1
|
Perez-Estrada JR, Tangeman JA, Proto-Newton M, Sanaka H, Smucker B, Del Rio-Tsonis K. Metabolic states influence chicken retinal pigment epithelium cell fate decisions. Development 2024; 151:dev202462. [PMID: 39120084 PMCID: PMC11708821 DOI: 10.1242/dev.202462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
During tissue regeneration, proliferation, dedifferentiation and reprogramming are necessary to restore lost structures. However, it is not fully understood how metabolism intersects with these processes. Chicken embryos can regenerate their retina through retinal pigment epithelium (RPE) reprogramming when treated with fibroblast factor 2 (FGF2). Using transcriptome profiling, we uncovered extensive regulation of gene sets pertaining to proliferation, neurogenesis and glycolysis throughout RPE-to-neural retina reprogramming. By manipulating cell media composition, we determined that glucose, glutamine or pyruvate are individually sufficient to support RPE reprogramming, identifying glycolysis as a requisite. Conversely, the activation of pyruvate dehydrogenase by inhibition of pyruvate dehydrogenase kinases, induces epithelial-to-mesenchymal transition, while simultaneously blocking the activation of neural retina fate. We also identified that epithelial-to-mesenchymal transition fate is partially driven by an oxidative environment. Our findings provide evidence that metabolism controls RPE cell fate decisions and provide insights into the metabolic state of RPE cells, which are prone to fate changes in regeneration and pathologies, such as proliferative vitreoretinopathy.
Collapse
Affiliation(s)
- J. Raúl Perez-Estrada
- Department of Biology, Miami University, Oxford, OH 45056, USA
- Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Jared A. Tangeman
- Department of Biology, Miami University, Oxford, OH 45056, USA
- Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | | | | | - Byran Smucker
- Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
- Department of Statistics, Miami University, Oxford, OH 45056, USA
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH 45056, USA
- Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| |
Collapse
|
2
|
Bose D, Ortolan D, Farnoodian M, Sharma R, Bharti K. Considerations for Developing an Autologous Induced Pluripotent Stem Cell (iPSC)-Derived Retinal Pigment Epithelium (RPE) Replacement Therapy. Cold Spring Harb Perspect Med 2024; 14:a041295. [PMID: 37487631 PMCID: PMC10910357 DOI: 10.1101/cshperspect.a041295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Cell-replacement therapies are a new class of treatments, which include induced pluripotent stem cell (iPSC)-derived tissues that aim to replace degenerated cells. iPSCs can potentially be used to generate any cell type of the body, making them a powerful tool for treating degenerative diseases. Cell replacement for retinal degenerative diseases is at the forefront of cell therapies, given the accessibility of the eye for surgical procedures and a huge unmet medical need for retinal degenerative diseases with no current treatment options. Clinical trials are ongoing in different parts of the world using stem cell-derived retinal pigment epithelium (RPE). This review focuses on scientific and regulatory considerations when developing an iPSC-derived RPE cell therapy from the development of a robust and efficient differentiation protocol to critical quality control assays for cell validation, the choice of an appropriate animal model for preclinical testing, and the regulatory aspects that dictate the final approval for proceeding to a first-in-human clinical trial.
Collapse
Affiliation(s)
- Devika Bose
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Davide Ortolan
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Mitra Farnoodian
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
3
|
Perez-Estrada JR, Tangeman JA, Proto-Newton M, Sanaka H, Smucker B, Del Rio-Tsonis K. DISTINCT METABOLIC STATES DIRECT RETINAL PIGMENT EPITHELIUM CELL FATE DECISIONS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559631. [PMID: 37808829 PMCID: PMC10557760 DOI: 10.1101/2023.09.26.559631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
During tissue regeneration, proliferation, dedifferentiation, and reprogramming are necessary to restore lost structures. However, it is not fully understood how metabolism intersects with these processes. Chicken embryos can regenerate their retina through retinal pigment epithelium (RPE) reprogramming when treated with fibroblast factor 2 (FGF2). Using transcriptome profiling, we uncovered extensive regulation of gene sets pertaining to proliferation, neurogenesis, and glycolysis throughout RPE-to-neural retina reprogramming. By manipulating cell media composition, we determined that glucose, glutamine, or pyruvate are sufficient to support RPE reprogramming identifying glycolysis as a requisite. Conversely, the induction of oxidative metabolism by activation of pyruvate dehydrogenase induces Epithelial-to-mesenchymal transition (EMT), while simultaneously blocking the activation of neural retina fate. We also identify that EMT is partially driven by an oxidative environment. Our findings provide evidence that metabolism controls RPE cell fate decisions and provide insights into the metabolic state of RPE cells, which are prone to fate changes in regeneration and pathologies, such as proliferative vitreoretinopathy.
Collapse
|
4
|
Casey MA, Lusk S, Kwan KM. Eye Morphogenesis in Vertebrates. Annu Rev Vis Sci 2023; 9:221-243. [PMID: 37040791 DOI: 10.1146/annurev-vision-100720-111125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Proper eye structure is essential for visual function: Multiple essential eye tissues must take shape and assemble into a precise three-dimensional configuration. Accordingly, alterations to eye structure can lead to pathological conditions of visual impairment. Changes in eye shape can also be adaptive over evolutionary time. Eye structure is first established during development with the formation of the optic cup, which contains the neural retina, retinal pigment epithelium, and lens. This crucial yet deceptively simple hemispherical structure lays the foundation for all later elaborations of the eye. Building on descriptions of the embryonic eye that started with hand drawings and micrographs, the field is beginning to identify mechanisms driving dynamic changes in three-dimensional cell and tissue shape. A combination of molecular genetics, imaging, and pharmacological approaches is defining connections among transcription factors, signaling pathways, and the intracellular machinery governing the emergence of this crucial structure.
Collapse
Affiliation(s)
- Macaulie A Casey
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA; , ,
| | - Sarah Lusk
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA; , ,
| | - Kristen M Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA; , ,
| |
Collapse
|
5
|
Emri E, Cappa O, Kelly C, Kortvely E, SanGiovanni JP, McKay BS, Bergen AA, Simpson DA, Lengyel I. Zinc Supplementation Induced Transcriptional Changes in Primary Human Retinal Pigment Epithelium: A Single-Cell RNA Sequencing Study to Understand Age-Related Macular Degeneration. Cells 2023; 12:773. [PMID: 36899910 PMCID: PMC10000409 DOI: 10.3390/cells12050773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Zinc supplementation has been shown to be beneficial to slow the progression of age-related macular degeneration (AMD). However, the molecular mechanism underpinning this benefit is not well understood. This study used single-cell RNA sequencing to identify transcriptomic changes induced by zinc supplementation. Human primary retinal pigment epithelial (RPE) cells could mature for up to 19 weeks. After 1 or 18 weeks in culture, we supplemented the culture medium with 125 µM added zinc for one week. RPE cells developed high transepithelial electrical resistance, extensive, but variable pigmentation, and deposited sub-RPE material similar to the hallmark lesions of AMD. Unsupervised cluster analysis of the combined transcriptome of the cells isolated after 2, 9, and 19 weeks in culture showed considerable heterogeneity. Clustering based on 234 pre-selected RPE-specific genes divided the cells into two distinct clusters, we defined as more and less differentiated cells. The proportion of more differentiated cells increased with time in culture, but appreciable numbers of cells remained less differentiated even at 19 weeks. Pseudotemporal ordering identified 537 genes that could be implicated in the dynamics of RPE cell differentiation (FDR < 0.05). Zinc treatment resulted in the differential expression of 281 of these genes (FDR < 0.05). These genes were associated with several biological pathways with modulation of ID1/ID3 transcriptional regulation. Overall, zinc had a multitude of effects on the RPE transcriptome, including several genes involved in pigmentation, complement regulation, mineralization, and cholesterol metabolism processes associated with AMD.
Collapse
Affiliation(s)
- Eszter Emri
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
- Section Ophthalmogenetics, Department of Human Genetics, Queen Emma Centre for Precision Medicine, Amsterdam UMC, Location AMC, 1105AZ Amsterdam, The Netherlands
| | - Oisin Cappa
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Caoimhe Kelly
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Elod Kortvely
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - John Paul SanGiovanni
- Biosciences Research Laboratories, BIO5 Institute, University of Arizona, 1230 North Cherry Avenue, Tucson, AZ 85724, USA
| | - Brian S. McKay
- Department of Ophthalmology and Vision Science, University of Arizona, 1656 E. Mabel Street, Tucson, AZ 85724, USA
| | - Arthur A. Bergen
- Section Ophthalmogenetics, Department of Human Genetics, Queen Emma Centre for Precision Medicine, Amsterdam UMC, Location AMC, 1105AZ Amsterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN-KNAW), 1105AZ Amsterdam, The Netherlands
| | - David A. Simpson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| |
Collapse
|
6
|
Lee JH, Park HS, Holmes DP. Elastic Instabilities Govern the Morphogenesis of the Optic Cup. PHYSICAL REVIEW LETTERS 2021; 127:138102. [PMID: 34623834 DOI: 10.1103/physrevlett.127.138102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Because the normal operation of the eye depends on sensitive morphogenetic processes for its eventual shape, developmental flaws can lead to wide-ranging ocular defects. However, the physical processes and mechanisms governing ocular morphogenesis are not well understood. Here, using analytical theory and nonlinear shell finite-element simulations, we show, for optic vesicles experiencing matrix-constrained growth, that elastic instabilities govern the optic cup morphogenesis. By capturing the stress amplification owing to mass increase during growth, we show that the morphogenesis is driven by two elastic instabilities analogous to the snap through in spherical shells, where the second instability is sensitive to the optic cup geometry. In particular, if the optic vesicle is too slender, it will buckle and break axisymmetry, thus, preventing normal development. Our results shed light on the morphogenetic mechanisms governing the formation of a functional biological system and the role of elastic instabilities in the shape selection of soft biological structures.
Collapse
Affiliation(s)
- Jeong-Ho Lee
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts 02215, USA
| | - Harold S Park
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts 02215, USA
| | - Douglas P Holmes
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts 02215, USA
| |
Collapse
|
7
|
Divya D, Bhattacharya TK. Bone morphogenetic proteins (BMPs) and their role in poultry. WORLD POULTRY SCI J 2021. [DOI: 10.1080/00439339.2021.1959274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- D. Divya
- Molecular Genetics and Breeding Division, ICAR-Directorate of Poultry Research, Hyderabad, India
| | - T. K. Bhattacharya
- Molecular Genetics and Breeding Division, ICAR-Directorate of Poultry Research, Hyderabad, India
| |
Collapse
|
8
|
Tangeman JA, Luz-Madrigal A, Sreeskandarajan S, Grajales-Esquivel E, Liu L, Liang C, Tsonis PA, Del Rio-Tsonis K. Transcriptome Profiling of Embryonic Retinal Pigment Epithelium Reprogramming. Genes (Basel) 2021; 12:genes12060840. [PMID: 34072522 PMCID: PMC8226911 DOI: 10.3390/genes12060840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/05/2021] [Accepted: 05/22/2021] [Indexed: 12/27/2022] Open
Abstract
The plasticity of human retinal pigment epithelium (RPE) has been observed during proliferative vitreoretinopathy, a defective repair process during which injured RPE gives rise to fibrosis. In contrast, following injury, the RPE of the embryonic chicken can be reprogrammed to regenerate neural retina in a fibroblast growth factor 2 (FGF2)-dependent manner. To better explore the mechanisms underlying embryonic RPE reprogramming, we used laser capture microdissection to isolate RNA from (1) intact RPE, (2) transiently reprogrammed RPE (t-rRPE) 6 h post-retinectomy, and (3) reprogrammed RPE (rRPE) 6 h post-retinectomy with FGF2 treatment. Using RNA-seq, we observed the acute repression of genes related to cell cycle progression in the injured t-rRPE, as well as up-regulation of genes associated with injury. In contrast, the rRPE was strongly enriched for mitogen-activated protein kinase (MAPK)-responsive genes and retina development factors, confirming that FGF2 and the downstream MAPK cascade are the main drivers of embryonic RPE reprogramming. Clustering and pathway enrichment analysis was used to create an integrated network of the core processes associated with RPE reprogramming, including key terms pertaining to injury response, migration, actin dynamics, and cell cycle progression. Finally, we employed gene set enrichment analysis to suggest a previously uncovered role for epithelial-mesenchymal transition (EMT) machinery in the initiation of embryonic chick RPE reprogramming. The EMT program is accompanied by extensive, coordinated regulation of extracellular matrix (ECM) associated factors, and these observations together suggest an early role for ECM and EMT-like dynamics during reprogramming. Our study provides for the first time an in-depth transcriptomic analysis of embryonic RPE reprogramming and will prove useful in guiding future efforts to understand proliferative disorders of the RPE and to promote retinal regeneration.
Collapse
Affiliation(s)
- Jared A. Tangeman
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
| | - Agustín Luz-Madrigal
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sutharzan Sreeskandarajan
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Erika Grajales-Esquivel
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
| | - Lin Liu
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
| | - Chun Liang
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- Department of Computer Science and Software Engineering, Miami University, Oxford, OH 45056, USA
| | - Panagiotis A. Tsonis
- Department of Biology, University of Dayton and Center for Tissue Regeneration and Engineering at the University of Dayton (TREND), Dayton, OH 45469, USA;
| | - Katia Del Rio-Tsonis
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH 45056, USA; (J.A.T.); (A.L.-M.); (S.S.); (E.G.-E.); (L.L.); (C.L.)
- Correspondence: ; Tel.: +513-529-3128; Fax: +513-529-6900
| |
Collapse
|
9
|
George SM, Lu F, Rao M, Leach LL, Gross JM. The retinal pigment epithelium: Development, injury responses, and regenerative potential in mammalian and non-mammalian systems. Prog Retin Eye Res 2021; 85:100969. [PMID: 33901682 DOI: 10.1016/j.preteyeres.2021.100969] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022]
Abstract
Diseases that result in retinal pigment epithelium (RPE) degeneration, such as age-related macular degeneration (AMD), are among the leading causes of blindness worldwide. Atrophic (dry) AMD is the most prevalent form of AMD and there are currently no effective therapies to prevent RPE cell death or restore RPE cells lost from AMD. An intriguing approach to treat AMD and other RPE degenerative diseases is to develop therapies focused on stimulating endogenous RPE regeneration. For this to become feasible, a deeper understanding of the mechanisms underlying RPE development, injury responses and regenerative potential is needed. In mammals, RPE regeneration is extremely limited; small lesions can be repaired by the expansion of adjacent RPE cells, but large lesions cannot be repaired as remaining RPE cells are unable to functionally replace lost RPE tissue. In some injury paradigms, RPE cells proliferate but do not regenerate a morphologically normal monolayer, while in others, proliferation is pathogenic and results in further disruption to the retina. This is in contrast to non-mammalian vertebrates, which possess tremendous RPE regenerative potential. Here, we discuss what is known about RPE formation during development in mammalian and non-mammalian vertebrates, we detail the processes by which RPE cells respond to injury, and we describe examples of RPE-to-retina and RPE-to-RPE regeneration in non-mammalian vertebrates. Finally, we outline barriers to RPE-dependent regeneration in mammals that could potentially be overcome to stimulate a regenerative response from the RPE.
Collapse
Affiliation(s)
- Stephanie M George
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Fangfang Lu
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Mishal Rao
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Lyndsay L Leach
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Jeffrey M Gross
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
10
|
Kadkhodaeian HA. Mesenchymal Stem Cells: Signaling Pathways in Transdifferentiation Into Retinal Progenitor Cells. Basic Clin Neurosci 2021; 12:29-42. [PMID: 33995925 PMCID: PMC8114861 DOI: 10.32598/bcn.9.10.510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/25/2018] [Accepted: 02/02/2020] [Indexed: 11/29/2022] Open
Abstract
Several signaling pathways and transcription factors control the cell fate in its in vitro development and differentiation. The orchestrated use of these factors results in cell specification. In coculture methods, many of these factors secrete from host cells but control the process. Today, transcription factors required for retinal progenitor cells are well known, but the generation of these cells from mesenchymal stem cells is an ideal goal. The purpose of the paper is to review novel methods for retinal progenitor cell production and selecting a set of signaling molecules in the presence of adult retinal pigment epithelium and extraocular mesenchyme acting as inducers of retinal cell differentiation.
Collapse
|
11
|
Grocott T, Lozano-Velasco E, Mok GF, Münsterberg AE. The Pax6 master control gene initiates spontaneous retinal development via a self-organising Turing network. Development 2020; 147:dev185827. [PMID: 33214222 PMCID: PMC7774904 DOI: 10.1242/dev.185827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/05/2020] [Indexed: 12/14/2022]
Abstract
Understanding how complex organ systems are assembled from simple embryonic tissues is a major challenge. Across the animal kingdom a great diversity of visual organs are initiated by a 'master control gene' called Pax6, which is both necessary and sufficient for eye development. Yet precisely how Pax6 achieves this deeply homologous function is poorly understood. Using the chick as a model organism, we show that vertebrate Pax6 interacts with a pair of morphogen-coding genes, Tgfb2 and Fst, to form a putative Turing network, which we have computationally modelled. Computer simulations suggest that this gene network is sufficient to spontaneously polarise the developing retina, establishing the first organisational axis of the eye and prefiguring its further development. Our findings reveal how retinal self-organisation may be initiated independently of the highly ordered tissue interactions that help to assemble the eye in vivo These results help to explain how stem cell aggregates spontaneously self-organise into functional eye-cups in vitro We anticipate these findings will help to underpin retinal organoid technology, which holds much promise as a platform for disease modelling, drug development and regenerative therapies.
Collapse
Affiliation(s)
- Timothy Grocott
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | | | | | | |
Collapse
|
12
|
Jabbehdari S, Handa JT. Oxidative stress as a therapeutic target for the prevention and treatment of early age-related macular degeneration. Surv Ophthalmol 2020; 66:423-440. [PMID: 32961209 DOI: 10.1016/j.survophthal.2020.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration, the leading cause of irreversible visual loss among older adults in developed countries, is a chronic, multifactorial, and progressive disease with the development of painless, central vision loss. Retinal pigment epithelial cell dysfunction is a core change in age-related macular degeneration that results from aging and the accumulated effects of genetic and environmental factors that, in part, is both caused by and leads to oxidative stress. In this review, we describe the role of oxidative stress, the cytoprotective oxidative stress pathways, and the impact of oxidative stress on critical cellular processes involved in age-related macular degeneration pathobiology. We also offer targeted therapy that may define how antioxidant therapy can either prevent or improve specific stages of age-related macular degeneration.
Collapse
Affiliation(s)
- Sayena Jabbehdari
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - James T Handa
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
13
|
Kalaskar VK, Alur RP, Li LK, Thomas JW, Sergeev YV, Blain D, Hufnagel RB, Cogliati T, Brooks BP. High-throughput custom capture sequencing identifies novel mutations in coloboma-associated genes: Mutation in DNA-binding domain of retinoic acid receptor beta affects nuclear localization causing ocular coloboma. Hum Mutat 2019; 41:678-695. [PMID: 31816153 PMCID: PMC7027867 DOI: 10.1002/humu.23954] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 10/04/2019] [Accepted: 11/09/2019] [Indexed: 12/11/2022]
Abstract
Uveal coloboma is a potentially blinding congenital ocular malformation caused by the failure of optic fissure closure during the fifth week of human gestation. We performed custom capture high‐throughput screening of 38 known coloboma‐associated genes in 66 families. Suspected causative novel variants were identified in TFAP2A and CHD7, as well as two previously reported variants of uncertain significance in RARB and BMP7. The variant in RARB, unlike previously reported disease mutations in the ligand‐binding domain, was a missense change in the highly conserved DNA‐binding domain predicted to affect the protein's DNA‐binding ability. In vitro studies revealed lower steady‐state protein levels, reduced transcriptional activity, and incomplete nuclear localization of the mutant RARB protein compared with wild‐type. Zebrafish studies showed that human RARB messenger RNA partially reduced the ocular phenotype caused by morpholino knockdown of rarga gene, a zebrafish homolog of human RARB. Our study indicates that sequence alterations in known coloboma genes account for a small percentage of coloboma cases and that mutations in the RARB DNA‐binding domain could result in human disease.
Collapse
Affiliation(s)
- Vijay K Kalaskar
- Pediatric, Developmental & Genetic Ophthalmology Section, Ophthalmic Genetics and Visual Function Branch (OGVFB), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Ramakrishna P Alur
- Pediatric, Developmental & Genetic Ophthalmology Section, Ophthalmic Genetics and Visual Function Branch (OGVFB), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, Maryland
| | - LeeAnn K Li
- Pediatric, Developmental & Genetic Ophthalmology Section, Ophthalmic Genetics and Visual Function Branch (OGVFB), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, Maryland
| | - James W Thomas
- National Institutes of Health Intramural Sequencing Center, National Human Genome Research Institute, NIH, Bethesda, Maryland
| | - Yuri V Sergeev
- Protein Biochemistry and Molecular Modeling Group, OGVFB, NEI, NIH, Bethesda, Maryland
| | - Delphine Blain
- Ophthalmic Clinical Genetics Section, OGVFB, NEI, NIH, Bethesda, Maryland
| | - Robert B Hufnagel
- Medical Genetics and Ophthalmic Genomics Unit, OGVFB, NEI, NIH, Bethesda, Maryland
| | - Tiziana Cogliati
- Pediatric, Developmental & Genetic Ophthalmology Section, Ophthalmic Genetics and Visual Function Branch (OGVFB), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Brian P Brooks
- Pediatric, Developmental & Genetic Ophthalmology Section, Ophthalmic Genetics and Visual Function Branch (OGVFB), National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, Maryland.,Ophthalmic Clinical Genetics Section, OGVFB, NEI, NIH, Bethesda, Maryland
| |
Collapse
|
14
|
Ma X, Li H, Chen Y, Yang J, Chen H, Arnheiter H, Hou L. The transcription factor MITF in RPE function and dysfunction. Prog Retin Eye Res 2019; 73:100766. [DOI: 10.1016/j.preteyeres.2019.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022]
|
15
|
Gamm DM, Clark E, Capowski EE, Singh R. The Role of FGF9 in the Production of Neural Retina and RPE in a Pluripotent Stem Cell Model of Early Human Retinal Development. Am J Ophthalmol 2019; 206:113-131. [PMID: 31078532 DOI: 10.1016/j.ajo.2019.04.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/11/2019] [Accepted: 04/18/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE To investigate the role of fibroblast growth factors (FGFs) in the production of neural retina (NR) and retinal pigmented epithelium (RPE) in a human pluripotent stem cell model of early retinal development. METHODS Human induced pluripotent stem cell (hiPSC) lines from an individual with microphthalmia caused by a functional null mutation (R200Q) in visual system homeobox 2 (VSX2), a transcription factor involved in early NR progenitor cell (NRPC) production, and a normal sibling were differentiated along the retinal and forebrain lineages using an established protocol. Quantitative and global gene expression analyses (microarray and RNAseq) were used to investigate endogenous FGF expression profiles in these cultures over time. Based on these results, mutant and control hiPSC cultures were treated exogenously with selected FGFs and subjected to gene and protein expression analyses to determine their effects on RPE and NR production. RESULTS We found that FGF9 and FGF19 were selectively increased in early hiPSC-derived optic vesicles (OVs) when compared to isogenic cultures of hiPSC-derived forebrain neurospheres. Furthermore, these same FGFs were downregulated over time in (R200Q)VSX2 hiPSC-OVs relative to sibling control hiPSC-OVs. Interestingly, long-term supplementation with FGF9, but not FGF19, partially rescued the mutant retinal phenotype of the (R200Q)VSX2 hiPSC-OV model. However, antagonizing FGF9 in wild-type control hiPSCs did not alter OV development. CONCLUSIONS Our results show that FGF9 acts in concert with VSX2 to promote NR differentiation in hiPSC-OVs and has potential to be used to manipulate early retinogenesis and mitigate ocular defects caused by functional loss of VSX2 activity. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
Affiliation(s)
- David M Gamm
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA; Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| | - Eric Clark
- Department of Cell Biology, Neurobiology, & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Ruchira Singh
- Department of Ophthalmology, University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
16
|
Valensi M, Goldman G, Marchant D, Van Den Berghe L, Jonet L, Daruich A, Robert MP, Krejci E, Klein C, Mascarelli F, Versaux-Botteri C, Moulin A, Putterman M, Guimiot F, Molina T, Terris B, Brémond-Gignac D, Behar-Cohen F, Abitbol MM. Sostdc1 is expressed in all major compartments of developing and adult mammalian eyes. Graefes Arch Clin Exp Ophthalmol 2019; 257:2401-2427. [PMID: 31529323 DOI: 10.1007/s00417-019-04462-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/20/2019] [Accepted: 09/04/2019] [Indexed: 01/16/2023] Open
Abstract
PURPOSE This study was conducted in order to study Sostdc1 expression in rat and human developing and adult eyes. METHODS Using the yeast signal sequence trap screening method, we identified the Sostdc1 cDNA encoding a protein secreted by the adult rat retinal pigment epithelium. We determined by in situ hybridization, RT-PCR, immunohistochemistry, and western blot analysis Sostdc1 gene and protein expression in developing and postnatal rat ocular tissue sections. We also investigated Sostdc1 immunohistolocalization in developing and adult human ocular tissues. RESULTS We demonstrated a prominent Sostdc1 gene expression in the developing rat central nervous system (CNS) and eyes at early developmental stages from E10.5 days postconception (dpc) to E13 dpc. Specific Sostdc1 immunostaining was also detected in most adult cells of rat ocular tissue sections. We also identified the rat ocular embryonic compartments characterized by a specific Sostdc1 immunohistostaining and specific Pax6, Sox2, Otx2, and Vsx2 immunohistostaining from embryonic stages E10.5 to E13 dpc. Furthermore, we determined the localization of SOSTDC1 immunoreactivity in ocular tissue sections of developing and adult human eyes. Indeed, we detected SOSTDC1 immunostaining in developing and adult human retinal pigment epithelium (RPE) and neural retina (NR) as well as in several developing and adult human ocular compartments, including the walls of choroidal and scleral vessels. Of utmost importance, we observed a strong SOSTDC1 expression in a pathological ocular specimen of type 2 Peters' anomaly complicated by retinal neovascularization as well in the walls ofother pathological extra-ocular vessels. CONCLUSION: As rat Sostdc1 and human SOSTDC1 are dual antagonists of the Wnt/β-catenin and BMP signaling pathways, these results underscore the potential crucial roles of these pathways and their antagonists, such as Sostdc1 and SOSTDC1, in developing and adult mammalian normal eyes as well as in syndromic and nonsyndromic congenital eye diseases.
Collapse
Affiliation(s)
- Maud Valensi
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Gabrielle Goldman
- APHP, Service de Pathologie de L'Hôpital Cochin-Hôtel-Dieu, Université Paris Descartes, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Dominique Marchant
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- Sorbonne Paris Cité, UFR SMBH, Laboratoire Hypoxie et poumons, Université Paris 13, EA 2363, 93017, Bobigny, France
| | - Loïc Van Den Berghe
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- Inserm UMR 1037, CRCT (Cancer Research Center of Toulouse), 31037, Toulouse, France
| | - Laurent Jonet
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Alejandra Daruich
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France
| | - Matthieu P Robert
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France
- COGnition and Action Group, UMR 8257, CNRS, Université Paris Descartes, Paris, France
| | - Eric Krejci
- COGnition and Action Group, UMR 8257, CNRS, Université Paris Descartes, Paris, France
| | - Christophe Klein
- Centre d'Imagerie Cellulaire et de Cytométrie (CICC), Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6, Université Paris Descartes - Paris 5, UMR_S 1138, 75006, Paris, France
| | - Frédéric Mascarelli
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Claudine Versaux-Botteri
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
| | - Alexandre Moulin
- Département de Pathologie, Hôpital Ophtalmique Jules-Gonin , 15, avenue de France, 1004, Lausanne, Switzerland
| | - Marc Putterman
- APHP, Service de Pathologie de l'Hôpital Universitaire Necker-Enfants-Malades, Université Paris Descartes, 149 rue de Sèvres, 75015, Paris, France
| | - Fabien Guimiot
- Unité Fonctionnelle de Foeto-Pathologie, Hôpital Universitaire Robert Debré, 48 Boulevard Serrurier, 75019, Paris, France
| | - Thierry Molina
- APHP, Service de Pathologie de l'Hôpital Universitaire Necker-Enfants-Malades, Université Paris Descartes, 149 rue de Sèvres, 75015, Paris, France
| | - Benoît Terris
- APHP, Service de Pathologie de L'Hôpital Cochin-Hôtel-Dieu, Université Paris Descartes, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Dominique Brémond-Gignac
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France
| | - Francine Behar-Cohen
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France
- AP-HP, Service d'Ophtalmologie, Hôpital Universitaire Cochin-Hôtel-Dieu, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Marc M Abitbol
- Centre de Recherches des Cordeliers, UMR_S INSERM 1138, Equipe 17, Université Paris Descartes, 15 rue de l'école de médecine, 75006, Paris, France.
- AP-HP, Hôpital Universitaire Necker-Enfants-Malades, Service d'Ophtalmologie, 149 rue de Sèvres, 75015, Paris, France.
| |
Collapse
|
17
|
Robust induction of retinal pigment epithelium cells from human induced pluripotent stem cells by inhibiting FGF/MAPK signaling. Stem Cell Res 2019; 39:101514. [DOI: 10.1016/j.scr.2019.101514] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 07/02/2019] [Accepted: 07/24/2019] [Indexed: 01/09/2023] Open
|
18
|
Shirahama M, Steinfeld I, Karaiwa A, Taketani S, Vogel-Höpker A, Layer PG, Araki M. Change in the developmental fate of the chick optic vesicle from the neural retina to the telencephalon. Dev Growth Differ 2019; 61:252-262. [PMID: 30843193 DOI: 10.1111/dgd.12599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 11/27/2022]
Abstract
The forebrain develops into the telencephalon, diencephalon, and optic vesicle (OV). The OV further develops into the optic cup, the inner and outer layers of which develop into the neural retina and retinal pigmented epithelium (RPE), respectively. We studied the change in fate of the OV by using embryonic transplantation and explant culture methods. OVs excised from 10-somite stage chick embryos were freed from surrounding tissues (the surface ectoderm and mesenchyme) and were transplanted back to their original position in host embryos. Expression of neural retina-specific genes, such as Rax and Vsx2 (Chx10), was downregulated in the transplants. Instead, expression of the telencephalon-specific gene Emx1 emerged in the proximal region of the transplants, and in the distal part of the transplants close to the epidermis, expression of an RPE-specific gene Mitf was observed. Explant culture studies showed that when OVs were cultured alone, Rax was continuously expressed regardless of surrounding tissues (mesenchyme and epidermis). When OVs without surrounding tissues were cultured in close contact with the anterior forebrain, Rax expression became downregulated in the explants, and Emx1 expression became upregulated. These findings indicate that chick OVs at stage 10 are bi-potential with respect to their developmental fates, either for the neural retina or for the telencephalon, and that the surrounding tissues have a pivotal role in their actual fates. An in vitro tissue culture model suggests that under the influence of the anterior forebrain and/or its surrounding tissues, the OV changes its fate from the retina to the telencephalon.
Collapse
Affiliation(s)
- Misaki Shirahama
- Developmental Neurobiology Laboratory, Nara Women's University, Nara, Japan
| | - Ichie Steinfeld
- Developmental Neurobiology Laboratory, Nara Women's University, Nara, Japan.,Entwicklungsbiologie & Neurogenetik, Technische Universität Darmstadt, Darmstadt, Germany
| | - Akari Karaiwa
- Developmental Neurobiology Laboratory, Nara Women's University, Nara, Japan
| | - Shigeru Taketani
- Department of Biotechnology, Kyoto Institute of Technology, Kyoto, Japan
| | - Astrid Vogel-Höpker
- Entwicklungsbiologie & Neurogenetik, Technische Universität Darmstadt, Darmstadt, Germany
| | - Paul G Layer
- Entwicklungsbiologie & Neurogenetik, Technische Universität Darmstadt, Darmstadt, Germany
| | - Masasuke Araki
- Developmental Neurobiology Laboratory, Nara Women's University, Nara, Japan.,Unit of Neural Development and Regeneration, Nara Medical University, Kashihara, Japan
| |
Collapse
|
19
|
Ji SL, Tang SB. Differentiation of retinal ganglion cells from induced pluripotent stem cells: a review. Int J Ophthalmol 2019; 12:152-160. [PMID: 30662854 DOI: 10.18240/ijo.2019.01.22] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/06/2018] [Indexed: 01/06/2023] Open
Abstract
Glaucoma is a common optic neuropathy that is characterized by the progressive degeneration of axons and the loss of retinal ganglion cells (RGCs). Glaucoma is one of the leading causes of irreversible blindness worldwide. Current glaucoma treatments only slow the progression of RGCs loss. Induced pluripotent stem cells (iPSCs) are capable of differentiating into all three germ layer cell lineages. iPSCs can be patient-specific, making iPSC-derived RGCs a promising candidate for cell replacement. In this review, we focus on discussing the detailed approaches used to differentiate iPSCs into RGCs.
Collapse
Affiliation(s)
- Shang-Li Ji
- Aier Eye Institute, Changsha 410015, Hunan Province, China
| | - Shi-Bo Tang
- Aier School of Ophthalmology, Central South University, Changsha 410015, Hunan Province, China
| |
Collapse
|
20
|
Phelep A, Laouari D, Bharti K, Burtin M, Tammaccaro S, Garbay S, Nguyen C, Vasseur F, Blanc T, Berissi S, Langa-Vives F, Fischer E, Druilhe A, Arnheiter H, Friedlander G, Pontoglio M, Terzi F. MITF - A controls branching morphogenesis and nephron endowment. PLoS Genet 2017; 13:e1007093. [PMID: 29240767 PMCID: PMC5746285 DOI: 10.1371/journal.pgen.1007093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 12/28/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022] Open
Abstract
Congenital nephron number varies widely in the human population and individuals with low nephron number are at risk of developing hypertension and chronic kidney disease. The development of the kidney occurs via an orchestrated morphogenetic process where metanephric mesenchyme and ureteric bud reciprocally interact to induce nephron formation. The genetic networks that modulate the extent of this process and set the final nephron number are mostly unknown. Here, we identified a specific isoform of MITF (MITF-A), a bHLH-Zip transcription factor, as a novel regulator of the final nephron number. We showed that overexpression of MITF-A leads to a substantial increase of nephron number and bigger kidneys, whereas Mitfa deficiency results in reduced nephron number. Furthermore, we demonstrated that MITF-A triggers ureteric bud branching, a phenotype that is associated with increased ureteric bud cell proliferation. Molecular studies associated with an in silico analyses revealed that amongst the putative MITF-A targets, Ret was significantly modulated by MITF-A. Consistent with the key role of this network in kidney morphogenesis, Ret heterozygosis prevented the increase of nephron number in mice overexpressing MITF-A. Collectively, these results uncover a novel transcriptional network that controls branching morphogenesis during kidney development and identifies one of the first modifier genes of nephron endowment. The number of nephrons, the functional unit of kidney, varies widely among humans. Indeed, it has been shown that kidneys may contain from 0.3 to more than 2 million of nephrons. Nephrons are formed during development via a coordinated morphogenetic program in which the metanephric mesenchyme reciprocally and recursively interacts with the ureteric bud. The fine-tuning of this cross-talk determines the final number of nephrons. Strong evidence indicates that suboptimal nephron endowment is associated with an increased risk of hypertension and chronic kidney disease, a major healthcare burden. Indeed, chronic kidney disease is characterized by the progressive decline of renal function towards end stage renal disease, which occurs once a critical number of nephrons has been lost. Elucidating the molecular mechanisms that control nephron endowment is, therefore, a critical issue for public health. However, little is known about the factors that determine the final number of nephrons in the healthy population. Our data showed that nephron endowment is genetically predetermined and identified Mitfa, a bHLH transcription factor, as one of the first modifiers of nephron formation during kidney development. By generating an allelic series of transgenic mice expressing different levels of MITF-A, we discovered that MITF-A promotes final nephron endowment. In addition, we elucidated the molecular mechanisms by which MITF-A promotes nephron formation and identified RET as one of the critical effectors.
Collapse
Affiliation(s)
- Aurélie Phelep
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Denise Laouari
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Kapil Bharti
- Unit on Ocular and Stem Cells Translational Research National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Martine Burtin
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Salvina Tammaccaro
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Serge Garbay
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Clément Nguyen
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Florence Vasseur
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Thomas Blanc
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Sophie Berissi
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | | | - Evelyne Fischer
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Anne Druilhe
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Heinz Arnheiter
- Scientist Emeritus, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Bethesda, MD, United States of America
| | - Gerard Friedlander
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Marco Pontoglio
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Fabiola Terzi
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
21
|
Steinfeld J, Steinfeld I, Bausch A, Coronato N, Hampel ML, Depner H, Layer PG, Vogel-Höpker A. BMP-induced reprogramming of the neural retina into retinal pigment epithelium requires Wnt signalling. Biol Open 2017; 6:979-992. [PMID: 28546339 PMCID: PMC5550904 DOI: 10.1242/bio.018739] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/21/2017] [Indexed: 12/13/2022] Open
Abstract
In vertebrates, the retinal pigment epithelium (RPE) and photoreceptors of the neural retina (NR) comprise a functional unit required for vision. During vertebrate eye development, a conversion of the RPE into NR can be induced by growth factors in vivo at optic cup stages, but the reverse process, the conversion of NR tissue into RPE, has not been reported. Here, we show that bone morphogenetic protein (BMP) signalling can reprogram the NR into RPE at optic cup stages in chick. Shortly after BMP application, expression of Microphthalmia-associated transcription factor (Mitf) is induced in the NR and selective cell death on the basal side of the NR induces an RPE-like morphology. The newly induced RPE differentiates and expresses Melanosomalmatrix protein 115 (Mmp115) and RPE65. BMP-induced Wnt2b expression is observed in regions of the NR that become pigmented. Loss of function studies show that conversion of the NR into RPE requires both BMP and Wnt signalling. Simultaneous to the appearance of ectopic RPE tissue, BMP application reprogrammed the proximal RPE into multi-layered retinal tissue. The newly induced NR expresses visual segment homeobox-containing gene (Vsx2), and the ganglion and photoreceptor cell markers Brn3α and Visinin are detected. Our results show that high BMP concentrations are required to induce the conversion of NR into RPE, while low BMP concentrations can still induce transdifferentiation of the RPE into NR. This knowledge may contribute to the development of efficient standardized protocols for RPE and NR generation for cell replacement therapies.
Collapse
Affiliation(s)
- Jörg Steinfeld
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Ichie Steinfeld
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Alexander Bausch
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Nicola Coronato
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Meggi-Lee Hampel
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Heike Depner
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Paul G Layer
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Astrid Vogel-Höpker
- Fachbereich Biologie, Abteilung Stammzell- und Entwicklungsbiologie, Schnittspahnstraße 13, Darmstadt 64287, Germany
| |
Collapse
|
22
|
Todd L, Palazzo I, Squires N, Mendonca N, Fischer AJ. BMP- and TGFβ-signaling regulate the formation of Müller glia-derived progenitor cells in the avian retina. Glia 2017; 65:1640-1655. [PMID: 28703293 DOI: 10.1002/glia.23185] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/24/2017] [Accepted: 06/13/2017] [Indexed: 01/20/2023]
Abstract
Müller glia-derived progenitor cells (MGPCs) have the capability to regenerate neurons in the retinas of different vertebrate orders. The formation of MGPCs is regulated by a network of cell-signaling pathways. The purpose of this study was to investigate how BMP/Smad1/5/8- and TGFβ/Smad2/3-signaling are coordinated to influence the formation of MGPCs in the chick model system. We find that pSmad1/5/8 is selectively up-regulated in the nuclei of Müller glia following treatment with BMP4, FGF2, or NMDA-induced damage, and this up-regulation is blocked by a dorsomorphin analogue DMH1. By comparison, Smad2/3 is found in the nuclei of Müller glia in untreated retinas, and becomes localized to the cytoplasm following NMDA- or FGF2-treatment. These findings suggest a decrease in TGFβ- and increase in BMP-signaling when MGPCs are known to form. In both NMDA-damaged and FGF2-treated retinas, inhibition of BMP-signaling suppressed the proliferation of MGPCs, whereas inhibition of TGFβ-signaling stimulated the proliferation of MGPCs. Consistent with these findings, TGFβ2 suppressed the formation of MGPCs in NMDA-damaged retinas. Our findings indicate that BMP/TGFβ/Smad-signaling is recruited into the network of signaling pathways that controls the formation of proliferating MGPCs. We conclude that signaling through BMP4/Smad1/5/8 promotes the formation of MGPCs, whereas signaling through TGFβ/Smad2/3 suppresses the formation 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, Ohio, 43210
| | - Isabella Palazzo
- Department of Neuroscience, College of Medicine, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave, Columbus, Ohio, 43210
| | - Natalie Squires
- Department of Neuroscience, College of Medicine, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave, Columbus, Ohio, 43210
| | - Ninoshka Mendonca
- Department of Neuroscience, College of Medicine, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave, Columbus, Ohio, 43210
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave, Columbus, Ohio, 43210
| |
Collapse
|
23
|
Zhao JJ, Afshari NA. Generation of Human Corneal Endothelial Cells via In Vitro Ocular Lineage Restriction of Pluripotent Stem Cells. Invest Ophthalmol Vis Sci 2017; 57:6878-6884. [PMID: 28002562 PMCID: PMC5215466 DOI: 10.1167/iovs.16-20024] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose We generate a renewable supply of corneal endothelial cells (CEC) from human pluripotent stem cells (PSCs) under defined culture conditions. Methods Corneal endothelial cell induction was driven by small molecules in a stepwise fashion of lineage specification. During the initial phase, PSC fate was restricted to the eye field-like state and became eye field stem cells (EFSCs). In the second phase, PSC-derived EFSCs were further directed toward either neural crest lineage or retinal lineage. The CECs were directly induced from ocular neural crest stem cells (NCSCs) by suppressing TGF-β and ROCK signaling. Results Under chemically defined conditions, PSCs were massively converted into EFSCs and subsequently NCSCs. Eye field cell identity was characterized by the expression of key fate restriction factors for early eye field cells, such as PAX6, LHX2, and VSX2. The induction of ocular NCSCs was initiated by promoting WNT signaling in EFSCs. Within 2 weeks of induction, the majority of cells expressed the typical neural crest markers p75NTR and HNK-1. Eye field stem cell-derived NCSCs can be propagated and cryopreserved. Subsequently, a CEC monolayer was induced from adherent NCSCs in the presence of small molecular inhibitors to suppress TGF-β and ROCK signaling. The polygon-shaped CEC-like cells became visible after a week in culture. The NCSC-derived CECs expressed typical CEC markers, such as N-Cadherin and Na+/K+-ATPase. Conclusions A novel small molecule-based approach was developed to derive human CECs from PSCs via ocular lineage specification. Moreover, EFSC-derived NCSCs could serve as an immediate source cell for rapid CEC induction in vitro.
Collapse
Affiliation(s)
- Jiagang J Zhao
- Shiley Eye Institute, Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Natalie A Afshari
- Shiley Eye Institute, Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
24
|
Zhao C, Wang Q, Temple S. Stem cell therapies for retinal diseases: recapitulating development to replace degenerated cells. Development 2017; 144:1368-1381. [PMID: 28400433 PMCID: PMC5399657 DOI: 10.1242/dev.133108] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Retinal degenerative diseases are the leading causes of blindness worldwide. Replacing lost retinal cells via stem cell-based therapies is an exciting, rapidly advancing area of translational research that has already entered the clinic. Here, we review the status of these clinical efforts for several significant retinal diseases, describe the challenges involved and discuss how basic developmental studies have contributed to and are needed to advance clinical goals.
Collapse
Affiliation(s)
- Cuiping Zhao
- Neural Stem Cell Institute, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Qingjie Wang
- Neural Stem Cell Institute, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Sally Temple
- Neural Stem Cell Institute, 1 Discovery Drive, Rensselaer, NY 12144, USA
| |
Collapse
|
25
|
Grigoryan EN, Markitantova YV. Cellular and Molecular Preconditions for Retinal Pigment Epithelium (RPE) Natural Reprogramming during Retinal Regeneration in Urodela. Biomedicines 2016; 4:E28. [PMID: 28536395 PMCID: PMC5344269 DOI: 10.3390/biomedicines4040028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/26/2016] [Accepted: 11/26/2016] [Indexed: 12/25/2022] Open
Abstract
Many regeneration processes in animals are based on the phenomenon of cell reprogramming followed by proliferation and differentiation in a different specialization direction. An insight into what makes natural (in vivo) cell reprogramming possible can help to solve a number of biomedical problems. In particular, the first problem is to reveal the intrinsic properties of the cells that are necessary and sufficient for reprogramming; the second, to evaluate these properties and, on this basis, to reveal potential endogenous sources for cell substitution in damaged tissues; and the third, to use the acquired data for developing approaches to in vitro cell reprogramming in order to obtain a cell reserve for damaged tissue repair. Normal cells of the retinal pigment epithelium (RPE) in newts (Urodela) can change their specialization and transform into retinal neurons and ganglion cells (i.e., actualize their retinogenic potential). Therefore, they can serve as a model that provides the possibility to identify factors of the initial competence of vertebrate cells for reprogramming in vivo. This review deals mainly with the endogenous properties of native newt RPE cells themselves and, to a lesser extent, with exogenous mechanisms regulating the process of reprogramming, which are actively discussed.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia.
| | - Yuliya V Markitantova
- Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia.
| |
Collapse
|
26
|
Hasegawa Y, Takata N, Okuda S, Kawada M, Eiraku M, Sasai Y. Emergence of dorsal-ventral polarity in ESC-derived retinal tissue. Development 2016; 143:3895-3906. [PMID: 27633992 DOI: 10.1242/dev.134601] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 09/01/2016] [Indexed: 01/08/2023]
Abstract
We previously demonstrated that mouse embryonic stem cell (mESC)-derived retinal epithelium self-forms an optic cup-like structure. In the developing retina, the dorsal and ventral sides differ in terms of local gene expression and morphological features. This aspect has not yet been shown in vitro Here, we demonstrate that mESC-derived retinal tissue spontaneously acquires polarity reminiscent of the dorsal-ventral (D-V) patterning of the embryonic retina. Tbx5 and Vax2 were expressed in a mutually exclusive manner, as seen in vivo Three-dimensional morphometric analysis showed that the in vitro-formed optic cup often contains cleft structures resembling the embryonic optic fissure. To elucidate the mechanisms underlying the spontaneous D-V polarization of mESC-derived retina, we examined the effects of patterning factors, and found that endogenous BMP signaling plays a predominant role in the dorsal specification. Further analysis revealed that canonical Wnt signaling, which was spontaneously activated at the proximal region, acts upstream of BMP signaling for dorsal specification. These observations suggest that D-V polarity could be established within the self-formed retinal neuroepithelium by intrinsic mechanisms involving the spatiotemporal regulation of canonical Wnt and BMP signals.
Collapse
Affiliation(s)
- Yuiko Hasegawa
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan.,Laboratory for in vitro Histogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan
| | - Nozomu Takata
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan.,Laboratory for in vitro Histogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan
| | - Satoru Okuda
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan.,Laboratory for in vitro Histogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan
| | - Masako Kawada
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan.,Laboratory for in vitro Histogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan
| | - Mototsugu Eiraku
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan .,Laboratory for in vitro Histogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan
| | - Yoshiki Sasai
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo, Kobe 650-0047, Japan
| |
Collapse
|
27
|
Choudhary P, Booth H, Gutteridge A, Surmacz B, Louca I, Steer J, Kerby J, Whiting PJ. Directing Differentiation of Pluripotent Stem Cells Toward Retinal Pigment Epithelium Lineage. Stem Cells Transl Med 2016; 6:490-501. [PMID: 28191760 PMCID: PMC5442825 DOI: 10.5966/sctm.2016-0088] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022] Open
Abstract
Development of efficient and reproducible conditions for directed differentiation of pluripotent stem cells into specific cell types is important not only to understand early human development but also to enable more practical applications, such as in vitro disease modeling, drug discovery, and cell therapies. The differentiation of stem cells to retinal pigment epithelium (RPE) in particular holds promise as a source of cells for therapeutic replacement in age‐related macular degeneration. Here we show development of an efficient method for deriving homogeneous RPE populations in a period of 45 days using an adherent, monolayer system and defined xeno‐free media and matrices. The method utilizes sequential inhibition and activation of the Activin and bone morphogenetic protein signaling pathways and can be applied to both human embryonic stem cells and induced pluripotent stem cells as the starting population. In addition, we use whole genome transcript analysis to characterize cells at different stages of differentiation that provides further understanding of the developmental dynamics and fate specification of RPE. We show that with the described method, RPE develop through stages consistent with their formation during embryonic development. This characterization— together with the absence of steps involving embryoid bodies, three‐dimensional culture, or manual dissections, which are common features of other protocols—makes this process very attractive for use in research as well as for clinical applications. Stem Cells Translational Medicine2017;6:490–501
Collapse
Affiliation(s)
- Parul Choudhary
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| | - Heather Booth
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| | - Alex Gutteridge
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| | - Beata Surmacz
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| | - Irene Louca
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| | - Juliette Steer
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| | - Julie Kerby
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| | - Paul John Whiting
- Pfizer Neuroscience and Pain Research Unit, Great Abington, Cambridge, United Kingdom
| |
Collapse
|
28
|
Wang Z, Yasugi S, Ishii Y. Chx10 functions as a regulator of molecular pathways controlling the regional identity in the primordial retina. Dev Biol 2016; 413:104-11. [PMID: 27001188 DOI: 10.1016/j.ydbio.2016.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/01/2016] [Accepted: 03/17/2016] [Indexed: 11/29/2022]
Abstract
The light-sensitive neural retina (NR) and the retinal pigmented epithelium (RPE) develop from a common primordium, the optic vesicle, raising the question of how they acquire and maintain distinct identities. Here, we demonstrate that sustained misexpression of the Chx10 homeobox gene in the presumptive RPE in chick suppresses accumulation of melanin pigments and promotes ectopic NR-like neural differentiation. This phenotypic change involved ectopic expression of NR transcription factor genes, Sox2, Six3, Rx1 and Optx2, which, when misexpressed, counteracted RPE development without upregulating Chx10. These results suggest that Chx10 can function as a cell autonomous regulator of the regional identity in the primordial retina, presumably through a downstream transcriptional cascade.
Collapse
Affiliation(s)
- Zi Wang
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Sadao Yasugi
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Yasuo Ishii
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan.
| |
Collapse
|
29
|
Pandit T, Jidigam VK, Patthey C, Gunhaga L. Neural retina identity is specified by lens-derived BMP signals. Development 2015; 142:1850-9. [PMID: 25968316 PMCID: PMC4440930 DOI: 10.1242/dev.123653] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The eye has served as a classical model to study cell specification and tissue induction for over a century. Nevertheless, the molecular mechanisms that regulate the induction and maintenance of eye-field cells, and the specification of neural retina cells are poorly understood. Moreover, within the developing anterior forebrain, how prospective eye and telencephalic cells are differentially specified is not well defined. In the present study, we have analyzed these issues by manipulating signaling pathways in intact chick embryo and explant assays. Our results provide evidence that at blastula stages, BMP signals inhibit the acquisition of eye-field character, but from neural tube/optic vesicle stages, BMP signals from the lens are crucial for the maintenance of eye-field character, inhibition of dorsal telencephalic cell identity and specification of neural retina cells. Subsequently, our results provide evidence that a Rax2-positive eye-field state is not sufficient for the progress to a neural retina identity, but requires BMP signals. In addition, our results argue against any essential role of Wnt or FGF signals during the specification of neural retina cells, but provide evidence that Wnt signals together with BMP activity are sufficient to induce cells of retinal pigment epithelial character. We conclude that BMP activity emanating from the lens ectoderm maintains eye-field identity, inhibits telencephalic character and induces neural retina cells. Our findings link the requirement of the lens ectoderm for neural retina specification with the molecular mechanism by which cells in the forebrain become specified as neural retina by BMP activity. SUMMARY: BMP signals from the lens are crucial to maintain eye-field character, inhibit dorsal telencephalic cell identity, and specificy neural retina cells in chick embryos.
Collapse
Affiliation(s)
- Tanushree Pandit
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| | - Vijay K Jidigam
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| | - Cedric Patthey
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| | - Lena Gunhaga
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| |
Collapse
|
30
|
Huang J, Liu Y, Oltean A, Beebe DC. Bmp4 from the optic vesicle specifies murine retina formation. Dev Biol 2015; 402:119-26. [PMID: 25792196 DOI: 10.1016/j.ydbio.2015.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/06/2015] [Accepted: 03/07/2015] [Indexed: 01/30/2023]
Abstract
Previous studies of mouse embryos concluded that after the optic vesicle evaginates from the ventral forebrain and contacts the surface ectoderm, signals from the ectoderm specify the distal region of the optic vesicle to become retina and signals from the optic vesicle induce the lens. Germline deletion of Bmp4 resulted in failure of lens formation. We performed conditional deletion of Bmp4 from the optic vesicle to test the function of Bmp4 in murine eye development. The optic vesicle evaginated normally and contacted the surface ectoderm. Lens induction did not occur. The optic cup failed to form and the expression of retina-specific genes decreased markedly in the distal optic vesicle. Instead, cells in the prospective retina expressed genes characteristic of the retinal pigmented epithelium. We conclude that Bmp4 is required for retina specification in mice. In the absence of Bmp4, formation of the retinal pigmented epithelium is the default differentiation pathway of the optic vesicle. Differences in the signaling pathways required for specification of the retina and retinal pigmented epithelium in chicken and mouse embryos suggest major changes in signaling during the evolution of the vertebrate eye.
Collapse
Affiliation(s)
- Jie Huang
- Department of Ophthalmology and Visual Science, USA
| | - Ying Liu
- Department of Ophthalmology and Visual Science, USA
| | - Alina Oltean
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, USA
| | - David C Beebe
- Department of Ophthalmology and Visual Science, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, USA.
| |
Collapse
|
31
|
Pillai-Kastoori L, Wen W, Morris AC. Keeping an eye on SOXC proteins. Dev Dyn 2015; 244:367-376. [PMID: 25476579 PMCID: PMC4344926 DOI: 10.1002/dvdy.24235] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/10/2014] [Accepted: 11/28/2014] [Indexed: 12/17/2022] Open
Abstract
The formation of a mature, functional eye requires a complex series of cell proliferation, migration, induction among different germinal layers, and cell differentiation. These processes are regulated by extracellular cues such as the Wnt/BMP/Hh/Fgf signaling pathways, as well as cell intrinsic transcription factors that specify cell fate. In this review article, we provide an overview of stages of embryonic eye morphogenesis, extrinsic and intrinsic factors that are required for each stage, and pediatric ocular diseases that are associated with defective eye development. In addition, we focus on recent findings about the roles of the SOXC proteins in regulating vertebrate ocular development and implicating SOXC mutations in human ocular malformations.
Collapse
Affiliation(s)
| | - Wen Wen
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Ann C. Morris
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
32
|
Heermann S, Schütz L, Lemke S, Krieglstein K, Wittbrodt J. Eye morphogenesis driven by epithelial flow into the optic cup facilitated by modulation of bone morphogenetic protein. eLife 2015; 4. [PMID: 25719386 PMCID: PMC4337729 DOI: 10.7554/elife.05216] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/26/2015] [Indexed: 01/07/2023] Open
Abstract
The hemispheric, bi-layered optic cup forms from an oval optic vesicle during early vertebrate eye development through major morphological transformations. The overall basal surface, facing the developing lens, is increasing, while, at the same time, the space basally occupied by individual cells is decreasing. This cannot be explained by the classical view of eye development. Using zebrafish (Danio rerio) as a model, we show that the lens-averted epithelium functions as a reservoir that contributes to the growing neuroretina through epithelial flow around the distal rims of the optic cup. We propose that this flow couples morphogenesis and retinal determination. Our 4D data indicate that future stem cells flow from their origin in the lens-averted domain of the optic vesicle to their destination in the ciliary marginal zone. BMP-mediated inhibition of the flow results in ectopic neuroretina in the RPE domain. Ultimately the ventral fissure fails to close resulting in coloboma. DOI:http://dx.doi.org/10.7554/eLife.05216.001 The eye is our most important organ for sensing and recognizing our environment. In humans and other vertebrates, the eye forms from an outgrowth of the brain as the embryo develops. This outgrowth is called the optic vesicle and it is rapidly transformed into a cup-shaped structure known as the optic cup. Defects in this process prevent the optic cup from closing completely, which leads to a severe condition called Coloboma—one of the most frequent causes of blindness in children. The optic cup has two distinct layers: the inside layer—known as the neuroretina—contains light sensitive cells and is surrounded by the other layer called the pigmented epithelium. It is thought that the neural retina is made from cells from the side of the optic vesicle that faces the lens, and the pigmented epithelium is formed by cells from the other side of the vesicle. This is a plausible explanation and is well accepted, but it cannot explain how the neuroretina can become five times larger as the cup forms. Heermann et al. addressed this problem by using four-dimensional in vivo microscopy to follow individual cells as the optic cup forms in living zebrafish embryos. The experiments show that the neuroretina is made of cells from both sides of the optic vesicle. Cells from the back of the optic vesicle (furthest away from the lens) join the rest of the cells by moving around the outside rim of the cup. Further experiments found that a signaling molecule called BMP—which is crucial to the normal development of the eye—controls the flow of cells around the developing optic cup. This factor needs to be carefully controlled during the development of the eye; when BMP activity was artificially increased, the flow of cells stopped, resulting in neuroretinal tissue developing in the wrong place (in the outer layer of the optic cup). The experiments also reveal that the stem cells in the retina—which divide to produce new cells throughout the life of the zebrafish—originate from two distinct areas in the optic vesicle. Heermann et al.'s findings challenge the textbook model of eye development by revealing that cells from both sides of the optic vesicle contribute to the neuroretina and that retinal stem cells originate from a specific place in the developing eye. A future challenge will be to understand how the movement of the cells into the neuroretina is coordinated to make a perfectly shaped eye. DOI:http://dx.doi.org/10.7554/eLife.05216.002
Collapse
Affiliation(s)
- Stephan Heermann
- Centre for Organismal Studies Heidelberg, Ruprecht Karls Universität, Heidelberg, Germany
| | - Lucas Schütz
- Centre for Organismal Studies Heidelberg, Ruprecht Karls Universität, Heidelberg, Germany
| | - Steffen Lemke
- Centre for Organismal Studies Heidelberg, Ruprecht Karls Universität, Heidelberg, Germany
| | - Kerstin Krieglstein
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University Freiburg, Freiburg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies Heidelberg, Ruprecht Karls Universität, Heidelberg, Germany
| |
Collapse
|
33
|
Generation of a ciliary margin-like stem cell niche from self-organizing human retinal tissue. Nat Commun 2015; 6:6286. [PMID: 25695148 DOI: 10.1038/ncomms7286] [Citation(s) in RCA: 237] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 01/12/2015] [Indexed: 12/19/2022] Open
Abstract
In the developing neural retina (NR), multipotent stem cells within the ciliary margin (CM) contribute to de novo retinal tissue growth. We recently reported the ability of human embryonic stem cells (hESCs) to self-organize stratified NR using a three-dimensional culture technique. Here we report the emergence of CM-like stem cell niches within human retinal tissue. First, we developed a culture method for selective NR differentiation by timed BMP4 treatment. We then found that inhibiting GSK3 and FGFR induced the transition from NR tissue to retinal pigment epithelium (RPE), and that removing this inhibition facilitated the reversion of this RPE-like tissue back to the NR fate. This step-wise induction-reversal method generated tissue aggregates with RPE at the margin of central-peripherally polarized NR. We demonstrate that the NR-RPE boundary tissue further self-organizes a niche for CM stem cells that functions to expand the NR peripherally by de novo progenitor generation.
Collapse
|
34
|
Grigoryan EN. Competence factors of retinal pigment epithelium cells for reprogramming in the neuronal direction during retinal regeneration in newts. BIOL BULL+ 2015. [DOI: 10.1134/s1062359015010045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
35
|
Kuznetsova AV, Kurinov AM, Aleksandrova MA. Cell models to study regulation of cell transformation in pathologies of retinal pigment epithelium. J Ophthalmol 2014; 2014:801787. [PMID: 25177495 PMCID: PMC4142280 DOI: 10.1155/2014/801787] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/16/2014] [Accepted: 06/30/2014] [Indexed: 11/25/2022] Open
Abstract
The retinal pigment epithelium (RPE) plays a key role in the development of many eye diseases leading to visual impairment and even blindness. Cell culture models of pathological changes in the RPE make it possible to study factors responsible for these changes and signaling pathways coordinating cellular and molecular mechanisms of cell interactions under pathological conditions. Moreover, they give an opportunity to reveal target cells and develop effective specific treatment for degenerative and dystrophic diseases of the retina. In this review, data are presented on RPE cell sources for culture models, approaches to RPE cell culturing, phenotypic changes of RPE cells in vitro, the role of signal pathways, and possibilities for their regulation in pathological processes.
Collapse
Affiliation(s)
- Alla V. Kuznetsova
- N.K. Koltsov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow 119334, Russia
| | - Alexander M. Kurinov
- N.K. Koltsov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow 119334, Russia
| | - Maria A. Aleksandrova
- N.K. Koltsov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow 119334, Russia
| |
Collapse
|
36
|
Capowski EE, Simonett JM, Clark EM, Wright LS, Howden SE, Wallace KA, Petelinsek AM, Pinilla I, Phillips MJ, Meyer JS, Schneider BL, Thomson JA, Gamm DM. Loss of MITF expression during human embryonic stem cell differentiation disrupts retinal pigment epithelium development and optic vesicle cell proliferation. Hum Mol Genet 2014; 23:6332-44. [PMID: 25008112 DOI: 10.1093/hmg/ddu351] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Microphthalmia-associated transcription factor (MITF) is a master regulator of pigmented cell survival and differentiation with direct transcriptional links to cell cycle, apoptosis and pigmentation. In mouse, Mitf is expressed early and uniformly in optic vesicle (OV) cells as they evaginate from the developing neural tube, and null Mitf mutations result in microphthalmia and pigmentation defects. However, homozygous mutations in MITF have not been identified in humans; therefore, little is known about its role in human retinogenesis. We used a human embryonic stem cell (hESC) model that recapitulates numerous aspects of retinal development, including OV specification and formation of retinal pigment epithelium (RPE) and neural retina progenitor cells (NRPCs), to investigate the earliest roles of MITF. During hESC differentiation toward a retinal lineage, a subset of MITF isoforms was expressed in a sequence and tissue distribution similar to that observed in mice. In addition, we found that promoters for the MITF-A, -D and -H isoforms were directly targeted by Visual Systems Homeobox 2 (VSX2), a transcription factor involved in patterning the OV toward a NRPC fate. We then manipulated MITF RNA and protein levels at early developmental stages and observed decreased expression of eye field transcription factors, reduced early OV cell proliferation and disrupted RPE maturation. This work provides a foundation for investigating MITF and other highly complex, multi-purposed transcription factors in a dynamic human developmental model system.
Collapse
Affiliation(s)
| | | | | | | | - Sara E Howden
- Morgridge Institute for Research, Madison, WI 53715, USA
| | | | | | - Isabel Pinilla
- Department of Ophthalmology, University Hospital Lozano Blesa, Zaragoza 50009, Spain, Aragon Institute of Health Sciences, Zaragoza 50009, Spain
| | | | - Jason S Meyer
- Department of Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Bernard L Schneider
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI 53715, USA, Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53715, USA and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - David M Gamm
- Waisman Center, McPherson Eye Research Institute and Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA,
| |
Collapse
|
37
|
Gjorevski N, Ranga A, Lutolf MP. Bioengineering approaches to guide stem cell-based organogenesis. Development 2014; 141:1794-804. [PMID: 24757002 DOI: 10.1242/dev.101048] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During organogenesis, various molecular and physical signals are orchestrated in space and time to sculpt multiple cell types into functional tissues and organs. The complex and dynamic nature of the process has hindered studies aimed at delineating morphogenetic mechanisms in vivo, particularly in mammals. Recent demonstrations of stem cell-driven tissue assembly in culture offer a powerful new tool for modeling and dissecting organogenesis. However, despite the highly organotypic nature of stem cell-derived tissues, substantial differences set them apart from their in vivo counterparts, probably owing to the altered microenvironment in which they reside and the lack of mesenchymal influences. Advances in the biomaterials and microtechnology fields have, for example, afforded a high degree of spatiotemporal control over the cellular microenvironment, making it possible to interrogate the effects of individual microenvironmental components in a modular fashion and rapidly identify organ-specific synthetic culture models. Hence, bioengineering approaches promise to bridge the gap between stem cell-driven tissue formation in culture and morphogenesis in vivo, offering mechanistic insight into organogenesis and unveiling powerful new models for drug discovery, as well as strategies for tissue regeneration in the clinic. We draw on several examples of stem cell-derived organoids to illustrate how bioengineering can contribute to tissue formation ex vivo. We also discuss the challenges that lie ahead and potential ways to overcome them.
Collapse
Affiliation(s)
- Nikolche Gjorevski
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
| | | | | |
Collapse
|
38
|
Luz-Madrigal A, Grajales-Esquivel E, McCorkle A, DiLorenzo AM, Barbosa-Sabanero K, Tsonis PA, Del Rio-Tsonis K. Reprogramming of the chick retinal pigmented epithelium after retinal injury. BMC Biol 2014; 12:28. [PMID: 24742279 PMCID: PMC4026860 DOI: 10.1186/1741-7007-12-28] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 03/31/2014] [Indexed: 01/01/2023] Open
Abstract
Background One of the promises in regenerative medicine is to regenerate or replace damaged tissues. The embryonic chick can regenerate its retina by transdifferentiation of the retinal pigmented epithelium (RPE) and by activation of stem/progenitor cells present in the ciliary margin. These two ways of regeneration occur concomitantly when an external source of fibroblast growth factor 2 (FGF2) is present after injury (retinectomy). During the process of transdifferentiation, the RPE loses its pigmentation and is reprogrammed to become neuroepithelium, which differentiates to reconstitute the different cell types of the neural retina. Somatic mammalian cells can be reprogrammed to become induced pluripotent stem cells by ectopic expression of pluripotency-inducing factors such as Oct4, Sox2, Klf4, c-Myc and in some cases Nanog and Lin-28. However, there is limited information concerning the expression of these factors during natural regenerative processes. Organisms that are able to regenerate their organs could share similar mechanisms and factors with the reprogramming process of somatic cells. Herein, we investigate the expression of pluripotency-inducing factors in the RPE after retinectomy (injury) and during transdifferentiation in the presence of FGF2. Results We present evidence that upon injury, the quiescent (p27Kip1+/BrdU-) RPE cells transiently dedifferentiate and express sox2, c-myc and klf4 along with eye field transcriptional factors and display a differential up-regulation of alternative splice variants of pax6. However, this transient process of dedifferentiation is not sustained unless FGF2 is present. We have identified lin-28 as a downstream target of FGF2 during the process of retina regeneration. Moreover, we show that overexpression of lin-28 after retinectomy was sufficient to induce transdifferentiation of the RPE in the absence of FGF2. Conclusion These findings delineate in detail the molecular changes that take place in the RPE during the process of transdifferentiation in the embryonic chick, and specifically identify Lin-28 as an important factor in this process. We propose a novel model in which injury signals initiate RPE dedifferentiation, while FGF2 up-regulates Lin-28, allowing for RPE transdifferentiation to proceed.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Katia Del Rio-Tsonis
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| |
Collapse
|
39
|
Boeckx C, Benítez-Burraco A. The shape of the human language-ready brain. Front Psychol 2014; 5:282. [PMID: 24772099 PMCID: PMC3983487 DOI: 10.3389/fpsyg.2014.00282] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/17/2014] [Indexed: 12/14/2022] Open
Abstract
Our core hypothesis is that the emergence of our species-specific language-ready brain ought to be understood in light of the developmental changes expressed at the levels of brain morphology and neural connectivity that occurred in our species after the split from Neanderthals–Denisovans and that gave us a more globular braincase configuration. In addition to changes at the cortical level, we hypothesize that the anatomical shift that led to globularity also entailed significant changes at the subcortical level. We claim that the functional consequences of such changes must also be taken into account to gain a fuller understanding of our linguistic capacity. Here we focus on the thalamus, which we argue is central to language and human cognition, as it modulates fronto-parietal activity. With this new neurobiological perspective in place, we examine its possible molecular basis. We construct a candidate gene set whose members are involved in the development and connectivity of the thalamus, in the evolution of the human head, and are known to give rise to language-associated cognitive disorders. We submit that the new gene candidate set opens up new windows into our understanding of the genetic basis of our linguistic capacity. Thus, our hypothesis aims at generating new testing grounds concerning core aspects of language ontogeny and phylogeny.
Collapse
Affiliation(s)
- Cedric Boeckx
- Catalan Institute for Advanced Studies and Research (ICREA) Barcelona, Spain ; Department of Linguistics, Universitat de Barcelona Barcelona, Spain
| | | |
Collapse
|
40
|
Layer PG, Araki M, Vogel-Höpker A. New concepts for reconstruction of retinal and pigment epithelial tissues. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/eop.10.42] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
41
|
Hägglund AC, Berghard A, Carlsson L. Canonical Wnt/β-catenin signalling is essential for optic cup formation. PLoS One 2013; 8:e81158. [PMID: 24324671 PMCID: PMC3852023 DOI: 10.1371/journal.pone.0081158] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/09/2013] [Indexed: 12/17/2022] Open
Abstract
A multitude of signalling pathways are involved in the process of forming an eye. Here we demonstrate that β-catenin is essential for eye development as inactivation of β-catenin prior to cellular specification in the optic vesicle caused anophthalmia in mice. By achieving this early and tissue-specific β-catenin inactivation we find that retinal pigment epithelium (RPE) commitment was blocked and eye development was arrested prior to optic cup formation due to a loss of canonical Wnt signalling in the dorsal optic vesicle. Thus, these results show that Wnt/β-catenin signalling is required earlier and play a more central role in eye development than previous studies have indicated. In our genetic model system a few RPE cells could escape β-catenin inactivation leading to the formation of a small optic rudiment. The optic rudiment contained several neural retinal cell classes surrounded by an RPE. Unlike the RPE cells, the neural retinal cells could be β-catenin-negative revealing that differentiation of the neural retinal cell classes is β-catenin-independent. Moreover, although dorsoventral patterning is initiated in the mutant optic vesicle, the neural retinal cells in the optic rudiment displayed almost exclusively ventral identity. Thus, β-catenin is required for optic cup formation, commitment to RPE cells and maintenance of dorsal identity of the retina.
Collapse
Affiliation(s)
| | - Anna Berghard
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Leif Carlsson
- Umeå Center for Molecular Medicine, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
42
|
Steinfeld J, Steinfeld I, Coronato N, Hampel ML, Layer PG, Araki M, Vogel-Höpker A. RPE specification in the chick is mediated by surface ectoderm-derived BMP and Wnt signalling. Development 2013; 140:4959-69. [PMID: 24227655 DOI: 10.1242/dev.096990] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The retinal pigment epithelium (RPE) is indispensable for vertebrate eye development and vision. In the classical model of optic vesicle patterning, the surface ectoderm produces fibroblast growth factors (FGFs) that specify the neural retina (NR) distally, whereas TGFβ family members released from the proximal mesenchyme are involved in RPE specification. However, we previously proposed that bone morphogenetic proteins (BMPs) released from the surface ectoderm are essential for RPE specification in chick. We now show that the BMP- and Wnt-expressing surface ectoderm is required for RPE specification. We reveal that Wnt signalling from the overlying surface ectoderm is involved in restricting BMP-mediated RPE specification to the dorsal optic vesicle. Wnt2b is expressed in the dorsal surface ectoderm and subsequently in dorsal optic vesicle cells. Activation of Wnt signalling by implanting Wnt3a-soaked beads or inhibiting GSK3β at optic vesicle stages inhibits NR development and converts the entire optic vesicle into RPE. Surface ectoderm removal at early optic vesicle stages or inhibition of Wnt, but not Wnt/β-catenin, signalling prevents pigmentation and downregulates the RPE regulatory gene Mitf. Activation of BMP or Wnt signalling can replace the surface ectoderm to rescue MITF expression and optic cup formation. We provide evidence that BMPs and Wnts cooperate via a GSK3β-dependent but β-catenin-independent pathway at the level of pSmad to ensure RPE specification in dorsal optic vesicle cells. We propose a new dorsoventral model of optic vesicle patterning, whereby initially surface ectoderm-derived Wnt signalling directs dorsal optic vesicle cells to develop into RPE through a stabilising effect of BMP signalling.
Collapse
Affiliation(s)
- Jörg Steinfeld
- Fachgebiet Entwicklungsbiologie und Neurogenetik, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Fuhrmann S, Zou C, Levine EM. Retinal pigment epithelium development, plasticity, and tissue homeostasis. Exp Eye Res 2013; 123:141-50. [PMID: 24060344 DOI: 10.1016/j.exer.2013.09.003] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/05/2013] [Accepted: 09/07/2013] [Indexed: 12/13/2022]
Abstract
The retinal pigment epithelium (RPE) is a simple epithelium interposed between the neural retina and the choroid. Although only 1 cell-layer in thickness, the RPE is a virtual workhorse, acting in several capacities that are essential for visual function and preserving the structural and physiological integrities of neighboring tissues. Defects in RPE function, whether through chronic dysfunction or age-related decline, are associated with retinal degenerative diseases including age-related macular degeneration. As such, investigations are focused on developing techniques to replace RPE through stem cell-based methods, motivated primarily because of the seemingly limited regeneration or self-repair properties of mature RPE. Despite this, RPE cells have an unusual capacity to transdifferentiate into various cell types, with the particular fate choices being highly context-dependent. In this review, we describe recent findings elucidating the mechanisms and steps of RPE development and propose a developmental framework for understanding the apparent contradiction in the capacity for low self-repair versus high transdifferentiation.
Collapse
Affiliation(s)
- Sabine Fuhrmann
- Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA.
| | - ChangJiang Zou
- Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA.
| | - Edward M Levine
- Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA.
| |
Collapse
|
44
|
Inoue J, Ueda Y, Bando T, Mito T, Noji S, Ohuchi H. The expression of LIM-homeobox genes,Lhx1andLhx5,in the forebrain is essential for neural retina differentiation. Dev Growth Differ 2013; 55:668-75. [DOI: 10.1111/dgd.12074] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Junji Inoue
- Department of Cytology and Histology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; 2-5-1 Shikata-cho; Okayama; 700-8558; Japan
| | - Yuuki Ueda
- Department of Life Systems; Institute of Technology and Science; The University of Tokushima Graduate School; 2-1 Minami-Josanjima-cho; Tokushima; 770-8506; Japan
| | - Tetsuya Bando
- Department of Cytology and Histology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; 2-5-1 Shikata-cho; Okayama; 700-8558; Japan
| | - Taro Mito
- Department of Life Systems; Institute of Technology and Science; The University of Tokushima Graduate School; 2-1 Minami-Josanjima-cho; Tokushima; 770-8506; Japan
| | - Sumihare Noji
- Department of Life Systems; Institute of Technology and Science; The University of Tokushima Graduate School; 2-1 Minami-Josanjima-cho; Tokushima; 770-8506; Japan
| | - Hideyo Ohuchi
- Department of Cytology and Histology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; 2-5-1 Shikata-cho; Okayama; 700-8558; Japan
| |
Collapse
|
45
|
Reynolds J, Lamba DA. Human embryonic stem cell applications for retinal degenerations. Exp Eye Res 2013; 123:151-60. [PMID: 23880530 DOI: 10.1016/j.exer.2013.07.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/27/2013] [Accepted: 07/08/2013] [Indexed: 12/12/2022]
Abstract
Loss of vision in severe retinal degenerations often is a result of photoreceptor cell or retinal pigment epithelial cell death or dysfunction. Cell replacement therapy has the potential to restore useful vision for these individuals especially after they have lost most or all of their light-sensing cells in the eye. A reliable, well-characterized source of retinal cells will be needed for replacement purposes. Human embryonic stem cells (ES cells) can provide an unlimited source of replacement retinal cells to take over the function of lost cells in the eye. The author's intent for this review is to provide an historical overview of the field of embryonic stem cells with relation to the retina. The review will provide a quick primer on key pathways involved in the development of the neural retina and RPE followed by a discussion of the various protocols out in the literature for generating these cells from non-human and human embryonic stem cells and end with in vivo application of ES cell-derived photoreceptors and RPE cells.
Collapse
Affiliation(s)
- Joseph Reynolds
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA
| | - Deepak A Lamba
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA.
| |
Collapse
|
46
|
Zhang R, Huang H, Cao P, Wang Z, Chen Y, Pan Y. Sma- and Mad-related protein 7 (Smad7) is required for embryonic eye development in the mouse. J Biol Chem 2013; 288:10275-85. [PMID: 23426374 PMCID: PMC3624411 DOI: 10.1074/jbc.m112.416719] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 02/19/2013] [Indexed: 01/16/2023] Open
Abstract
Smad7 is an intracellular inhibitory protein that antagonizes the signaling of TGF-β family members. Deletion of Smad7 in the mouse leads to an abnormality in heart development. However, whether Smad7 has a functional role in the development of other organs has been elusive. Here we present evidence that Smad7 imparts a role to eye development in the mouse. Smad7 is expressed in both the lens and retina in the developing embryonic eye. Depletion of Smad7 caused various degrees of coloboma and microphthalmia with alterations in cell apoptosis and proliferation in eyes. Smad7 was implicated in lens differentiation but was not required for the induction of the lens placode. The development of the periocular mesenchyme was retarded with the down-regulation of Bmp7 and Pitx2 in mutant mice. Retinal spatial patterning was affected by Smad7 deletion and was accompanied by altered bone morphogenetic protein (BMP) signaling. At late gestation stages, TGF-β signaling was up-regulated in the differentiating retina. Smad7 mutant mice displayed an expanded optic disc with increasing of sonic hedgehog (SHH) signaling. Furthermore, loss of Smad7 led to a temporal change in retinal neurogenesis. In conclusion, our study suggests that Smad7 is essential for eye development. In addition, our data indicate that alterations in the signaling of BMP, TGF-β, and SHH likely underlie the defects in eye development caused by Smad7 deletion.
Collapse
Affiliation(s)
- Rui Zhang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Heng Huang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Peijuan Cao
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenzhen Wang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Chen
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi Pan
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
47
|
Ou J, Bharti K, Nodari A, Bertuzzi S, Arnheiter H. Vax1/2 genes counteract Mitf-induced respecification of the retinal pigment epithelium. PLoS One 2013; 8:e59247. [PMID: 23555005 PMCID: PMC3598659 DOI: 10.1371/journal.pone.0059247] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 02/13/2013] [Indexed: 01/22/2023] Open
Abstract
During vertebrate eye development, the transcription factor MITF acts to promote the development of the retinal pigment epithelium (RPE). In embryos with Mitf mutations, the future RPE hyperproliferates and is respecified as retinal tissue but only in a small portion of the dorsal RPE. Using a series of genetic crosses, we show that this spatial restriction of RPE respecification is brought about by persistent expression of the anti-retinogenic ventral homeodomain gene Vax2 in the dorso-proximal and both Vax1 and Vax2 in the ventral RPE. We further show that dorso-proximal RPE respecification in Vax2/Mitf double mutants and dorso-proximal and ventral RPE respecification in Vax1/2/Mitf triple mutants result from increased FGF/MAP kinase signaling. In none of the mutants, however, does the distal RPE show signs of hyperproliferation or respecification, likely due to local JAGGED1/NOTCH signaling. Expression studies and optic vesicle culture experiments also suggest a role for NOTCH signaling within the mutant dorsal RPE domains, where ectopic JAGGED1 expression may partially counteract the effects of FGF/ERK1/2 signaling on RPE respecification. The results indicate the presence of complex interplays between distinct transcription factors and signaling molecules during eye development and show how RPE phenotypes associated with mutations in one gene are modulated by expression changes in other genes.
Collapse
Affiliation(s)
- Jingxing Ou
- Mammalian Development Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | |
Collapse
|
48
|
Effects of imposed defocus of opposite sign on temporal gene expression patterns of BMP4 and BMP7 in chick RPE. Exp Eye Res 2013; 109:98-106. [PMID: 23428741 DOI: 10.1016/j.exer.2013.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 01/15/2013] [Accepted: 02/05/2013] [Indexed: 12/29/2022]
Abstract
This study investigated the effects of imposed optical defocus on the expression patterns of bone morphogenetic protein 4 and 7 (BMP4, BMP7) in chick retinal pigment epithelium (RPE), as indicators of roles in postnatal eye growth regulation. BMP4 and BMP7 gene and protein expression patterns were characterized for retina, RPE and choroid tissues of young normal White-Leghorn chickens. The effects of short-term (2 and 48 h) exposure to monocular +10 and -10 diopter (D) lenses on RPE gene expression of BMP4 and BMP7 were also examined. Tissues from both treated and fellow eyes as well as from eyes of age-matched untreated birds were included in the latter experiment. Of ocular tissues comprising the posterior wall of the chick eye, RPE showed the highest expression of BMP4 and BMP7 mRNA, compared to retina and choroid. Western blots and immunohistochemistry confirmed the expression of BMP4 and BMP7 protein in all layers - retina, RPE, choroid and sclera. With imposed defocus, both BMP4 and BMP7 showed bidirectional changes in expression in RPE, however, with different temporal patterns. With +10 D lenses, BMP4 gene expression was up-regulated after both 2 and 48 h of treatment, while BMP7 expression was up-regulated only after 48 h of lens wear. With -10 D lenses, both BMP4 and BMP7 showed down-regulation of gene expression for both 2 and 48 h treatment durations. With the -10 D lens treatment applied for 48 h, gene expression for both BMP4 and BMP7 was also down-regulated in contralateral fellows of treated eyes compared to eyes of untreated chicks. The rapid changes in gene expression in chick RPE observed for both BMP4 and BMP7, up or down according to the sign of imposed optical defocus, resemble similar trends reported for BMP2. Further studies are needed to confirm the roles of BMPs as ocular growth modulators, as suggested by these data. The data also suggest a role for the RPE as a conduit for relaying growth modulatory retinal signals.
Collapse
|
49
|
Sasai Y, Eiraku M, Suga H. In vitro organogenesis in three dimensions: self-organising stem cells. Development 2013; 139:4111-21. [PMID: 23093423 DOI: 10.1242/dev.079590] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organ formation during embryogenesis is a complex process that involves various local cell-cell interactions at the molecular and mechanical levels. Despite this complexity, organogenesis can be modelled in vitro. In this article, we focus on two recent examples in which embryonic stem cells can self-organise into three-dimensional structures - the optic cup and the pituitary epithelium; and one case of self-organising adult stem cells - the gut epithelium. We summarise how these approaches have revealed intrinsic programs that drive locally autonomous modes of organogenesis and homeostasis. We also attempt to interpret the results of previous in vivo studies of retinal development in light of the self-organising nature of the retina.
Collapse
Affiliation(s)
- Yoshiki Sasai
- Neurogenesis and Organogenesis Group, RIKEN Center for Developmental Biology, Kobe, Japan.
| | | | | |
Collapse
|
50
|
Reh TA. The Development of the Retina. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|