1
|
Yin Z, Kang J, Cheng X, Gao H, Huo S, Xu H. Investigating Müller glia reprogramming in mice: a retrospective of the last decade, and a look to the future. Neural Regen Res 2025; 20:946-959. [PMID: 38989930 PMCID: PMC11438324 DOI: 10.4103/nrr.nrr-d-23-01612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
Müller glia, as prominent glial cells within the retina, plays a significant role in maintaining retinal homeostasis in both healthy and diseased states. In lower vertebrates like zebrafish, these cells assume responsibility for spontaneous retinal regeneration, wherein endogenous Müller glia undergo proliferation, transform into Müller glia-derived progenitor cells, and subsequently regenerate the entire retina with restored functionality. Conversely, Müller glia in the mouse and human retina exhibit limited neural reprogramming. Müller glia reprogramming is thus a promising strategy for treating neurodegenerative ocular disorders. Müller glia reprogramming in mice has been accomplished with remarkable success, through various technologies. Advancements in molecular, genetic, epigenetic, morphological, and physiological evaluations have made it easier to document and investigate the Müller glia programming process in mice. Nevertheless, there remain issues that hinder improving reprogramming efficiency and maturity. Thus, understanding the reprogramming mechanism is crucial toward exploring factors that will improve Müller glia reprogramming efficiency, and for developing novel Müller glia reprogramming strategies. This review describes recent progress in relatively successful Müller glia reprogramming strategies. It also provides a basis for developing new Müller glia reprogramming strategies in mice, including epigenetic remodeling, metabolic modulation, immune regulation, chemical small-molecules regulation, extracellular matrix remodeling, and cell-cell fusion, to achieve Müller glia reprogramming in mice.
Collapse
Affiliation(s)
- Zhiyuan Yin
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | | | | | | | | | | |
Collapse
|
2
|
Naef V, Damiani D, Licitra R, Marchese M, Vecchia SD, Baggiani M, Brogi L, Galatolo D, Landi S, Santorelli FM. Modeling sacsin depletion in Danio Rerio offers new insight on retinal defects in ARSACS. Neurobiol Dis 2025; 205:106793. [PMID: 39778749 PMCID: PMC11757156 DOI: 10.1016/j.nbd.2025.106793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/17/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025] Open
Abstract
Biallelic mutations in the SACS gene, encoding sacsin, cause early-onset autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS), a neurodegenerative disease also characterized by unique and poorly understood retinal abnormalities. While two murine models replicate the phenotypic and neuronal features observed in patients, no retinal phenotype has been described so far. In a zebrafish knock-out strain that faithfully mirrors the main aspects of ARSACS, we observed impaired visual function due to photoreceptor degeneration, likely caused by cell cycle defects in progenitor cells. RNA-seq analysis in embryos revealed dysfunction in proteins related to fat-soluble vitamins (e.g., TTPA, RDH5, VKORC) and suggested a key role of neuroinflammation in driving the retinal defects. Our findings indicate that studying retinal pathology in ARSACS could be crucial for understanding the impact of sacsin depletion and may offer insights into halting disease progression.
Collapse
Affiliation(s)
- Valentina Naef
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy.
| | - Devid Damiani
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Rosario Licitra
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Maria Marchese
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Stefania Della Vecchia
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Matteo Baggiani
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Letizia Brogi
- Bio@SNS, Department of Neurosciences, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Daniele Galatolo
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | | | | |
Collapse
|
3
|
Hernández-Núñez I, Clark BS. Experimental Framework for Assessing Mouse Retinal Regeneration Through Single-Cell RNA-Sequencing. Methods Mol Biol 2025; 2848:117-134. [PMID: 39240520 DOI: 10.1007/978-1-0716-4087-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Retinal degenerative diseases including age-related macular degeneration and glaucoma are estimated to currently affect more than 14 million people in the United States, with an increased prevalence of retinal degenerations in aged individuals. An expanding aged population who are living longer forecasts an increased prevalence and economic burden of visual impairments. Improvements to visual health and treatment paradigms for progressive retinal degenerations slow vision loss. However, current treatments fail to remedy the root cause of visual impairments caused by retinal degenerations-loss of retinal neurons. Stimulation of retinal regeneration from endogenous cellular sources presents an exciting treatment avenue for replacement of lost retinal cells. In multiple species including zebrafish and Xenopus, Müller glial cells maintain a highly efficient regenerative ability to reconstitute lost cells throughout the organism's lifespan, highlighting potential therapeutic avenues for stimulation of retinal regeneration in humans. Here, we describe how the application of single-cell RNA-sequencing (scRNA-seq) has enhanced our understanding of Müller glial cell-derived retinal regeneration, including the characterization of gene regulatory networks that facilitate/inhibit regenerative responses. Additionally, we provide a validated experimental framework for cellular preparation of mouse retinal cells as input into scRNA-seq experiments, including insights into experimental design and analyses of resulting data.
Collapse
Affiliation(s)
- Ismael Hernández-Núñez
- John F Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S Clark
- John F Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Yin Z, Kang J, Xu H, Huo S, Xu H. Recent progress of principal techniques used in the study of Müller glia reprogramming in mice. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:30. [PMID: 39663301 PMCID: PMC11635068 DOI: 10.1186/s13619-024-00211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
In zebrafish, Müller glia (MG) cells retain the ability to proliferate and de-differentiate into retinal progenitor-like cells, subsequently differentiating into retinal neurons that can replace those damaged or lost due to retinal injury. In contrast, the reprogramming potential of MG in mammals has been lost, with these cells typically responding to retinal damage through gliosis. Considerable efforts have been dedicated to achieving the reprogramming of MG cells in mammals. Notably, significant advancements have been achieved in reprogramming MG cells in mice employing various methodologies. At the same time, some inevitable challenges have hindered identifying accurate MG cell reprogramming rather than the illusion, let alone improving the reprogramming efficiency and maturity of daughter cells. Recently, several strategies, including lineage tracking, multi-omics techniques, and functional analysis, have been developed to investigate the MG reprogramming process in mice. This review summarizes both the advantages and limitations of these novel strategies for analyzing MG reprogramming in mice, offering insights into enhancing the reliability and efficiency of MG reprogramming.
Collapse
Affiliation(s)
- Zhiyuan Yin
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China
| | - Jiahui Kang
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China
| | - Haoan Xu
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shujia Huo
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China.
| | - Haiwei Xu
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China.
| |
Collapse
|
5
|
Hu H, Qin Y, Qu Z, Huang Y, Ren X, Liu M, Liu F, Gao P. The Zpr-3 Antibody Recognizes the 320-354 Region of Rho and Labels Both Rods and Green Cones in Zebrafish. Zebrafish 2024; 21:394-400. [PMID: 39316468 DOI: 10.1089/zeb.2024.0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
Retinal markers with high quality and specificity are important for the observation of pathologic changes of retinal cells during retinal development, degeneration, and regeneration. The zpr-3 antibody is widely used to label rods in zebrafish, but the exact antigen is still unknown. In this study, we provided evidence to demonstrate that the antigen gene of zpr-3 is rho, which encodes the rod opsin, and the exact epitope of zpr-3 is the 320-354 region of Rho protein. More importantly, our immunofluorescence assays indicated that zpr-3 labels both the outer segments of rods and green cones on zebrafish retinal sections, probably due to the cross-reaction with the green-cone opsin. Our work is valuable for the scientific community to interpret the experimental data involving the zpr-3 antibody.
Collapse
Affiliation(s)
- Hualei Hu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yayun Qin
- Medical Genetics Center, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Zhen Qu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwen Huang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Ren
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Liu
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| | - Pan Gao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
La Pietra A, Bianchi AR, Capriello T, Mobilio T, Guagliardi A, De Maio A, Ferrandino I. Regeneration of zebrafish retina following toxic injury. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 112:104582. [PMID: 39481820 DOI: 10.1016/j.etap.2024.104582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
The structure of the zebrafish retina appears to be very similar to that of mammals, that is why it is used as a model for studying the eye. Indeed, the zebrafish retina can regenerate itself through mechanisms of Müller cell reprogramming. In this research, adult zebrafish were exposed to aluminum to cause damage in the retina and thus evaluate the regenerative capacity of the damaged tissue. Histological and histochemical analyses assessed the retinal structure and the neurodegenerative process, respectively. An expression analysis of PARPs was carried out to verify whether a potential oxidative DNA damage happens. In addition, some genes involved in the regeneration process (pax6a, pax2a, ngn1, and notch1a) were analyzed. The data confirmed the toxicity of aluminum which caused retinal neurodegeneration, but also highlighted the ability of zebrafish to regenerate the retinal structure, repairing the damage and confirming its use as a good model for translational studies.
Collapse
Affiliation(s)
- Alessandra La Pietra
- Department of Biology, University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy.
| | - Anna Rita Bianchi
- Department of Biology, University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy.
| | - Teresa Capriello
- Department of Biology, University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy.
| | - Teresa Mobilio
- Department of Biology, University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy.
| | - Annamaria Guagliardi
- Department of Biology, University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy.
| | - Anna De Maio
- Department of Biology, University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy.
| | - Ida Ferrandino
- Department of Biology, University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy.
| |
Collapse
|
7
|
Volkov LI, Ogawa Y, Somjee R, Vedder HE, Powell HE, Poria D, Meiselman S, Kefalov VJ, Corbo JC. Samd7 represses short-wavelength cone genes to preserve long-wavelength cone and rod photoreceptor identity. Proc Natl Acad Sci U S A 2024; 121:e2402121121. [PMID: 39531499 PMCID: PMC11588049 DOI: 10.1073/pnas.2402121121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 09/16/2024] [Indexed: 11/16/2024] Open
Abstract
The role of transcription factors in photoreceptor gene regulation is fairly well understood, but knowledge of the cell-type-specific function of transcriptional cofactors remains incomplete. Here, we show that the transcriptional corepressor samd7 promotes rod differentiation and represses short-wavelength cone genes in long-wavelength cones in zebrafish. In samd7-/- retinas, red cones are transformed into hybrid red/ultraviolet (UV) cones, green cones are absent, the number of blue cones is approximately doubled, and the number of rods is greatly reduced. We also find that mouse Samd7 represses S-opsin expression in dorsal M-cones-analogous to its role in repressing UV cone genes in zebrafish red cones. Thus, samd7 plays a key role in ensuring appropriate patterns of gene expression in rods and cone subtypes of both zebrafish and mice.
Collapse
Affiliation(s)
- Leo I. Volkov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Yohey Ogawa
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Ramiz Somjee
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Hannah E. Vedder
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Hannah E. Powell
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Deepak Poria
- Department of Ophthalmology, University of California Irvine, Irvine, CA92697
| | - Sam Meiselman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Vladimir J. Kefalov
- Department of Ophthalmology, University of California Irvine, Irvine, CA92697
| | - Joseph C. Corbo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
8
|
Jui J, Goldman D. Müller Glial Cell-Dependent Regeneration of the Retina in Zebrafish and Mice. Annu Rev Genet 2024; 58:67-90. [PMID: 38876121 DOI: 10.1146/annurev-genet-111523-102000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Sight is one of our most precious senses. People fear losing their sight more than any other disability. Thus, restoring sight to the blind is an important goal of vision scientists. Proregenerative species, such as zebrafish, provide a system for studying endogenous mechanisms underlying retina regeneration. Nonregenerative species, such as mice, provide a system for testing strategies for stimulating retina regeneration. Key to retina regeneration in zebrafish and mice is the Müller glial cell, a malleable cell type that is amenable to a variety of regenerative strategies. Here, we review cellular and molecular mechanisms used by zebrafish to regenerate a retina, as well as the application of these mechanisms, and other strategies to stimulate retina regeneration in mice. Although our focus is on Müller glia (MG), niche components and their impact on MG reprogramming are also discussed.
Collapse
Affiliation(s)
- Jonathan Jui
- Molecular Neuroscience Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA; ,
| | - Daniel Goldman
- Molecular Neuroscience Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA; ,
| |
Collapse
|
9
|
Mikula Mrstakova S, Kozmik Z. Genetic analysis of medaka fish illuminates conserved and divergent roles of Pax6 in vertebrate eye development. Front Cell Dev Biol 2024; 12:1448773. [PMID: 39512904 PMCID: PMC11541176 DOI: 10.3389/fcell.2024.1448773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024] Open
Abstract
Landmark discovery of eye defects caused by Pax6 gene mutations in humans, rodents, and even fruit flies combined with Pax6 gene expression studies in various phyla, led to the master control gene hypothesis postulating that the gene is required almost universally for animal visual system development. However, this assumption has not been broadly tested in genetically trackable organisms such as vertebrates. Here, to determine the functional role of the fish orthologue of mammalian Pax6 in eye development we analyzed mutants in medaka Pax6.1 gene generated by genome editing. We found that transcription factors implicated in vertebrate lens development (Prox1a, MafB, c-Maf, FoxE3) failed to initiate expression in the presumptive lens tissue of Pax6.1 mutant fish resulting in aphakia, a phenotype observed previously in Pax6 mutant mice. Surprisingly, the overall differentiation potential of Pax6.1-deficient retinal progenitor cells (RPCs) is not severely compromised, and the only cell types affected by the absence of Pax6.1 transcription factor are retinal ganglion cells. This is in stark contrast to the situation in mice where the Pax6 gene is required cell-autonomously for the expansion of RPCs, and the differentiation of all retina cell types. Our results provide novel insight into the conserved and divergent roles of Pax6 gene orthologues in vertebrate eye development indicating that the lens-specific role is more evolutionarily conserved than the role in retina differentiation.
Collapse
Affiliation(s)
| | - Zbynek Kozmik
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
10
|
Prieto-López L, Pereiro X, Vecino E. The mechanics of the retina: Müller glia role on retinal extracellular matrix and modelling. Front Med (Lausanne) 2024; 11:1393057. [PMID: 39296899 PMCID: PMC11410058 DOI: 10.3389/fmed.2024.1393057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
The retina is a highly heterogeneous tissue, both cell-wise but also regarding its extracellular matrix (ECM). The stiffness of the ECM is pivotal in retinal development and maturation and has also been associated with the onset and/or progression of numerous retinal pathologies, such as glaucoma, proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), epiretinal membrane (ERM) formation or uveitis. Nonetheless, much remains unknown about the biomechanical milieu of the retina, and specifically the role that Müller glia play as principal mechanosensors and major producers of ECM constituents. So far, new approaches need to be developed to further the knowledge in the field of retinal mechanobiology for ECM-target applications to arise. In this review, we focus on the involvement of Müller glia in shaping and altering the retinal ECM under both physiological and pathological conditions and look into various biomaterial options to more accurately replicate the impact of matrix stiffness in vitro.
Collapse
Affiliation(s)
- Laura Prieto-López
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| |
Collapse
|
11
|
Williams AL, Bohnsack BL. Keratin 8/18a.1 Expression Influences Embryonic Neural Crest Cell Dynamics and Contributes to Postnatal Corneal Regeneration in Zebrafish. Cells 2024; 13:1473. [PMID: 39273043 PMCID: PMC11394277 DOI: 10.3390/cells13171473] [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: 12/26/2023] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
A complete understanding of neural crest cell mechanodynamics during ocular development will provide insight into postnatal neural crest cell contributions to ophthalmic abnormalities in adult tissues and inform regenerative strategies toward injury repair. Herein, single-cell RNA sequencing in zebrafish during early eye development revealed keratin intermediate filament genes krt8 and krt18a.1 as additional factors expressed during anterior segment development. In situ hybridization and immunofluorescence microscopy confirmed krt8 and krt18a.1 expression in the early neural plate border and migrating cranial neural crest cells. Morpholino oligonucleotide (MO)-mediated knockdown of K8 and K18a.1 markedly disrupted the migration of neural crest cell subpopulations and decreased neural crest cell marker gene expression in the craniofacial region and eye at 48 h postfertilization (hpf), resulting in severe phenotypic defects reminiscent of neurocristopathies. Interestingly, the expression of K18a.1, but not K8, is regulated by retinoic acid (RA) during early-stage development. Further, both keratin proteins were detected during postnatal corneal regeneration in adult zebrafish. Altogether, we demonstrated that both K8 and K18a.1 contribute to the early development and postnatal repair of neural crest cell-derived ocular tissues.
Collapse
Affiliation(s)
- Antionette L. Williams
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave., Chicago, IL 60611, USA;
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, 645 N. Michigan Ave., Chicago, IL 60611, USA
| | - Brenda L. Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave., Chicago, IL 60611, USA;
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, 645 N. Michigan Ave., Chicago, IL 60611, USA
| |
Collapse
|
12
|
Blackshaw S, Qian J, Hyde DR. New pathways to neurogenesis: Insights from injury-induced retinal regeneration. Bioessays 2024; 46:e2400133. [PMID: 38990084 DOI: 10.1002/bies.202400133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
The vertebrate retina is a tractable system for studying control of cell neurogenesis and cell fate specification. During embryonic development, retinal neurogenesis is under strict temporal regulation, with cell types generated in fixed but overlapping temporal intervals. The temporal sequence and relative numbers of retinal cell types generated during development are robust and show minimal experience-dependent variation. In many cold-blooded vertebrates, acute retinal injury induces a different form of neurogenesis, where Müller glia reprogram into retinal progenitor-like cells that selectively regenerate retinal neurons lost to injury. The extent to which the molecular mechanisms controlling developmental and injury-induced neurogenesis resemble one another has long been unclear. However, a recent study in zebrafish has shed new light on this question, using single-cell multiomic analysis to show that selective loss of different retinal cell types induces the formation of fate-restricted Müller glia-derived progenitors that differ both from one another and from progenitors in developing retina. Here, we discuss the broader implications of these findings, and their possible therapeutic relevance.
Collapse
Affiliation(s)
- Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David R Hyde
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, USA
- Center for Zebrafish Research, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
13
|
Abraham E, Hartmann H, Yoshimatsu T, Baden T, Brand M. Restoration of cone-circuit functionality in the regenerating adult zebrafish retina. Dev Cell 2024; 59:2158-2170.e6. [PMID: 39096897 DOI: 10.1016/j.devcel.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/28/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024]
Abstract
Unlike humans, teleosts like zebrafish exhibit robust retinal regeneration after injury from endogenous stem cells. However, it is unclear if regenerating cone photoreceptors regain physiological function and integrate correctly into post-synaptic circuits. We used two-photon calcium imaging of living adult retina to examine photoreceptor responses before and after light-induced lesions. To assess functional recovery of cones and downstream outer retinal circuits, we exploited color opponency; UV cones exhibit intrinsic Off-response to blue light, but On-response to green light, which depends on feedback signals from outer retinal circuits. Accordingly, we assessed the presence and quality of Off- vs. On-responses and found that regenerated UV cones regain both Off-responses to short-wavelength and On-responses to long-wavelength light within 3 months after lesion. Therefore, physiological circuit functionality is restored in regenerated cone photoreceptors, suggesting that inducing endogenous regeneration is a promising strategy for human retinal repair.
Collapse
Affiliation(s)
- Evelyn Abraham
- CRTD - Center for Regenerative Therapies TU Dresden, CMCB, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Hella Hartmann
- CMCB - Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Takeshi Yoshimatsu
- School of Life Sciences, University of Sussex, Biology Road, Brighton BN1 9QG, UK
| | - Tom Baden
- School of Life Sciences, University of Sussex, Biology Road, Brighton BN1 9QG, UK
| | - Michael Brand
- CRTD - Center for Regenerative Therapies TU Dresden, CMCB, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; CMCB - Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany; PoL - Excellence Cluster Physics of Life, TU Dresden, Fetscherstrasse 105, 01307 Dresden, Germany.
| |
Collapse
|
14
|
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
|
15
|
Lee MS, Jui J, Sahu A, Goldman D. Mycb and Mych stimulate Müller glial cell reprogramming and proliferation in the uninjured and injured zebrafish retina. Development 2024; 151:dev203062. [PMID: 38984586 PMCID: PMC11369687 DOI: 10.1242/dev.203062] [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: 05/17/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
In the injured zebrafish retina, Müller glial cells (MG) reprogram to adopt retinal stem cell properties and regenerate damaged neurons. The strongest zebrafish reprogramming factors might be good candidates for stimulating a similar regenerative response by mammalian MG. Myc proteins are potent reprogramming factors that can stimulate cellular plasticity in differentiated cells; however, their role in MG reprogramming and retina regeneration remains poorly explored. Here, we report that retinal injury stimulates mycb and mych expression and that, although both Mycb and Mych stimulate MG reprogramming and proliferation, only Mych enhances retinal neuron apoptosis. RNA-sequencing analysis of wild-type, mychmut and mycbmut fish revealed that Mycb and Mych regulate ∼40% and ∼16%, respectively, of the genes contributing to the regeneration-associated transcriptome of MG. Of these genes, those that are induced are biased towards regulation of ribosome biogenesis, protein synthesis, DNA synthesis, and cell division, which are the top cellular processes affected by retinal injury, suggesting that Mycb and Mych are potent MG reprogramming factors. Consistent with this, forced expression of either of these proteins is sufficient to stimulate MG proliferation in the uninjured retina.
Collapse
Affiliation(s)
- Mi-Sun Lee
- Michigan Neuroscience Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan Jui
- Michigan Neuroscience Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aresh Sahu
- Michigan Neuroscience Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Goldman
- Michigan Neuroscience Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Le N, Vu TD, Palazzo I, Pulya R, Kim Y, Blackshaw S, Hoang T. Robust reprogramming of glia into neurons by inhibition of Notch signaling and nuclear factor I (NFI) factors in adult mammalian retina. SCIENCE ADVANCES 2024; 10:eadn2091. [PMID: 38996013 PMCID: PMC11244444 DOI: 10.1126/sciadv.adn2091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 06/10/2024] [Indexed: 07/14/2024]
Abstract
Generation of neurons through direct reprogramming has emerged as a promising therapeutic approach for treating neurodegenerative diseases. In this study, we present an efficient method for reprogramming retinal glial cells into neurons. By suppressing Notch signaling by disrupting either Rbpj or Notch1/2, we induced mature Müller glial cells to reprogram into bipolar- and amacrine-like neurons. We demonstrate that Rbpj directly activates both Notch effector genes and genes specific to mature Müller glia while indirectly repressing expression of neurogenic basic helix-loop-helix (bHLH) factors. Combined loss of function of Rbpj and Nfia/b/x resulted in conversion of nearly all Müller glia to neurons. Last, inducing Müller glial proliferation by overexpression of dominant-active Yap promotes neurogenesis in both Rbpj- and Nfia/b/x/Rbpj-deficient Müller glia. These findings demonstrate that Notch signaling and NFI factors act in parallel to inhibit neurogenic competence in mammalian Müller glia and help clarify potential strategies for regenerative therapies aimed at treating retinal dystrophies.
Collapse
Affiliation(s)
- Nguyet Le
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Trieu-Duc Vu
- Department of Ophthalmology and Visual Sciences, University of Michigan School of Medicine, Ann Arbor, MI 48105
- Michigan Neuroscience Institute, University of Michigan School of Medicine, Ann Arbor, MI 48105, USA
| | - Isabella Palazzo
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ritvik Pulya
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yehna Kim
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thanh Hoang
- Department of Ophthalmology and Visual Sciences, University of Michigan School of Medicine, Ann Arbor, MI 48105
- Michigan Neuroscience Institute, University of Michigan School of Medicine, Ann Arbor, MI 48105, USA
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48105, USA
| |
Collapse
|
17
|
Taylor OB, Patel SP, Hawthorn EC, El-Hodiri HM, Fischer AJ. ID factors regulate the ability of Müller glia to become proliferating neurogenic progenitor-like cells. Glia 2024; 72:1236-1258. [PMID: 38515287 PMCID: PMC11334223 DOI: 10.1002/glia.24523] [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: 09/29/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
The purpose of this study was to investigate how ID factors regulate the ability of Müller glia (MG) to reprogram into proliferating MG-derived progenitor cells (MGPCs) in the chick retina. We found that ID1 is transiently expressed by maturing MG (mMG), whereas ID4 is maintained in mMG in embryonic retinas. In mature retinas, ID4 was prominently expressed by resting MG, but following retinal damage ID4 was rapidly upregulated and then downregulated in MGPCs. By contrast, ID1, ID2, and ID3 were low in resting MG and then upregulated in MGPCs. Inhibition of ID factors following retinal damage decreased numbers of proliferating MGPCs. Inhibition of IDs, after MGPC proliferation, significantly increased numbers of progeny that differentiated as neurons. In damaged or undamaged retinas inhibition of IDs increased levels of p21Cip1 in MG. In response to damage or insulin+FGF2 levels of CDKN1A message and p21Cip1 protein were decreased, absent in proliferating MGPCs, and elevated in MG returning to a resting phenotype. Inhibition of notch- or gp130/Jak/Stat-signaling in damaged retinas increased levels of ID4 but not p21Cip1 in MG. Although ID4 is the predominant isoform expressed by MG in the chick retina, id1 and id2a are predominantly expressed by resting MG and downregulated in activated MG and MGPCs in zebrafish retinas. We conclude that ID factors have a significant impact on regulating the responses of MG to retinal damage, controlling the ability of MG to proliferate by regulating levels of p21Cip1, and suppressing the neurogenic potential of MGPCs.
Collapse
Affiliation(s)
- Olivia B. Taylor
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Snehal P. Patel
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Evan C. Hawthorn
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Heithem M. El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
18
|
Kelly LE, El-Hodiri HM, Crider A, Fischer AJ. Protein phosphatases regulate the formation of Müller glia-derived progenitor cells in the chick retina. Mol Cell Neurosci 2024; 129:103932. [PMID: 38679247 PMCID: PMC11362962 DOI: 10.1016/j.mcn.2024.103932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024] Open
Abstract
Different kinase-dependent cell signaling pathways are known to play important roles in glia-mediated neuroprotection and reprogramming of Müller glia (MG) into Müller glia-derived progenitor cells (MGPCs) in the retina. However, very little is known about the phosphatases that regulate kinase-dependent signaling in MG. Using single-cell RNA-sequencing (scRNA-seq) databases, we investigated patterns of expression of Dual Specificity Phosphatases (DUSP1/6) and other protein phosphatases in normal and damaged chick retinas. We found that DUSP1, DUSP6, PPP3CB, PPP3R1 and PPPM1A/B/D/E/G are widely expressed by many types of retinal neurons and are dynamically expressed by MG and MGPCs in retinas during the process of reprogramming. We find that inhibition of DUSP1/6 and PP2C phosphatases enhances the formation of proliferating MGPCs in damaged retinas and in retinas treated with insulin and FGF2 in the absence of damage. By contrast, inhibition of PP2B phosphatases suppressed the formation of proliferating MGPCs, but increased numbers of proliferating MGPCs in undamaged retinas treated with insulin and FGF2. In damaged retinas, inhibition of DUSP1/6 increased levels of pERK1/2 and cFos in MG whereas inhibition of PP2B's decreased levels of pStat3 and pS6 in MG. Analyses of scRNA-seq libraries identified numerous differentially activated gene modules in MG in damaged retinas versus MG in retinas treated with insulin+FGF2 suggesting significant differences in kinase-dependent signaling pathways that converge on the formation of MGPCs. Inhibition of phosphatases had no significant effects upon numbers of dying cells in damaged retinas. We conclude that the activity of different protein phosphatases acting through retinal neurons and MG "fine-tune" the cell signaling responses of MG in damaged retinas and during the reprogramming of MG into MGPCs.
Collapse
Affiliation(s)
- Lisa E Kelly
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Heithem M El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Andrew Crider
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
19
|
Han S, Hu Y, Jia D, Lv Y, Liu M, Wang D, Chao J, Xia X, Wang Q, Liu P, Cai Y, Ren X. IFT27 regulates the long-term maintenance of photoreceptor outer segments in zebrafish. Gene 2024; 905:148237. [PMID: 38310983 DOI: 10.1016/j.gene.2024.148237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Approximately a quarter of Retinitis Pigmentosa (RP) is caused by mutations in transport-related genes in cilia. IFT27 (Intraflagellar Transport 27), a core component of the ciliary intraflagellar transport (IFT) system, has been implicated as a significant pathogenic gene in RP. The pathogenic mechanisms and subsequent pathology related to IFT27 mutations in RP are largely obscure. Here, we utilized TALEN technology to create an ift27 knockout (ift27-/-) zebrafish model. Electroretinography (ERG) detection showed impaired vision in this model. Histopathological examinations disclosed that ift27 mutations cause progressive degeneration of photoreceptors in zebrafish, and this degeneration was late-onset. Immunofluorescence labeling of outer segments showed that rods degenerated before cones, aligning with the conventional characterization of RP. In cultured human retinal pigment epithelial cells, we found that IFT27 was involved in maintaining ciliary morphology. Furthermore, decreased IFT27 expression resulted in the inhibition of the Hedgehog (Hh) signaling pathway, including decreased expression of key factors in the Hh pathway and abnormal localization of the ciliary mediator Gli2. In summary, we generated an ift27-/- zebrafish line with retinal degeneration which mimicked the symptoms of RP patients, highlighting IFT27's integral role in the long-term maintenance of cilia via the Hh signaling pathway. This work may furnish new insights into the treatment or delay of RP caused by IFT27 mutations.
Collapse
Affiliation(s)
- Shanshan Han
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China.
| | - Yue Hu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Danna Jia
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Yuexia Lv
- Prenatal Diagnosis Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Decheng Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Jin Chao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Xuan Xia
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Qiong Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Pei Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Yu Cai
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Xiang Ren
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
20
|
Konar GJ, Flickinger Z, Sharma S, Vallone KT, Lyon CE, Doshier C, Lingan A, Lyon W, Patton JG. Damage-Induced Senescent Immune Cells Regulate Regeneration of the Zebrafish Retina. AGING BIOLOGY 2024; 2:e20240021. [PMID: 39156966 PMCID: PMC11328971 DOI: 10.59368/agingbio.20240021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Zebrafish spontaneously regenerate their retinas in response to damage through the action of Müller glia (MG). Even though MG are conserved in higher vertebrates, the capacity to regenerate retinal damage is lost. Recent work has focused on the regulation of inflammation during tissue regeneration, with temporal roles for macrophages and microglia. Senescent cells that have withdrawn from the cell cycle have mostly been implicated in aging but are still metabolically active, releasing a variety of signaling molecules as part of the senescence-associated secretory phenotype. Here, we discover that in response to retinal damage, a subset of cells expressing markers of microglia/macrophages also express markers of senescence. These cells display a temporal pattern of appearance and clearance during retina regeneration. Premature removal of senescent cells by senolytic treatment led to a decrease in proliferation and incomplete repair of the ganglion cell layer after N-methyl-D-aspartate damage. Our results demonstrate a role for modulation of senescent cell responses to balance inflammation, regeneration, plasticity, and repair as opposed to fibrosis and scarring.
Collapse
Affiliation(s)
- Gregory J. Konar
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Zachary Flickinger
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Shivani Sharma
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Kyle T. Vallone
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Charles E. Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Claire Doshier
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Audrey Lingan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - William Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - James G. Patton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
21
|
Pavlou M, Probst M, Blasdel N, Prieve AR, Reh TA. The impact of timing and injury mode on induced neurogenesis in the adult mammalian retina. Stem Cell Reports 2024; 19:239-253. [PMID: 38278154 PMCID: PMC10874861 DOI: 10.1016/j.stemcr.2023.12.010] [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: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024] Open
Abstract
Regeneration of neurons has important implications for human health, and the retina provides an accessible system to study the potential of replacing neurons following injury. In previous work, we generated transgenic mice in which neurogenic transcription factors were expressed in Müller glia (MG) and showed that they stimulated neurogenesis following inner retinal damage. It was unknown, however, whether the timing or mode of injury mattered in this process. Here, we explored these parameters on induced neurogenesis from MG and show that MG expressing Ascl1 will generate new bipolar neurons with similar efficiency irrespective of injury mode or timing. However, MG that express Ascl1-Atoh1 produce a new type of retinal ganglion-like cell after outer retinal damage, which is absent with inner retinal damage. Our data suggest that although cell fate is primarily dictated by neurogenic transcription factors, the inflammatory state of MG relative to injury can influence the outcome of induced neurogenesis.
Collapse
Affiliation(s)
- Marina Pavlou
- Department of Biological Structure, University of Washington, Seattle, WA, USA; Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Marlene Probst
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Nicolai Blasdel
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Aric R Prieve
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA; Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
22
|
Kondoh H. Organ Regeneration Without Relying on Regeneration-Dedicated Stem Cells. Results Probl Cell Differ 2024; 72:105-118. [PMID: 38509254 DOI: 10.1007/978-3-031-39027-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
The classic conception of tissue regeneration assumed the existence of tissue-proper regeneration stem cells that are set aside during normal tissue development and reserved as stem cells for regeneration. However, modern studies using cell tracing and other approaches have ruled out the presence of regeneration-proper stem cells in most cases in vertebrate tissue regeneration. The only experimentally validated regeneration-dedicated reserve cells are the satellite cells in skeletal muscle (e.g., Michele 2022) (see Sect. 5.2.3 ). Here, we will first discuss examples of large-scale tissue regeneration, liver regeneration in mammals, and lens and limb regeneration in newts. Then, attempts to widen the tissue regeneration capacity in mammals with exogenous transcription factor genes will be reviewed.
Collapse
Affiliation(s)
- Hisato Kondoh
- Osaka University, Suita, Osaka, Japan
- Biohistory Research Hall, Takatsuki, Osaka, Japan
| |
Collapse
|
23
|
Guo YM, Jiang X, Min J, Huang J, Huang XF, Ye L. Advances in the study of Müller glia reprogramming in mammals. Front Cell Neurosci 2023; 17:1305896. [PMID: 38155865 PMCID: PMC10752929 DOI: 10.3389/fncel.2023.1305896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
Müller cells play an integral role in the development, maintenance, and photopic signal transmission of the retina. While lower vertebrate Müller cells can differentiate into various types of retinal neurons to support retinal repair following damage, there is limited neurogenic potential of mammalian Müller cells. Therefore, it is of great interest to harness the neurogenic potential of mammalian Müller cells to achieve self-repair of the retina. While multiple studies have endeavored to induce neuronal differentiation and proliferation of mammalian Müller cells under defined conditions, the efficiency and feasibility of these methods often fall short, rendering them inadequate for the requisites of retinal repair. As the mechanisms and methodologies of Müller cell reprogramming have been extensively explored, a summary of the reprogramming process of unlocking the neurogenic potential of Müller cells can provide insight into Müller cell fate development and facilitate their therapeutic use in retinal repair. In this review, we comprehensively summarize the progress in reprogramming mammalian Müller cells and discuss strategies for optimizing methods and enhancing efficiency based on the mechanisms of fate regulation.
Collapse
Affiliation(s)
- Yi-Ming Guo
- Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Xinyi Jiang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Min
- Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Juan Huang
- Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lu Ye
- Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, China
| |
Collapse
|
24
|
Kelly LE, El-Hodiri HM, Crider A, Fischer AJ. Protein phosphatases regulate the formation of Müller glia-derived progenitor cells in the chick retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.570629. [PMID: 38168320 PMCID: PMC10760049 DOI: 10.1101/2023.12.11.570629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Different kinase-dependent cell signaling pathways are known to play important roles in glia-mediated neuroprotection and reprogramming of Müller glia (MG) into Müller glia-derived progenitor cells (MGPCs) in the retina. However, very little is known about the phosphatases that regulate kinase-dependent signaling in MG. Using single-cell RNA-sequencing (scRNA-seq) databases, we investigated patterns of expression of Dual Specificity Phosphatases (DUSP1/6) and other protein phosphatases in normal and damaged chick retinas. We found that DUSP1, DUSP6, PPP3CB, PPP3R1 and PPPM1A/B/D/E/G are dynamically expressed by MG and MGPCs in retinas during the process of reprogramming. We find that inhibition of DUSP1/6 and PP2C phosphatases enhances the formation of proliferating MGPCs in damaged retinas and in retinas treated with insulin in FGF2 in the absence of damage. By contrast, inhibition of PP2B phosphatases suppressed the formation of proliferating MGPCs, but increased numbers of proliferating MGPCs in undamaged retinas treated with insulin and FGF2. In damaged retinas, inhibition of DUSP1/6 increased levels of pERK1/2 and cFos in MG whereas inhibition of PP2B's decreased levels of pStat3 and pS6 in MG. Analyses of scRNA-seq libraries identified numerous differentially activated gene modules in MG in damaged retinas versus MG in retinas treated with insulin+FGF2 suggesting significant differences in kinase-dependent signaling pathways that converge on the formation of MGPCs. Inhibition of phosphatases had no significant effects upon numbers of dying cells in damaged retinas. We conclude that the activity of different protein phosphatases "fine-tune" the cell signaling responses of MG in damaged retinas and during the reprogramming of MG into MGPCs.
Collapse
Affiliation(s)
- Lisa E. Kelly
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Heithem M. El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Andrew Crider
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
25
|
Wohlschlegel J, Finkbeiner C, Hoffer D, Kierney F, Prieve A, Murry AD, Haugan AK, Ortuño-Lizarán I, Rieke F, Golden SA, Reh TA. ASCL1 induces neurogenesis in human Müller glia. Stem Cell Reports 2023; 18:2400-2417. [PMID: 38039971 PMCID: PMC10724232 DOI: 10.1016/j.stemcr.2023.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 12/03/2023] Open
Abstract
In mammals, loss of retinal cells due to disease or trauma is an irreversible process that can lead to blindness. Interestingly, regeneration of retinal neurons is a well established process in some non-mammalian vertebrates and is driven by the Müller glia (MG), which are able to re-enter the cell cycle and reprogram into neurogenic progenitors upon retinal injury or disease. Progress has been made to restore this mechanism in mammals to promote retinal regeneration: MG can be stimulated to generate new neurons in vivo in the adult mouse retina after the over-expression of the pro-neural transcription factor Ascl1. In this study, we applied the same strategy to reprogram human MG derived from fetal retina and retinal organoids into neurons. Combining single cell RNA sequencing, single cell ATAC sequencing, immunofluorescence, and electrophysiology we demonstrate that human MG can be reprogrammed into neurogenic cells in vitro.
Collapse
Affiliation(s)
| | - Connor Finkbeiner
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Dawn Hoffer
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Faith Kierney
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Aric Prieve
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Alexandria D Murry
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Alexandra K Haugan
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | | | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Sam A Golden
- Department of Biological Structure, University of Washington, Seattle, WA, USA; Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA; Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
26
|
El-Hodiri HM, Bentley JR, Reske AG, Taylor OB, Palazzo I, Campbell WA, Halloy NR, Fischer AJ. Heparin-binding epidermal growth factor and fibroblast growth factor 2 rescue Müller glia-derived progenitor cell formation in microglia- and macrophage-ablated chick retinas. Development 2023; 150:dev202070. [PMID: 37971210 PMCID: PMC10730090 DOI: 10.1242/dev.202070] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Recent studies have demonstrated the impact of pro-inflammatory signaling and reactive microglia/macrophages on the formation of Müller glial-derived progenitor cells (MGPCs) in the retina. In chick retina, ablation of microglia/macrophages prevents the formation of MGPCs. Analyses of single-cell RNA-sequencing chick retinal libraries revealed that quiescent and activated microglia/macrophages have a significant impact upon the transcriptomic profile of Müller glia (MG). In damaged monocyte-depleted retinas, MG fail to upregulate genes related to different cell signaling pathways, including those related to Wnt, heparin-binding epidermal growth factor (HBEGF), fibroblast growth factor (FGF) and retinoic acid receptors. Inhibition of GSK3β, to simulate Wnt signaling, failed to rescue the deficit in MGPC formation, whereas application of HBEGF or FGF2 completely rescued the formation of MGPCs in monocyte-depleted retinas. Inhibition of Smad3 or activation of retinoic acid receptors partially rescued the formation of MGPCs in monocyte-depleted retinas. We conclude that signals produced by reactive microglia/macrophages in damaged retinas stimulate MG to upregulate cell signaling through HBEGF, FGF and retinoic acid, and downregulate signaling through TGFβ/Smad3 to promote the reprogramming of MG into proliferating MGPCs.
Collapse
Affiliation(s)
- Heithem M. El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43221, USA
| | - James R. Bentley
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43221, USA
| | - Alana G. Reske
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43221, USA
| | - Olivia B. Taylor
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43221, USA
| | - Isabella Palazzo
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Warren A. Campbell
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43221, USA
| | - Nicklaus R. Halloy
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43221, USA
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43221, USA
| |
Collapse
|
27
|
Celotto L, Rost F, Machate A, Bläsche J, Dahl A, Weber A, Hans S, Brand M. Single-cell RNA sequencing unravels the transcriptional network underlying zebrafish retina regeneration. eLife 2023; 12:RP86507. [PMID: 37988404 PMCID: PMC10662954 DOI: 10.7554/elife.86507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
In the lesioned zebrafish retina, Müller glia produce multipotent retinal progenitors that generate all retinal neurons, replacing lost cell types. To study the molecular mechanisms linking Müller glia reactivity to progenitor production and neuronal differentiation, we used single-cell RNA sequencing of Müller glia, progenitors and regenerated progeny from uninjured and light-lesioned retinae. We discover an injury-induced Müller glia differentiation trajectory that leads into a cell population with a hybrid identity expressing marker genes of Müller glia and progenitors. A glial self-renewal and a neurogenic trajectory depart from the hybrid cell population. We further observe that neurogenic progenitors progressively differentiate to generate retinal ganglion cells first and bipolar cells last, similar to the events observed during retinal development. Our work provides a comprehensive description of Müller glia and progenitor transcriptional changes and fate decisions in the regenerating retina, which are key to tailor cell differentiation and replacement therapies for retinal dystrophies in humans.
Collapse
Affiliation(s)
- Laura Celotto
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| | - Fabian Rost
- Technische Universität Dresden, DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| | - Anja Machate
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| | - Juliane Bläsche
- Technische Universität Dresden, DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| | - Andreas Dahl
- Technische Universität Dresden, DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| | - Anke Weber
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| | - Stefan Hans
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| | - Michael Brand
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), FetscherstraßeDresdenGermany
| |
Collapse
|
28
|
Le N, Vu TD, Palazzo I, Pulya R, Kim Y, Blackshaw S, Hoang T. Robust reprogramming of glia into neurons by inhibition of Notch signaling and NFI factors in adult mammalian retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.29.560483. [PMID: 37961663 PMCID: PMC10634926 DOI: 10.1101/2023.10.29.560483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Generation of neurons through direct reprogramming has emerged as a promising therapeutic approach for neurodegenerative diseases. Despite successful applications in vitro , in vivo implementation has been hampered by low efficiency. In this study, we present a highly efficient strategy for reprogramming retinal glial cells into neurons by simultaneously inhibiting key negative regulators. By suppressing Notch signaling through the removal of its central mediator Rbpj, we induced mature Müller glial cells to reprogram into bipolar and amacrine neurons in uninjured adult mouse retinas, and observed that this effect was further enhanced by retinal injury. We found that specific loss of function of Notch1 and Notch2 receptors in Müller glia mimicked the effect of Rbpj deletion on Müller glia-derived neurogenesis. Integrated analysis of multiome (scRNA- and scATAC-seq) and CUT&Tag data revealed that Rbpj directly activates Notch effector genes and genes specific to mature Müller glia while also indirectly represses the expression of neurogenic bHLH factors. Furthermore, we found that combined loss of function of Rbpj and Nfia/b/x resulted in a robust conversion of nearly all Müller glia to neurons. Finally, we demonstrated that inducing Müller glial proliferation by AAV (adeno-associated virus)-mediated overexpression of dominant- active Yap supports efficient levels of Müller glia-derived neurogenesis in both Rbpj - and Nfia/b/x/Rbpj - deficient Müller glia. These findings demonstrate that, much like in zebrafish, Notch signaling actively represses neurogenic competence in mammalian Müller glia, and suggest that inhibition of Notch signaling and Nfia/b/x in combination with overexpression of activated Yap could serve as an effective component of regenerative therapies for degenerative retinal diseases.
Collapse
|
29
|
Markitantova YV, Grigoryan EN. Cellular and Molecular Triggers of Retinal Regeneration in Amphibians. Life (Basel) 2023; 13:1981. [PMID: 37895363 PMCID: PMC10608152 DOI: 10.3390/life13101981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Understanding the mechanisms triggering the initiation of retinal regeneration in amphibians may advance the quest for prevention and treatment options for degenerating human retina diseases. Natural retinal regeneration in amphibians requires two cell sources, namely retinal pigment epithelium (RPE) and ciliary marginal zone. The disruption of RPE interaction with photoreceptors through surgery or injury triggers local and systemic responses for retinal protection. In mammals, disease-induced damage to the retina results in the shutdown of the function, cellular or oxidative stress, pronounced immune response, cell death and retinal degeneration. In contrast to retinal pathology in mammals, regenerative responses in amphibians have taxon-specific features ensuring efficient regeneration. These include rapid hemostasis, the recruitment of cells and factors of endogenous defense systems, activities of the immature immune system, high cell viability, and the efficiency of the extracellular matrix, cytoskeleton, and cell surface remodeling. These reactions are controlled by specific signaling pathways, transcription factors, and the epigenome, which are insufficiently studied. This review provides a summary of the mechanisms initiating retinal regeneration in amphibians and reveals its features collectively directed at recruiting universal responses to trauma to activate the cell sources of retinal regeneration. This study of the integrated molecular network of these processes is a prospect for future research in demand biomedicine.
Collapse
Affiliation(s)
| | - Eleonora N. Grigoryan
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| |
Collapse
|
30
|
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
|
31
|
Zhang H, Guo Y, Yang Y, Wang Y, Zhang Y, Zhuang J, Zhang Y, Shen M, Zhao J, Zhang R, Qiu Y, Li S, Hu J, Li W, Wu J, Xu H, Fliesler SJ, Liao Y, Liu Z. MAP4Ks inhibition promotes retinal neuron regeneration from Müller glia in adult mice. NPJ Regen Med 2023; 8:36. [PMID: 37443319 DOI: 10.1038/s41536-023-00310-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Mammalian Müller glia (MG) possess limited regenerative capacities. However, the intrinsic capacity of mammalian MG to transdifferentiate to generate mature neurons without transgenic manipulations remains speculative. Here we show that MAP4K4, MAP4K6 and MAP4K7, which are conserved Misshapen subfamily of ste20 kinases homologs, repress YAP activity in mammalian MG and therefore restrict their ability to be reprogrammed. However, by treating with a small molecule inhibitor of MAP4K4/6/7, mouse MG regain their ability to proliferate and enter into a retinal progenitor cell (RPC)-like state after NMDA-induced retinal damage; such plasticity was lost in YAP knockout MG. Moreover, spontaneous trans-differentiation of MG into retinal neurons expressing both amacrine and retinal ganglion cell (RGC) markers occurs after inhibitor withdrawal. Taken together, these findings suggest that MAP4Ks block the reprogramming capacity of MG in a YAP-dependent manner in adult mammals, which provides a novel avenue for the pharmaceutical induction of retinal regeneration in vivo.
Collapse
Affiliation(s)
- Houjian Zhang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
- Xiamen University Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China
- Department of Ophthalmology, the First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Yuli Guo
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
- Xiamen University Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China
- Department of Ophthalmology, the First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Yaqiong Yang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yuqian Wang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Youwen Zhang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jingbin Zhuang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yuting Zhang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Mei Shen
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jiankai Zhao
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Rongrong Zhang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yan Qiu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Shiying Li
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jiaoyue Hu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Wei Li
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jianfeng Wu
- Laboratory animal research center, Xiamen University, Xiamen, Fujian, 361102, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Steven J Fliesler
- Departments of Ophthalmology and Biochemistry and Neuroscience Graduate School, Jacobs School of Medicine and Biomedical Sciences, SUNY- University at Buffalo, Buffalo, NY, USA
- Research Service, VA Western New York Healthcare System, Buffalo, NY, USA
| | - Yi Liao
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China.
| | - Zuguo Liu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China.
- Xiamen University Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.
- Department of Ophthalmology, the First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
32
|
El-Hodiri HM, Bentley J, Reske A, Palazzo I, Campbell WA, Halloy NR, Fischer AJ. Formation of Müller glia-derived progenitor cells in retinas depleted of microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544205. [PMID: 37333380 PMCID: PMC10274900 DOI: 10.1101/2023.06.08.544205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Recent studies have demonstrated the complex coordination of pro-inflammatory signaling and reactive microglia/macrophage on the formation Müller glial-derived progenitor cells (MGPCs) in the retinas of fish, birds and mice. We generated scRNA-seq libraries to identify transcriptional changes in Müller glia (MG) that result from the depletion of microglia from the chick retina. We found significant changes in different networks of genes in MG in normal and damaged retinas when the microglia are ablated. We identified a failure of MG to upregulate Wnt-ligands, Heparin binding epidermal growth factor (HBEGF), Fibroblast growth factor (FGF), retinoic acid receptors and genes related to Notch-signaling. Inhibition of GSK3β, to simulate Wnt-signaling, failed to rescue the deficit in formation of proliferating MGPCs in damaged retinas missing microglia. By comparison, application of HBEGF or FGF2 completely rescued the formation of proliferating MGPCs in microglia-depleted retinas. Similarly, injection of a small molecule inhibitor to Smad3 or agonist to retinoic acid receptors partially rescued the formation of proliferating MGPCs in microglia-depleted damaged retinas. According to scRNA-seq libraries, patterns of expression of ligands, receptors, signal transducers and/or processing enzymes to cell-signaling via HBEGF, FGF, retinoic acid and TGFβ are rapidly and transiently upregulated by MG after neuronal damage, consistent with important roles for these cell-signaling pathways in regulating the formation of MGPCs. We conclude that quiescent and activated microglia have a significant impact upon the transcriptomic profile of MG. We conclude that signals produced by reactive microglia in damaged retinas stimulate MG to upregulate cell signaling through HBEGF, FGF and retinoic acid, and downregulate signaling through TGFβ/Smad3 to promote the reprogramming on MG into proliferating MGPCs.
Collapse
Affiliation(s)
- Heithem M. El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - James Bentley
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Alana Reske
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Isabella Palazzo
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD
| | - Warren A. Campbell
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Nicklaus R. Halloy
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
33
|
Meehan SD, Hu M, Veldman MB, Bhattacharya SK. Metabolomics dataset of zebrafish optic nerve regeneration after injury. Data Brief 2023; 48:109102. [PMID: 37383800 PMCID: PMC10293924 DOI: 10.1016/j.dib.2023.109102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 06/30/2023] Open
Abstract
Zebrafish (Danio rerio) have the capacity for successful adult optic nerve regeneration. In contrast, mammals lack this intrinsic ability and undergo irreversible neurodegeneration seen in glaucoma and other optic neuropathies. Optic nerve regeneration is often studied using optic nerve crush, a mechanical neurodegenerative model. Untargeted metabolomic studies within successful regenerative models are deficient. Evaluation of tissue metabolomic changes in active zebrafish optic nerve regeneration can elucidate prioritized metabolite pathways that can be targeted in mammalian systems for therapeutic development. Female and male (6 month to 1 year old wild type) right zebrafish optic nerves were crushed and collected three days after. Contralateral, uninjured optic nerves were collected as controls. The tissue was dissected from euthanized fish and frozen on dry ice. Samples were pooled for each category (female crush, female control, male crush, male control) and pooled at n = 31 to obtain sufficient metabolite concentrations for analysis. Optic nerve regeneration at 3 days post crush was demonstrated by microscope visualization of GFP fluorescence in Tg(gap43:GFP) transgenic fish. Metabolites were extracted using a Precellys Homogenizer and a serial extraction method: (1) 1:1 Methanol/Water and (2) 8:1:1 Acetonitrile/Methanol/Acetone. Metabolites were analyzed by untargeted liquid chromatography-mass spectrometry (LC MS-MS) profiling using a Q-Exactive Orbitrap instrument coupled with Vanquish Horizon Binary UHPLC LC-MS system. Metabolites were identified and quantified using Compound Discoverer 3.3 and isotopic internal metabolites standards.
Collapse
Affiliation(s)
- Sean D. Meehan
- Bascom Palmer Eye Institute University of Miami, 1638 NW 10th Avenue, Suite 707A, Miami, FL, 33136, United States
- Miami Integrative Metabolomics Research Center, Miami, FL, 33136, United States
- University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| | - Mengming Hu
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Matthew B. Veldman
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Sanjoy K. Bhattacharya
- Bascom Palmer Eye Institute University of Miami, 1638 NW 10th Avenue, Suite 707A, Miami, FL, 33136, United States
- Miami Integrative Metabolomics Research Center, Miami, FL, 33136, United States
- University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| |
Collapse
|
34
|
Pannetier P, Poulsen R, Gölz L, Coordes S, Stegeman H, Koegst J, Reger L, Braunbeck T, Hansen M, Baumann L. Reversibility of Thyroid Hormone System-Disrupting Effects on Eye and Thyroid Follicle Development in Zebrafish (Danio rerio) Embryos. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2023; 42:1276-1292. [PMID: 36920003 DOI: 10.1002/etc.5608] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 03/10/2023] [Indexed: 05/27/2023]
Abstract
Early vertebrate development is partially regulated by thyroid hormones (THs). Environmental pollutants that interact with the TH system (TH system-disrupting chemicals [THSDCs]) can have massively disrupting effects on this essential phase. Eye development of fish is directly regulated by THs and can, therefore, be used as a thyroid-related endpoint in endocrine disruptor testing. To evaluate the effects of THSDC-induced eye malformations during early development, zebrafish (Danio rerio) embryos were exposed for 5 days postfertilization (dpf) to either propylthiouracil, a TH synthesis inhibitor, or tetrabromobisphenol A, which interacts with TH receptors. Subsequently, one half of the embryos were exposed further to the THSDCs until 8 dpf, while the other half of the embryos were raised in clean water for 3 days to check for reversibility of effects. Continued THSDC exposure altered eye size and pigmentation and induced changes in the cellular structure of the retina. This correlated with morphological alterations of thyroid follicles as revealed by use of a transgenic zebrafish line. Interestingly, effects were partly reversible after a recovery period as short as 3 days. Results are consistent with changes in TH levels measured in different tissues of the embryos, for example, in the eyes. The results show that eye development in zebrafish embryos is very sensitive to THSDC treatment but able to recover quickly from early exposure by effective repair mechanisms. Environ Toxicol Chem 2023;42:1276-1292. © 2023 The Authors. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Pauline Pannetier
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Rikke Poulsen
- Environmental Metabolomics Laboratory, Department of Environmental Science, University of Aarhus, Aarhus, Denmark
| | - Lisa Gölz
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Sara Coordes
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Hanna Stegeman
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Johannes Koegst
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Luisa Reger
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Thomas Braunbeck
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Martin Hansen
- Environmental Metabolomics Laboratory, Department of Environmental Science, University of Aarhus, Aarhus, Denmark
| | - Lisa Baumann
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
- Amsterdam Institute for Life and Environment (A-LIFE), Section on Environmental Health & Toxicology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Sokolova N, Zilova L, Wittbrodt J. Unravelling the link between embryogenesis and adult stem cell potential in the ciliary marginal zone: A comparative study between mammals and teleost fish. Cells Dev 2023; 174:203848. [PMID: 37172718 DOI: 10.1016/j.cdev.2023.203848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
The discovery and study of adult stem cells have revolutionized regenerative medicine by offering new opportunities for treating various medical conditions. Anamniote stem cells, which retain their full proliferative capacity and full differentiation range throughout their lifetime, harbour a greater potential compared to mammalian adult stem cells, which only exhibit limited stem cell potential. Therefore, understanding the mechanisms underlying these differences is of significant interest. In this review, we examine the similarities and differences of adult retinal stem cells in anamniotes and mammals, from their embryonic stages in the optic vesicle to their residence in the postembryonic retinal stem cell niche, the ciliary marginal zone located in the retinal periphery. In anamniotes, developing precursors of retinal stem cells are exposed to various environmental cues during their migration in the complex morphogenetic remodelling of the optic vesicle to the optic cup. In contrast, their mammalian counterparts in the retinal periphery are primarily instructed by neighbouring tissues once they are in place. We explore the distinct modes of optic cup morphogenesis in mammals and teleost fish and highlight molecular mechanisms governing morphogenesis and stem cells instruction. The review concludes with the molecular mechanisms of ciliary marginal zone formation and offers a perspective on the impact of comparative single cell transcriptomic studies to reveal the evolutionary similarities and differences.
Collapse
Affiliation(s)
- Natalia Sokolova
- Centre for Organismal Studies Heidelberg, Germany; Heidelberg Biosciences International Graduate School, Germany
| | - Lucie Zilova
- Centre for Organismal Studies Heidelberg, Germany.
| | | |
Collapse
|
36
|
Caioni G, Merola C, Bertolucci C, Lucon-Xiccato T, Savaşçı BB, Massimi M, Colasante M, Fioravanti G, Cacciola NA, Ippoliti R, d'Angelo M, Perugini M, Benedetti E. Early-life exposure to environmentally relevant concentrations of triclocarban impairs ocular development in zebrafish larvae. CHEMOSPHERE 2023; 324:138348. [PMID: 36898440 DOI: 10.1016/j.chemosphere.2023.138348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Triclocarban (TCC), is an antimicrobial component in personal care products and it is one of the emerging contaminants since it has been detected in various environmental matrices. Its presence in human cord blood, breast milk, and maternal urine raised issues about its possible impact on development and increased concerns about the safety of daily exposure. This study aims to provide additional information about the effects of zebrafish early-life exposure to TCC on eye development and visual function. Zebrafish embryos were exposed to two concentrations of TCC (5 and 50 μg/L) for 4 days. TCC-mediated toxicity was assessed in larvae at the end of exposure and in the long term (20 days post fertilization; dpf), through different biological end-points. The experiments showed that TCC exposure influences the retinal architecture. In 4 dpf treated larvae, we found a less organized ciliary marginal zone, a decrease in the inner nuclear and inner plexiform layers, and a decrease in the retinal ganglion cell layer. Photoreceptor and inner plexiform layers showed an increase in 20 dpf larvae at lower and both concentrations, respectively. The expression levels of two genes involved in eye development (mitfb and pax6a) were both decreased at the concentration of 5 μg/L in 4 dpf larvae, and an increase in mitfb was observed in 5 μg/L-exposed 20 dpf larvae. Interestingly, 20 dpf larvae failed to discriminate between visual stimuli, demonstrating notable visual perception impairments due to compound. The results prompt us to hypothesize that early-life exposure to TCC may have severe and potentially long-term effect on zebrafish visual function.
Collapse
Affiliation(s)
- Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Carmine Merola
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy.
| | - Cristiano Bertolucci
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Tyrone Lucon-Xiccato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Beste Başak Savaşçı
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy; Unit of Evolutionary Biology/Systematic Zoology, Institute for Biochemistry and Biology, University of Potsdam, Potsdam, Germany.
| | - Mara Massimi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Martina Colasante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Giulia Fioravanti
- Department of Physical and Chemical Sciences University of L'Aquila, L'Aquila, Italy.
| | - Nunzio Antonio Cacciola
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy.
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Monia Perugini
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy.
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
37
|
Spitsbergen JB, Webster SE, Linn CL. Functional Changes in the Adult Mouse Retina using an Alpha7 Nicotinic Acetylcholine Receptor Agonist after Blast Exposure. Neuroscience 2023; 512:1-15. [PMID: 36572172 DOI: 10.1016/j.neuroscience.2022.12.017] [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: 06/17/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 12/25/2022]
Abstract
Currently, there is a lack of treatments for retinal neurotrauma. To address this issue, this study uses an alpha7 nAChR agonist, PNU-282987, to determine it effects on functional activity in the retina shortly after a traumatic blast exposure. The objectives of this research include: (1) examination of the cellular and functional damage associated with ocular blast exposure, and (2) evaluation of structural and functional changes that occur post PNU-282987 treatment. Significant ocular blast damage was induced in adult mice after exposure to a single blast of 35 psi to the left eye. Blast-exposed transgenic mice expressing tdTomato Müller glia were treated daily with eyedrops containing PNU-282987 for 4 weeks following the blast exposure. Antibody staining studies in these transgenic mice was conducted to examine lineage tracing and electroretinograms (ERGs) were obtained to examine functional changes. Blast exposure caused a significant loss of cells in all retinal layers after 4 weeks. Immunohistochemical analysis demonstrated tdTomato-positive labeled photoreceptors and retinal ganglion cells in blast-exposed mice treated with PNU-282987. ERG recordings were taken from control animals, from blast-damaged animals and from animals exposed to blast followed by 4 weeks of PNU-282987 treatment. Scotopic ERG recordings from blast-exposed mice had significantly decreased amplitudes of a-wave, b-wave, oscillatory potentials and flicker frequencies, which were prevented after PNU-282987 treatment. In photopic experiments, the PhNR response was reduced significantly after blast exposure but the decrease was prevented after treatment with PNU-282987. These are the first experiments that demonstrate preservation of retinal function after blast exposure using an alpha7 nAChR agonist.
Collapse
Affiliation(s)
- Jake B Spitsbergen
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI 49008, USA
| | - Sarah E Webster
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI 49008, USA
| | - Cindy L Linn
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI 49008, USA.
| |
Collapse
|
38
|
Boyd P, Campbell LJ, Hyde DR. Clcf1/Crlf1a-mediated signaling is neuroprotective and required for Müller glia proliferation in the light-damaged zebrafish retina. Front Cell Dev Biol 2023; 11:1142586. [PMID: 36846595 PMCID: PMC9950120 DOI: 10.3389/fcell.2023.1142586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Zebrafish possess the innate ability to fully regenerate any neurons lost following a retinal injury. This response is mediated by Müller glia that reprogram and divide asymmetrically to produce neuronal precursor cells that differentiate into the lost neurons. However, little is understood about the early signals that induce this response. Ciliary neurotrophic factor (CNTF) was previously shown to be both neuroprotective and pro-proliferative within the zebrafish retina, however CNTF is not expressed following injury. Here we demonstrate that alternative ligands of the Ciliary neurotrophic factor receptor (CNTFR), such as Cardiotrophin-like cytokine factor 1 (Clcf1) and Cytokine receptor-like factor 1a (Crlf1a), are expressed within Müller glia of the light-damaged retina. We found that CNTFR, Clcf1, and Crlf1a are required for Müller glia proliferation in the light-damaged retina. Furthermore, intravitreal injection of CLCF1/CRLF1 protected against rod photoreceptor cell death in the light-damaged retina and induced proliferation of rod precursor cells in the undamaged retina, but not Müller glia. While rod precursor cell proliferation was previously shown to be Insulin-like growth factor 1 receptor (IGF-1R)-dependent, co-injection of IGF-1 with CLCF1/CRLF1 failed to induce further proliferation of either Müller glia or rod precursor cells. Together, these findings demonstrate that CNTFR ligands have a neuroprotective effect and are required for induction of Müller glia proliferation in the light-damaged zebrafish retina.
Collapse
Affiliation(s)
| | | | - David R. Hyde
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, and Center for Zebrafish Research, Galvin Life Sciences Building, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
39
|
Gupta S, Sharma P, Chaudhary M, Premraj S, Kaur S, Vijayan V, Arun MG, Prasad NG, Ramachandran R. Pten associates with important gene regulatory network to fine-tune Müller glia-mediated zebrafish retina regeneration. Glia 2023; 71:259-283. [PMID: 36128720 DOI: 10.1002/glia.24270] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/11/2022]
Abstract
Unlike mammals, zebrafish possess a remarkable ability to regenerate damaged retina after an acute injury. Retina regeneration in zebrafish involves the induction of Müller glia-derived progenitor cells (MGPCs) exhibiting stem cell-like characteristics, which are capable of restoring all retinal cell-types. The induction of MGPC through Müller glia-reprograming involves several cellular, genetic and biochemical events soon after a retinal injury. Despite the knowledge on the importance of Phosphatase and tensin homolog (Pten), which is a dual-specificity phosphatase and tumor suppressor in the maintaining of cellular homeostasis, its importance during retina regeneration remains unknown. Here, we explored the importance of Pten during zebrafish retina regeneration. The Pten gets downregulated upon retinal injury and is absent from the MGPCs, which is essential to trigger Akt-mediated cellular proliferation essential for retina regeneration. We found that the downregulation of Pten in the post-injury retina accelerates MGPCs formation, while its overexpression restricts the regenerative response. We observed that Pten regulates the proliferation of MGPCs not only through Akt pathway but also by Mmp9/Notch signaling. Mmp9-activity is essential to induce the proliferation of MGPCs in the absence of Pten. Lastly, we show that expression of Pten is fine-tuned through Mycb/histone deacetylase1 and Tgf-β signaling. The present study emphasizes on the stringent regulation of Pten and its crucial involvement during the zebrafish retina regeneration.
Collapse
Affiliation(s)
- Shivangi Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Poonam Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Mansi Chaudhary
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Sharanya Premraj
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Simran Kaur
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Vijithkumar Vijayan
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Manas Geeta Arun
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Nagaraj Guru Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Rajesh Ramachandran
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| |
Collapse
|
40
|
Kaplan L, Drexler C, Pfaller AM, Brenna S, Wunderlich KA, Dimitracopoulos A, Merl-Pham J, Perez MT, Schlötzer-Schrehardt U, Enzmann V, Samardzija M, Puig B, Fuchs P, Franze K, Hauck SM, Grosche A. Retinal regions shape human and murine Müller cell proteome profile and functionality. Glia 2023; 71:391-414. [PMID: 36334068 DOI: 10.1002/glia.24283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/08/2022]
Abstract
The human macula is a highly specialized retinal region with pit-like morphology and rich in cones. How Müller cells, the principal glial cell type in the retina, are adapted to this environment is still poorly understood. We compared proteomic data from cone- and rod-rich retinae from human and mice and identified different expression profiles of cone- and rod-associated Müller cells that converged on pathways representing extracellular matrix and cell adhesion. In particular, epiplakin (EPPK1), which is thought to play a role in intermediate filament organization, was highly expressed in macular Müller cells. Furthermore, EPPK1 knockout in a human Müller cell-derived cell line led to a decrease in traction forces as well as to changes in cell size, shape, and filopodia characteristics. We here identified EPPK1 as a central molecular player in the region-specific architecture of the human retina, which likely enables specific functions under the immense mechanical loads in vivo.
Collapse
Affiliation(s)
- Lew Kaplan
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Corinne Drexler
- Max Perutz Labs, Department of Biochemistry and Cell Biology, University of Vienna, Vienna Biocenter Campus (VBC), Vienna, Austria
- Vienna Biocenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Anna M Pfaller
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kirsten A Wunderlich
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Andrea Dimitracopoulos
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Juliane Merl-Pham
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Maria-Theresa Perez
- Department of Clinical Sciences, Division of Ophthalmology, Lund University, Lund, Sweden
- NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden
| | | | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Marijana Samardzija
- Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Fuchs
- Max Perutz Labs, Department of Biochemistry and Cell Biology, University of Vienna, Vienna Biocenter Campus (VBC), Vienna, Austria
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Institute of Medical Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
41
|
Konar G, Flickinger Z, Sharma S, Vallone K, Lyon C, Doshier C, Lyon W, Patton JG. Damage-induced senescent immune cells regulate regeneration of the zebrafish retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524296. [PMID: 36711649 PMCID: PMC9882244 DOI: 10.1101/2023.01.16.524296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Zebrafish spontaneously regenerate their retina in response to damage through the action of Müller glia. Even though Müller glia (MG) are conserved in higher vertebrates, the capacity to regenerate retinal damage is lost. Recent work has focused on the regulation of inflammation during tissue regeneration with precise temporal roles for macrophages and microglia. Senescent cells that have withdrawn from the cell cycle have mostly been implicated in aging, but are still metabolically active, releasing proinflammatory signaling molecules as part of the Senescence Associated Secretory Phenotype (SASP). Here, we discover that in response to retinal damage, a subset of cells expressing markers of microglia/macrophages also express markers of senescence. These cells display a temporal pattern of appearance and clearance during retina regeneration. Premature removal of senescent cells by senolytic treatment led to a decrease in proliferation and incomplete repair of the ganglion cell layer after NMDA damage. Our results demonstrate a role for modulation of senescent cell responses to balance inflammation, regeneration, plasticity, and repair as opposed to fibrosis and scarring.
Collapse
Affiliation(s)
| | | | - Shivani Sharma
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - Kyle Vallone
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - Charles Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - Claire Doshier
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - William Lyon
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| | - James G. Patton
- Department of Biological Sciences, Vanderbilt University, Nashville TN, USA
| |
Collapse
|
42
|
Marchese NA, Ríos MN, Guido ME. Müller glial cell photosensitivity: a novel function bringing higher complexity to vertebrate retinal physiology. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2023. [DOI: 10.1016/j.jpap.2023.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
43
|
Bise T, Pfefferli C, Bonvin M, Taylor L, Lischer HEL, Bruggmann R, Jaźwińska A. The regeneration-responsive element careg monitors activation of Müller glia after MNU-induced damage of photoreceptors in the zebrafish retina. Front Mol Neurosci 2023; 16:1160707. [PMID: 37138703 PMCID: PMC10149768 DOI: 10.3389/fnmol.2023.1160707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
In contrast to mammals, zebrafish can regenerate their damaged photoreceptors. This capacity depends on the intrinsic plasticity of Müller glia (MG). Here, we identified that the transgenic reporter careg, a marker of regenerating fin and heart, also participates in retina restoration in zebrafish. After methylnitrosourea (MNU) treatment, the retina became deteriorated and contained damaged cell types including rods, UV-sensitive cones and the outer plexiform layer. This phenotype was associated with the induction of careg expression in a subset of MG until the reconstruction of the photoreceptor synaptic layer. Single-cell RNA sequencing (scRNAseq) analysis of regenerating retinas revealed a population of immature rods, defined by high expression of rhodopsin and the ciliogenesis gene meig1, but low expression of phototransduction genes. Furthermore, cones displayed deregulation of metabolic and visual perception genes in response to retina injury. Comparison between careg:EGFP expressing and non-expressing MG demonstrated that these two subpopulations are characterized by distinct molecular signatures, suggesting their heterogenous responsiveness to the regenerative program. Dynamics of ribosomal protein S6 phosphorylation showed that TOR signaling became progressively switched from MG to progenitors. Inhibition of TOR with rapamycin reduced the cell cycle activity, but neither affected careg:EGFP expression in MG, nor prevented restoration of the retina structure. This indicates that MG reprogramming, and progenitor cell proliferation might be regulated by distinct mechanisms. In conclusion, the careg reporter detects activated MG, and provides a common marker of regeneration-competent cells in diverse zebrafish organs, including the retina.
Collapse
Affiliation(s)
- Thomas Bise
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Marylène Bonvin
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Lea Taylor
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Heidi E. L. Lischer
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- *Correspondence: Anna Jaźwińska,
| |
Collapse
|
44
|
Lee Y, Yang J. Frozen Block Tissue Staining for Eye Structure of Zebrafish Embryo. Methods Mol Biol 2023; 2678:191-198. [PMID: 37326715 DOI: 10.1007/978-1-0716-3255-0_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Histology is a technique used to study the morphology of cell structures by cutting samples into thin sections. Histological cross-section and staining are the techniques needed to visualize the morphology of cell tissues. A suitable tissue staining experiment was created to observe changes in the retinal layer in zebrafish embryos. Zebrafish have a human-like visual system, retina, and eye structures. Due to the small size of zebrafish and undeveloped bones in the embryonic stage, the resistance through cross-section is inevitably small. Here, we present optimized protocol changes in eye tissue of zebrafish using frozen blocks.
Collapse
Affiliation(s)
- Yujin Lee
- Department of Ophthalmology, Inje University Busan Paik Hospital, Busan, South Korea
| | - Jaewook Yang
- Department of Ophthalmology, Inje University Busan Paik Hospital, Busan, South Korea.
| |
Collapse
|
45
|
Diacou R, Nandigrami P, Fiser A, Liu W, Ashery-Padan R, Cvekl A. Cell fate decisions, transcription factors and signaling during early retinal development. Prog Retin Eye Res 2022; 91:101093. [PMID: 35817658 PMCID: PMC9669153 DOI: 10.1016/j.preteyeres.2022.101093] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The development of the vertebrate eyes is a complex process starting from anterior-posterior and dorso-ventral patterning of the anterior neural tube, resulting in the formation of the eye field. Symmetrical separation of the eye field at the anterior neural plate is followed by two symmetrical evaginations to generate a pair of optic vesicles. Next, reciprocal invagination of the optic vesicles with surface ectoderm-derived lens placodes generates double-layered optic cups. The inner and outer layers of the optic cups develop into the neural retina and retinal pigment epithelium (RPE), respectively. In vitro produced retinal tissues, called retinal organoids, are formed from human pluripotent stem cells, mimicking major steps of retinal differentiation in vivo. This review article summarizes recent progress in our understanding of early eye development, focusing on the formation the eye field, optic vesicles, and early optic cups. Recent single-cell transcriptomic studies are integrated with classical in vivo genetic and functional studies to uncover a range of cellular mechanisms underlying early eye development. The functions of signal transduction pathways and lineage-specific DNA-binding transcription factors are dissected to explain cell-specific regulatory mechanisms underlying cell fate determination during early eye development. The functions of homeodomain (HD) transcription factors Otx2, Pax6, Lhx2, Six3 and Six6, which are required for early eye development, are discussed in detail. Comprehensive understanding of the mechanisms of early eye development provides insight into the molecular and cellular basis of developmental ocular anomalies, such as optic cup coloboma. Lastly, modeling human development and inherited retinal diseases using stem cell-derived retinal organoids generates opportunities to discover novel therapies for retinal diseases.
Collapse
Affiliation(s)
- Raven Diacou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Prithviraj Nandigrami
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wei Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ruth Ashery-Padan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ales Cvekl
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
46
|
Ahmad I. CRISPR/Cas9-A Promising Therapeutic Tool to Cure Blindness: Current Scenario and Future Prospects. Int J Mol Sci 2022; 23:11482. [PMID: 36232782 PMCID: PMC9569777 DOI: 10.3390/ijms231911482] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
CRISPR-based targeted genome editing is bringing revolutionary changes in the research arena of biological sciences. CRISPR/Cas9 has been explored as an efficient therapeutic tool for the treatment of genetic diseases. It has been widely used in ophthalmology research by using mouse models to correct pathogenic mutations in the eye stem cells. In recent studies, CRISPR/Cas9 has been used to correct a large number of mutations related to inherited retinal disorders. In vivo therapeutic advantages for retinal diseases have been successfully achieved in some rodents. Current advances in the CRISPR-based gene-editing domain, such as modified Cas variants and delivery approaches have optimized its application to treat blindness. In this review, recent progress and challenges of the CRISPR-Cas system have been discussed to cure blindness and its prospects.
Collapse
Affiliation(s)
- Irshad Ahmad
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; ; Tel.: +966-13-8608393
- Interdisciplinary Research Center for Membranes and Water Security, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia
| |
Collapse
|
47
|
Cao J, Zhang Y, Zhang P, Zhang Z, Zhang B, Feng Y, Li Z, Yang Y, Meng Q, He L, Cai Y, Wang Z, Li J, Chen X, Liu H, Hong A, Zheng W, Chen X. Turning gray selenium into a nanoaccelerator of tissue regeneration by PEG modification. Bioact Mater 2022; 15:131-144. [PMID: 35386336 PMCID: PMC8940942 DOI: 10.1016/j.bioactmat.2021.12.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/30/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023] Open
Abstract
Selenium (Se) is an essential trace element involved in nearly all human physiological processes but suffers from a narrow margin between benefit and toxicity. The nanoform of selenium has been proven shown to be more bioavailable and less toxic, yet significant challenges remain regarding the efficient and feasible synthesis of biologically active nanoselenium. In addition, although nanoselenium has shown a variety of biological activities, more interesting nanoselenium features are expected. In this work, hydrosoluble nanoselenium termed Nano-Se in the zero oxidation state was synthesized between gray Se and PEG. A zebrafish screen was carried out in zebrafish larvae cocultured with Nano-Se. Excitingly, Nano-Se promoted the action of the FGFR, Wnt, and VEGF signaling pathways, which play crucial roles in tissue regeneration. As expected, Nano-Se not only achieved the regeneration of zebrafish tail fins and mouse skin but also promoted the repair of skin in diabetic mice while maintaining a profitable safe profile. In brief, the Nano-Se reported here provided an efficient and feasible method for bioactive nanoselenium synthesis and not only expanded the application of nanoselenium to regenerative medicine but also likely reinvigorated efforts for discovering more peculiarunique biofunctions of nanoselenium in a great variety of human diseases. It was found that selenium nanoparticles through FGFR、Wnt、VEGFR signal pathway to promote tissue regeneration; Development a new water-soluble, bio-compatible, zero oxidation state Nano-Se; Development a new efficient and safe nano-biologic agent for promoting tissue regeneration.
Collapse
Affiliation(s)
- Jieqiong Cao
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Yibo Zhang
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Peiguang Zhang
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Zilei Zhang
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Bihui Zhang
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Yanxian Feng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, China
| | - Zhixin Li
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Yiqi Yang
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Qilin Meng
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Liu He
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Yulin Cai
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Zhenyu Wang
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Jie Li
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Xue Chen
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
| | - Hongwei Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - An Hong
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
- Corresponding author.
| | - Wenjie Zheng
- Department of Chemistry, Jinan University, Guangzhou, China
- Corresponding author.
| | - Xiaojia Chen
- Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China
- Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510240, China
- Corresponding author. Institute of Biomedicine & Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial biotechnology drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, China.
| |
Collapse
|
48
|
Sharma P, Ramachandran R. Retina regeneration: lessons from vertebrates. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac012. [PMID: 38596712 PMCID: PMC10913848 DOI: 10.1093/oons/kvac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 04/11/2024]
Abstract
Unlike mammals, vertebrates such as fishes and frogs exhibit remarkable tissue regeneration including the central nervous system. Retina being part of the central nervous system has attracted the interest of several research groups to explore its regenerative ability in different vertebrate models including mice. Fishes and frogs completely restore the size, shape and tissue structure of an injured retina. Several studies have unraveled molecular mechanisms underlying retina regeneration. In teleosts, soon after injury, the Müller glial cells of the retina reprogram to form a proliferating population of Müller glia-derived progenitor cells capable of differentiating into various neural cell types and Müller glia. In amphibians, the transdifferentiation of retinal pigment epithelium and differentiation of ciliary marginal zone cells contribute to retina regeneration. In chicks and mice, supplementation with external growth factors or genetic modifications cause a partial regenerative response in the damaged retina. The initiation of retina regeneration is achieved through sequential orchestration of gene expression through controlled modulations in the genetic and epigenetic landscape of the progenitor cells. Several developmental biology pathways are turned on during the Müller glia reprogramming, retinal pigment epithelium transdifferentiation and ciliary marginal zone differentiation. Further, several tumorigenic pathways and gene expression events also contribute to the complete regeneration cascade of events. In this review, we address the various retinal injury paradigms and subsequent gene expression events governed in different vertebrate species. Further, we compared how vertebrates such as teleost fishes and amphibians can achieve excellent regenerative responses in the retina compared with their mammalian counterparts.
Collapse
Affiliation(s)
- Poonam Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| | - Rajesh Ramachandran
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| |
Collapse
|
49
|
Zebrafish and inherited photoreceptor disease: Models and insights. Prog Retin Eye Res 2022; 91:101096. [PMID: 35811244 DOI: 10.1016/j.preteyeres.2022.101096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/21/2022]
Abstract
Photoreceptor dysfunctions and degenerative diseases are significant causes of vision loss in patients, with few effective treatments available. Targeted interventions to prevent or reverse photoreceptor-related vision loss are not possible without a thorough understanding of the underlying mechanism leading to disease, which is exceedingly difficult to accomplish in the human system. Cone diseases are particularly challenging to model, as some popular genetically modifiable model animals are nocturnal with a rod-dominant visual system and cones that have dissimilarities to human cones. As a result, cone diseases, which affect visual acuity, colour perception, and central vision in patients, are generally poorly understood in terms of pathology and mechanism. Zebrafish (Danio rerio) provide the opportunity to model photoreceptor diseases in a diurnal vertebrate with a cone-rich retina which develops many macular degeneration-like pathologies. Zebrafish undergo external development, allowing early-onset retinal diseases to be detected and studied, and many ophthalmic tools are available for zebrafish visual assessment during development and adulthood. There are numerous zebrafish models of photoreceptor disease, spanning the various types of photoreceptor disease (developmental, rod, cone, and mixed photoreceptor diseases) and genetic/molecular cause. In this review, we explore the features of zebrafish that make them uniquely poised to model cone diseases, summarize the established zebrafish models of inherited photoreceptor disease, and discuss how disease in these models compares to the human presentation, where applicable. Further, we highlight the contributions of these zebrafish models to our understanding of photoreceptor biology and disease, and discuss future directions for utilising and investigating these diverse models.
Collapse
|
50
|
Lee CJ, Paull GC, Tyler CR. Improving zebrafish laboratory welfare and scientific research through understanding their natural history. Biol Rev Camb Philos Soc 2022; 97:1038-1056. [PMID: 34983085 PMCID: PMC9303617 DOI: 10.1111/brv.12831] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
Globally, millions of zebrafish (Danio rerio) are used for scientific laboratory experiments for which researchers have a duty of care, with legal obligations to consider their welfare. Considering the growing use of the zebrafish as a vertebrate model for addressing a diverse range of scientific questions, optimising their laboratory conditions is of major importance for both welfare and improving scientific research. However, most guidelines for the care and breeding of zebrafish for research are concerned primarily with maximising production and minimising costs and pay little attention to the effects on welfare of the environments in which the fish are maintained, or how those conditions affect their scientific research. Here we review the physical and social conditions in which laboratory zebrafish are kept, identifying and drawing attention to factors likely to affect their welfare and experimental science. We also identify a fundamental lack knowledge of how zebrafish interact with many biotic and abiotic features in their natural environment to support ways to optimise zebrafish health and well-being in the laboratory, and in turn the quality of scientific data produced. We advocate that the conditions under which zebrafish are maintained need to become a more integral part of research and that we understand more fully how they influence experimental outcome and in turn interpretations of the data generated.
Collapse
Affiliation(s)
- Carole J. Lee
- Biosciences, Geoffrey Pope BuildingUniversity of ExeterStocker RoadExeterEX4 4QDU.K.
| | - Gregory C. Paull
- Biosciences, Geoffrey Pope BuildingUniversity of ExeterStocker RoadExeterEX4 4QDU.K.
| | - Charles R. Tyler
- Biosciences, Geoffrey Pope BuildingUniversity of ExeterStocker RoadExeterEX4 4QDU.K.
| |
Collapse
|