthalmology and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093
| | - Jing Luo
- Department of Ophthalmology and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093; Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 41001, China
| | - Sherrina Patel
- Department of Ophthalmology and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093
| | - Yuanchao Xue
- Department of Cellular and Molecular Medicine and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093
| | - John Quach
- Department of Ophthalmology and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093
| | - Nicole Sfeir
- Department of Ophthalmology and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093
| | - Meixia Zhang
- Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China
| | - Xiangdong Fu
- Department of Cellular and Molecular Medicine and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093
| | - Sheng Ding
- Gladstone Institutes, University of California, San Francisco, San Francisco, California 94158-2261
| | - Shaochen Chen
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093
| | - Kang Zhang
- Department of Ophthalmology and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093; Veterans Administration Healthcare System, San Diego, California 92161.
| |
Collapse
|
35
|
Lenkowski JR, Raymond PA. Müller glia: Stem cells for generation and regeneration of retinal neurons in teleost fish. Prog Retin Eye Res 2014; 40:94-123. [PMID: 24412518 DOI: 10.1016/j.preteyeres.2013.12.007] [Citation(s) in RCA: 223] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/28/2013] [Accepted: 12/30/2013] [Indexed: 12/31/2022]
Abstract
Adult zebrafish generate new neurons in the brain and retina throughout life. Growth-related neurogenesis allows a vigorous regenerative response to damage, and fish can regenerate retinal neurons, including photoreceptors, and restore functional vision following photic, chemical, or mechanical destruction of the retina. Müller glial cells in fish function as radial-glial-like neural stem cells. During adult growth, Müller glial nuclei undergo sporadic, asymmetric, self-renewing mitotic divisions in the inner nuclear layer to generate a rod progenitor that migrates along the radial fiber of the Müller glia into the outer nuclear layer, proliferates, and differentiates exclusively into rod photoreceptors. When retinal neurons are destroyed, Müller glia in the immediate vicinity of the damage partially and transiently dedifferentiate, re-express retinal progenitor and stem cell markers, re-enter the cell cycle, undergo interkinetic nuclear migration (characteristic of neuroepithelial cells), and divide once in an asymmetric, self-renewing division to generate a retinal progenitor. This daughter cell proliferates rapidly to form a compact neurogenic cluster surrounding the Müller glia; these multipotent retinal progenitors then migrate along the radial fiber to the appropriate lamina to replace missing retinal neurons. Some aspects of the injury-response in fish Müller glia resemble gliosis as observed in mammals, and mammalian Müller glia exhibit some neurogenic properties, indicative of a latent ability to regenerate retinal neurons. Understanding the specific properties of fish Müller glia that facilitate their robust capacity to generate retinal neurons will inform and inspire new clinical approaches for treating blindness and visual loss with regenerative medicine.
Collapse
Affiliation(s)
- Jenny R Lenkowski
- Department of Molecular, Cellular, and Developmental Biology, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA.
| | - Pamela A Raymond
- Department of Molecular, Cellular, and Developmental Biology, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
36
|
Bai L, Kiyama T, Li H, Wang SW. Birth of cone bipolar cells, but not rod bipolar cells, is associated with existing RGCs. PLoS One 2014; 9:e83686. [PMID: 24392091 PMCID: PMC3879276 DOI: 10.1371/journal.pone.0083686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/14/2013] [Indexed: 12/12/2022] Open
Abstract
Retinal ganglion cells (RGCs) play important roles in retinogenesis. They are required for normal retinal histogenesis and retinal cell number balance. Developmental RGC loss is typically characterized by initial retinal neuronal number imbalance and subsequent loss of retinal neurons. However, it is not clear whether loss of a specific non-RGC cell type in the RGC-depleted retina is due to reduced cell production or subsequent degeneration. Taking advantage of three knockout mice with varying degrees of RGC depletion, we re-examined bipolar cell production in these retinas from various aspects. Results show that generation of the cone bipolar cells is correlated with the existing number of RGCs. However, generation of the rod bipolar cells is unaffected by RGC shortage. Results report the first observation that RGCs selectively influence the genesis of subsequent retinal cell types.
Collapse
Affiliation(s)
- Ling Bai
- Department of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, China
| | - Takae Kiyama
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston Medical School, Houston, Texas, United States of America
| | - Hongyan Li
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston Medical School, Houston, Texas, United States of America
| | - Steven W. Wang
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston Medical School, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
37
|
A role for the ciliary marginal zone in the melanopsin-dependent intrinsic pupillary light reflex. Exp Eye Res 2013; 119:8-18. [PMID: 24316157 DOI: 10.1016/j.exer.2013.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/18/2013] [Accepted: 11/20/2013] [Indexed: 11/20/2022]
Abstract
Maintenance of pupillary constriction in light-adapted rodents has traditionally been thought to involve a reflex between retina, brain and iris, with recent work identifying the melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) as the major conduits for retinal input to the brain. There is also a less well-understood phenomenon whereby the iris of some mammals, including mice, will constrict to light when either the eye, or the iris itself is physically isolated from the brain. The intrinsic pupillary light reflex (iPLR) is the term given to pupil constriction in the absence of retinal input to the brain. Here, using an intraocular axotomy approach, we show that the iPLR in conscious mice spans a dynamic range over 3 log units of irradiance. This iPLR response is absent in melanopsin knockout (MKO) mice and can be significantly inhibited by atropine. Immunohistochemistry for cfos and melanopsin, in combination with light exposure revealed a population of small ipRGCs in the retinal ciliary marginal zone (CMZ), which remain responsive to light in axotomised mice. We report that damage to the CMZ in a novel in vitro preparation removes a significant component of the iPLR response, while a detailed immunohistochemical analysis of the CMZ in wildtype mice revealed a melanopsin-rich plexus, which was consistently most intense in nasal retina. There were clear examples of melanopsin-positive, direct retino-ciliary projections, which appear to emanate from Brn3b negative, M1 type ipRGCs. These cells are clustered along the melanopsin-rich plexus nasally and may channel ipRGC signals from retina into the iris via ciliary body. Comparison between wildtype and MKO mice reveals that the ciliary body is also weakly stained for melanopsin. Our results show that the full extent of iPLR in mice requires cholinergic neurotransmission and intact signalling at the CMZ/ciliary body. This response may be mediated to some extent by ipRGCs, which send direct projections from the retina into ciliary body. In addition to the melanopsin-mediated iris sphincter constriction suggested by others, we propose a new mechanism, which may involve constriction of the ciliary body and ipRGC-mediated relaxation of the iris dilator muscle.
Collapse
|
38
|
Balenci L, van der Kooy D. Notch signaling induces retinal stem-like properties in perinatal neural retina progenitors and promotes symmetric divisions in adult retinal stem cells. Stem Cells Dev 2013; 23:230-44. [PMID: 24050115 DOI: 10.1089/scd.2013.0177] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Understanding the mechanisms regulating retinal stem cell (RSC) activity is fundamental for future stem cell-based therapeutic purposes. By combining gain and loss of function approaches, we addressed whether Notch signaling may play a selective role in retinal stem versus retinal progenitor cells in both developing and adult eyes. Inhibition of either Notch or fibroblast growth factor signaling reduced proliferation of retinal stem and retinal progenitor cells, and inhibited RSC self-renewal. Conversely, exogenous Delta-like 3 and direct intrinsic Notch activation stimulated expansionary symmetric divisions in adult RSCs with the concomitant upregulation of Hes5. Knocking down Hes5 expression specifically decreased the numbers, but not the diameters, of adult RSC primary spheres, indicating that HES5 is the downstream effector of Notch receptor in controlling adult RSC proliferation. In addition, constitutive Notch activation induced retinal stem-like asymmetric self-renewal properties, with no expansion (no symmetrical division) in perinatal neural retina progenitor cells. These findings highlight central roles of Notch signaling activity in regulating the modes of division of retinal stem and retinal progenitor cells.
Collapse
Affiliation(s)
- Laurent Balenci
- Department of Molecular Genetics, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto , Toronto, Canada
| | | |
Collapse
|
39
|
Fang Y, Cho KS, Tchedre K, Lee SW, Guo C, Kinouchi H, Fried S, Sun X, Chen DF. Ephrin-A3 suppresses Wnt signaling to control retinal stem cell potency. Stem Cells 2013; 31:349-59. [PMID: 23165658 DOI: 10.1002/stem.1283] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 10/23/2012] [Indexed: 11/09/2022]
Abstract
The ciliary epithelium (CE) of adult mammals has been reported to provide a source of retinal stem cells (RSCs) that can give rise to all retinal cell types in vitro. A recent study, however, suggests that CE-derived cells possess properties of pigmented ciliary epithelial cells and display little neurogenic potential. Here we show that the neurogenic potential of CE-derived cells is negatively regulated by ephrin-A3, which is upregulated in the CE of postnatal mice and presents a strong prohibitory niche for adult RSCs. Addition of ephrin-A3 inhibits proliferation of CE-derived RSCs and increases pigment 349 cell 359. In contrast, absence of ephrin-A3 promotes proliferation and increases expression of neural progenitor cell markers and photoreceptor progeny. The negative effects of ephrin-A3 on CE-derived RSCs are mediated through activation of an EphA4 receptor and suppression of Wnt3a/β-catenin signaling. Together, our data suggest that CE-derived RSCs contain the intrinsic machinery to generate photoreceptors and other retinal neurons, while the CE of adult mice expresses negative regulators that prohibit the proliferation and neural differentiation of RSCs. Manipulating ephrin and Wnt/β-catenin signaling may, thus, represent a viable approach in activating the endogenous neurogenic potential of CE-derived RSCs for treating photoreceptor damage and retinal degenerative disorders.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
McNeill B, Mazerolle C, Bassett EA, Mears AJ, Ringuette R, Lagali P, Picketts DJ, Paes K, Rice D, Wallace VA. Hedgehog regulates Norrie disease protein to drive neural progenitor self-renewal. Hum Mol Genet 2012. [PMID: 23201751 DOI: 10.1093/hmg/dds505] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Norrie disease (ND) is a congenital disorder characterized by retinal hypovascularization and cognitive delay. ND has been linked to mutations in 'Norrie Disease Protein' (Ndp), which encodes the secreted protein Norrin. Norrin functions as a secreted angiogenic factor, although its role in neural development has not been assessed. Here, we show that Ndp expression is initiated in retinal progenitors in response to Hedgehog (Hh) signaling, which induces Gli2 binding to the Ndp promoter. Using a combination of genetic epistasis and acute RNAi-knockdown approaches, we show that Ndp is required downstream of Hh activation to induce retinal progenitor proliferation in the retina. Strikingly, Ndp regulates the rate of cell-cycle re-entry and not cell-cycle kinetics, thereby uncoupling the self-renewal and cell-cycle progression functions of Hh. Taken together, we have uncovered a cell autonomous function for Ndp in retinal progenitor proliferation that is independent of its function in the retinal vasculature, which could explain the neural defects associated with ND.
Collapse
Affiliation(s)
- Brian McNeill
- Vision Program, Ottawa Hospital Research Institute, Ottawa, Ont. K1H 8L6, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Distinct neurogenic potential in the retinal margin and the pars plana of mammalian eye. J Neurosci 2012; 32:12797-807. [PMID: 22973003 DOI: 10.1523/jneurosci.0118-12.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Unlike many other vertebrates, a healthy mammalian retina does not grow throughout life and lacks a ciliary margin zone capable of actively generating new neurons. The isolation of stem-like cells from the ciliary epithelium has led to speculation that the mammalian retina and/or surrounding tissues may retain neurogenic potential capable of responding to retinal damage. Using genetically altered mouse lines with varying degrees of retinal ganglion cell loss, we show that the retinal margin responds to ganglion cell loss by prolonging specific neurogenic activity, as characterized by increased numbers of Atoh7(LacZ)-expressing cells. The extent of neurogenic activity correlated with the degree of ganglion cell deficiency. In the pars plana, but not the retinal margin, cells remain proliferative into adulthood, marking the junction of pars plana and retinal margin as a niche capable of producing proliferative cells in the mammalian retina and a potential cellular source for retinal regeneration.
Collapse
|
42
|
Cerveny KL, Varga M, Wilson SW. Continued growth and circuit building in the anamniote visual system. Dev Neurobiol 2012; 72:328-45. [PMID: 21563317 DOI: 10.1002/dneu.20917] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Fish and amphibia are capable of lifelong growth and regeneration. The two core components of their visual system, the retina and tectum both maintain small populations of stem cells that contribute new neurons and glia to these tissues as they grow. As the animals age, the initial retinal projections onto the tectum are continuously remodeled to maintain retinotopy. These properties raise several biological challenges related to the control of proliferation and differentiation of retinal and tectal stem cells. For instance, how do stem and progenitor cells integrate intrinsic and extrinsic cues to produce the appropriate type and number of cells needed by the growing tissue. Does retinal growth or neuronal activity influence tectal growth? What are the cellular and molecular mechanisms that enable retinal axons to shift their tectal connections as these two tissues grow in incongruent patterns? While we cannot yet provide answers to these questions, this review attempts to supply background and context, laying the ground work for new investigations.
Collapse
Affiliation(s)
- Kara L Cerveny
- Department of Cell and Developmental Biology, University College, London, UK
| | | | | |
Collapse
|
43
|
The emerging role of stem cells in ocular neurodegeneration: hype or hope? Mol Cell Biochem 2012; 365:65-76. [PMID: 22290231 DOI: 10.1007/s11010-012-1244-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 01/14/2012] [Indexed: 01/14/2023]
Abstract
Affecting over a hundred million individuals worldwide, retinal diseases are among the leading causes of irreversible visual impairment and blindness. Thus, an appropriate study models, especially animal models, are essential to furthering our understanding of the etiology, pathology, and progression of these diseases. In this review, we provide an overview of retinal disorders in the context of biotherapeutic approaches in these disorders.
Collapse
|
44
|
Abstract
Distinct stem cell types have been established from embryos and identified in the fetal tissues and umbilical cord blood as well as in specific niches in many adult mammalian tissues and organs such as bone marrow, brain, skin, eyes, heart, kidneys, lungs, gastrointestinal tract, pancreas, liver, breast, ovaries, and prostate. All stem cells are undifferentiated cells that exhibit unlimited self-renewal and can generate multiple cell lineages or more restricted progenitor populations that can contribute to tissue homeostasis by replenishing the cells or to tissue regeneration after injury. The remarkable progress of regenerative medicine in the last few years indicates promise for the use of stem cells in the treatment of ophthalmic disorders. Experimental and human studies with intravitreal bone marrow-derived stem cells have begun. This paper reviews recent advances and potential sources of stem cells for cell therapy in retinal diseases.
Collapse
|
45
|
Jakobiec FA, Zakka FR, D'Amato R, Deangelis MM, Walton DS, Rao RC. Unilateral sporadic retinal dysplasia: results of histopathologic, immunohistochemical, chromosomal, genetic, and VEGF-A analyses. J AAPOS 2011; 15:579-86. [PMID: 22153404 PMCID: PMC4242509 DOI: 10.1016/j.jaapos.2011.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 07/28/2011] [Accepted: 08/19/2011] [Indexed: 11/16/2022]
Abstract
PURPOSE To describe new findings in a case of unilateral retinal dysplasia. METHODS Histopathologic evaluation of an enucleated globe and analysis with immunohistochemical probes, karyotyping, and genetic analysis for the Norrie gene, and aqueous assay for vascular endothelial growth factor A (VEGF-A). RESULTS Histopathological examination of the globe revealed retinal dysplasia with pseudorosette formation, abnormal or absent retinal nuclear lamination, a paucity of disorganized retinal microvasculature, retinal infoldings, advanced gliosis, persistent hyperplastic vitreous, exuberant neovascularization of the vitreous, and iris neovascularization (identical to the findings observed in bilateral Norrie disease). Immunohistochemistry disclosed GFAP-positive and GLUT-1-positive gliosis and retinal and persistent hyperplastic vitreous microvessels that were CD34-positive and GLUT-1-negative. Ki-67-positive retinal cells were polarized toward the subretinal space and absent in the retinal invaginations and pseudorosettes. A normal karyotype was found, and DNA sequencing revealed no known mutation in the region of the Norrie gene (NDP) in sputum or retinal DNA. Aqueous obtained immediately after enucleation contained an exceptionally high concentration of VEGF-A (4.5 ng/mL). CONCLUSIONS Despite the failure to find an abnormal NDP allele, other unexplored NDP regions, an undetected defect restricted to retinal tissues, or an autosomal mutation coupled with disrupted signaling pathways may be responsible for the condition. High aqueous VEGF-A suggests that this cytokine may play a role in pathogenesis in conjunction with other pathways.
Collapse
Affiliation(s)
- Frederick A Jakobiec
- David G. Cogan Laboratory of Ophthalmic Pathology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Bibliowicz J, Gross JM. Ectopic proliferation contributes to retinal dysplasia in the juvenile zebrafish patched2 mutant eye. Invest Ophthalmol Vis Sci 2011; 52:8868-77. [PMID: 22003118 DOI: 10.1167/iovs.11-8033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Patched is a well-studied tumor suppressor and negative regulator of the Hedgehog (Hh) pathway. Earlier work in this laboratory has shown that embryonic zebrafish patched2 (ptc2) mutant retinas possess an expanded ciliary marginal zone (CMZ) and phenotypes similar to those in human patients with basal cell naevus syndrome (BCNS), a congenital disorder linked to mutations in the human PTCH gene. This study extends the analysis of retinal structure and homeostasis in ptc2-/- mutants to juvenile stages, to determine whether Patched 2 function is essential in the postembryonic eye. METHODS Histologic, immunohistochemical, and molecular analyses were used to characterize retinal defects in the 6-week-old juvenile ptc2-/- retina. RESULTS Juvenile ptc2-/- mutants exhibited peripheral retinal dysplasias that included the presence of ectopic neuronal clusters in the inner nuclear layer (INL) and regions of disrupted retinal lamination. Retinal dysplasias were locally associated with ectopic proliferation. BrdU/EdU labeling and immunohistochemistry assays demonstrated that a population of ectopically proliferating cells gave rise to the ectopic neuronal clusters in the INL of ptc2-/- mutants and that this contributed to retinal dysplasia in the mutant eye. CONCLUSIONS These results demonstrate a direct link between overproliferation and retinal dysplasia in the ptc2-/- juvenile retina and establish ectopic proliferation as the likely cellular underpinning of retinal dysplasia in juvenile ptc2-/- mutants.
Collapse
Affiliation(s)
- Jonathan Bibliowicz
- Section of Molecular Cell and Developmental Biology, The University of Texas at Austin, Austin, Texas, USA
| | | |
Collapse
|
47
|
Abstract
Retinal degenerative disease has limited therapeutic options and the possibility of stem cell-mediated regenerative treatments is being actively explored for these blinding retinal conditions. The relative accessibility of this central nervous system tissue and the ability to visually monitor changes after transplantation make the retina and adjacent retinal pigment epithelium prime targets for pioneering stem cell therapeutics. Prior work conducted for several decades indicated the promise of cell transplantation for retinal disease, and new strategies that combine these established surgical approaches with stem cell-derived donor cells is ongoing. A variety of tissue-specific and pluripotent-derived donor cells are being advanced to replace lost or damaged retinal cells and/or to slow the disease processes by providing neuroprotective factors, with the ultimate aim of long-term improvement in visual function. Clinical trials are in the early stages, and data on safety and efficacy are widely anticipated. Positive outcomes from these stem cell-based clinical studies would radically change the way that blinding disorders are approached in the clinic.
Collapse
|
48
|
Proliferation of the ciliary epithelium with retinal neuronal and photoreceptor cell differentiation in human eyes with retinal detachment and proliferative vitreoretinopathy. Graefes Arch Clin Exp Ophthalmol 2011; 250:409-23. [DOI: 10.1007/s00417-011-1797-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 07/22/2011] [Accepted: 08/01/2011] [Indexed: 01/07/2023] Open
|
49
|
Ouchi Y, Baba Y, Koso H, Taketo MM, Iwamoto T, Aburatani H, Watanabe S. β-Catenin signaling regulates the timing of cell differentiation in mouse retinal progenitor cells. Mol Cell Neurosci 2011; 46:770-80. [DOI: 10.1016/j.mcn.2011.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 02/17/2011] [Accepted: 02/18/2011] [Indexed: 12/27/2022] Open
|
50
|
Fischer AJ, Bongini R. Turning Müller glia into neural progenitors in the retina. Mol Neurobiol 2010; 42:199-209. [PMID: 21088932 DOI: 10.1007/s12035-010-8152-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 11/10/2010] [Indexed: 12/22/2022]
Abstract
Stimulating neuronal regeneration is a potential strategy to treat sight-threatening diseases of the retina. In some classes of vertebrates, retinal regeneration occurs spontaneously to effectively replace neurons lost to acute damage in order to restore visual function. There are different mechanisms and cellular sources of retinal regeneration in different species, include the retinal pigmented epithelium, progenitors seeded across the retina, and the Müller glia. This review briefly summarizes the different mechanisms of retinal regeneration in frogs, fish, chicks, and rodents. The bulk of this review summarizes and discusses recent findings regarding regeneration from Müller glia-derived progenitors, with emphasis on findings in the chick retina. The Müller glia are a promising source of regeneration-supporting cells that are intrinsic to the retina and significant evidence indicated these glias can be stimulated to produce neurons in different classes of vertebrates. The key to harnessing the neurogenic potential of Müller glia is to identify the secreted factors, signaling pathways, and transcription factors that enable de-differentiation, proliferation, and neurogenesis. We review findings regarding the roles of mitogen-activated protein kinase and notch signaling in the proliferation and generation of Müller glia-derived retinal progenitors.
Collapse
Affiliation(s)
- Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, 3020 Graves Hall, 333 West 10th Ave, Columbus, OH 43210-1239, USA.
| | | |
Collapse
|