Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Glia ; 72(4): 759-776, 2024 04.
Article in English | MEDLINE | ID: mdl-38225726

ABSTRACT

Regenerative abilities are not evenly distributed across the animal kingdom. The underlying modalities are also highly variable. Retinal repair can involve the mobilization of different cellular sources, including ciliary marginal zone (CMZ) stem cells, the retinal pigmented epithelium (RPE), or Müller glia. To investigate whether the magnitude of retinal damage influences the regeneration modality of the Xenopus retina, we developed a model based on cobalt chloride (CoCl2 ) intraocular injection, allowing for a dose-dependent control of cell death extent. Analyses in Xenopus laevis revealed that limited CoCl2 -mediated neurotoxicity only triggers cone loss and results in a few Müller cells reentering the cell cycle. Severe CoCl2 -induced retinal degeneration not only potentializes Müller cell proliferation but also enhances CMZ activity and unexpectedly triggers RPE reprogramming. Surprisingly, reprogrammed RPE self-organizes into an ectopic mini-retina-like structure laid on top of the original retina. It is thus likely that the injury paradigm determines the awakening of different stem-like cell populations. We further show that these cellular sources exhibit distinct neurogenic capacities without any bias towards lost cells. This is particularly striking for Müller glia, which regenerates several types of neurons, but not cones, the most affected cell type. Finally, we found that X. tropicalis also has the ability to recruit Müller cells and reprogram its RPE following CoCl2 -induced damage, whereas only CMZ involvement was reported in previously examined degenerative models. Altogether, these findings highlight the critical role of the injury paradigm and reveal that three cellular sources can be reactivated in the very same degenerative model.


Subject(s)
Cobalt , Retinal Degeneration , Animals , Xenopus laevis/physiology , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Retina , Regeneration/physiology , Cell Proliferation , Neuroglia/metabolism
2.
Cell Death Discov ; 10(1): 48, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38272861

ABSTRACT

Glaucoma is a multifactorial neurodegenerative disease characterized by the progressive and irreversible degeneration of the optic nerve and retinal ganglion cells. Despite medical advances aiming at slowing degeneration, around 40% of treated glaucomatous patients will undergo vision loss. It is thus of utmost importance to have a better understanding of the disease and to investigate more deeply its early causes. The transcriptional coactivator YAP, an important regulator of eye homeostasis, has recently drawn attention in the glaucoma research field. Here we show that Yap conditional knockout mice (Yap cKO), in which the deletion of Yap is induced in both Müller glia (i.e. the only retinal YAP-expressing cells) and the non-pigmented epithelial cells of the ciliary body, exhibit a breakdown of the aqueous-blood barrier, accompanied by a progressive collapse of the ciliary body. A similar phenotype is observed in human samples that we obtained from patients presenting with uveitis. In addition, aged Yap cKO mice harbor glaucoma-like features, including deregulation of key homeostatic Müller-derived proteins, retinal vascular defects, optic nerve degeneration and retinal ganglion cell death. Finally, transcriptomic analysis of Yap cKO retinas pointed to early-deregulated genes involved in extracellular matrix organization potentially underlying the onset and/or progression of the observed phenotype. Together, our findings reveal the essential role of YAP in preserving the integrity of the ciliary body and retinal ganglion cells, thereby preventing the onset of uveitic glaucoma-like features.

3.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38108453

ABSTRACT

A growing wealth of data suggest that reactive oxygen species (ROS) signalling might be crucial in conferring embryonic or adult stem cells their specific properties. However, how stem cells control ROS production and scavenging, and how ROS in turn contribute to stemness, remain poorly understood. Using the Xenopus retina as a model system, we first investigated the redox status of retinal stem cells (RSCs). We discovered that they exhibit higher ROS levels compared with progenitors and retinal neurons, and express a set of specific redox genes. We next addressed the question of ROS functional involvement in these cells. Using pharmacological or genetic tools, we demonstrate that inhibition of NADPH oxidase-dependent ROS production increases the proportion of quiescent RSCs. Surprisingly, this is accompanied by an apparent acceleration of the mean division speed within the remaining proliferating pool. Our data further unveil that such impact on RSC cell cycling is achieved by modulation of the Wnt/Hedgehog signalling balance. Altogether, we highlight that RSCs exhibit distinctive redox characteristics and exploit NADPH oxidase signalling to limit quiescence and fine-tune their proliferation rate.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Animals , Xenopus laevis/metabolism , Reactive Oxygen Species , Cell Proliferation , Hedgehog Proteins , Retina/metabolism , Adult Stem Cells/metabolism , NADPH Oxidases/genetics , Wnt Signaling Pathway
4.
J Vis Exp ; (200)2023 10 13.
Article in English | MEDLINE | ID: mdl-37902314

ABSTRACT

Retinal neurodegenerative diseases are the leading causes of blindness. Among numerous therapeutic strategies being explored, stimulating self-repair recently emerged as particularly appealing. A cellular source of interest for retinal repair is the Müller glial cell, which harbors stem cell potential and an extraordinary regenerative capacity in anamniotes. This potential is, however, very limited in mammals. Studying the molecular mechanisms underlying retinal regeneration in animal models with regenerative capabilities should provide insights into how to unlock the latent ability of mammalian Müller cells to regenerate the retina. This is a key step for the development of therapeutic strategies in regenerative medicine. To this aim, we developed several retinal injury paradigms in Xenopus: a mechanical retinal injury, a transgenic line allowing for nitroreductase-mediated photoreceptor conditional ablation, a retinitis pigmentosa model based on CRISPR/Cas9-mediated rhodopsin knockout, and a cytotoxic model driven by intraocular CoCl2 injections. Highlighting their advantages and disadvantages, we describe here this series of protocols that generate various degenerative conditions and allow the study of retinal regeneration in Xenopus.


Subject(s)
Retina , Retinitis Pigmentosa , Animals , Xenopus laevis , Larva , Retina/metabolism , Animals, Genetically Modified , Retinitis Pigmentosa/metabolism , Mammals
5.
JCI Insight ; 8(21)2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37768732

ABSTRACT

Retinitis pigmentosa (RP) is the most common inherited retinal disease (IRD) and is characterized by photoreceptor degeneration and progressive vision loss. We report 4 patients presenting with RP from 3 unrelated families with variants in TBC1D32, which to date has never been associated with an IRD. To validate TBC1D32 as a putative RP causative gene, we combined Xenopus in vivo approaches and human induced pluripotent stem cell-derived (iPSC-derived) retinal models. Our data showed that TBC1D32 was expressed during retinal development and that it played an important role in retinal pigment epithelium (RPE) differentiation. Furthermore, we identified a role for TBC1D32 in ciliogenesis of the RPE. We demonstrated elongated ciliary defects that resulted in disrupted apical tight junctions, loss of functionality (delayed retinoid cycling and altered secretion balance), and the onset of an epithelial-mesenchymal transition-like phenotype. Last, our results suggested photoreceptor differentiation defects, including connecting cilium anomalies, that resulted in impaired trafficking to the outer segment in cones and rods in TBC1D32 iPSC-derived retinal organoids. Overall, our data highlight a critical role for TBC1D32 in the retina and demonstrate that TBC1D32 mutations lead to RP. We thus identify TBC1D32 as an IRD-causative gene.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Retinitis Pigmentosa , Humans , Retina , Retinitis Pigmentosa/genetics , Retinal Degeneration/genetics , Retinal Pigment Epithelium , Adaptor Proteins, Signal Transducing
6.
Cells ; 11(18)2022 09 16.
Article in English | MEDLINE | ID: mdl-36139472

ABSTRACT

Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. Although recent works strongly suggest that inhibiting GSK3 might also be a promising therapeutic approach for retinal degenerative diseases, its full potential is still under-evaluated. In this review, we summarize the literature on the role of GSK3 on the main cellular functions reported as deregulated during retinal degeneration, such as glucose homeostasis which is critical for photoreceptor survival, or oxidative stress, a major component of retinal degeneration. We also discuss the interest in targeting GSK3 for its beneficial effects on inflammation, for reducing neovascularization that occurs in some retinal dystrophies, or for cell-based therapy by enhancing Müller glia cell proliferation in diseased retina. Together, although GSK3 inhibitors hold promise as therapeutic agents, we highlight the complexity of targeting such a multitasked kinase and the need to increase our knowledge of the impact of reducing GSK3 activity on these multiple cellular pathways and biological processes.


Subject(s)
Retinal Degeneration , Ependymoglial Cells , Glucose/pharmacology , Glycogen Synthase Kinase 3/pharmacology , Humans , Retina , Retinal Degeneration/drug therapy
7.
Elife ; 112022 07 15.
Article in English | MEDLINE | ID: mdl-35838349

ABSTRACT

In multicellular eukaryotic organisms, the initiation of DNA replication occurs asynchronously throughout S-phase according to a regulated replication timing program. Here, using Xenopus egg extracts, we showed that Yap (Yes-associated protein 1), a downstream effector of the Hippo signalling pathway, is required for the control of DNA replication dynamics. We found that Yap is recruited to chromatin at the start of DNA replication and identified Rif1, a major regulator of the DNA replication timing program, as a novel Yap binding protein. Furthermore, we show that either Yap or Rif1 depletion accelerates DNA replication dynamics by increasing the number of activated replication origins. In Xenopus embryos, using a Trim-Away approach during cleavage stages devoid of transcription, we found that either Yap or Rif1 depletion triggers an acceleration of cell divisions, suggesting a shorter S-phase by alterations of the replication program. Finally, our data show that Rif1 knockdown leads to defects in the partitioning of early versus late replication foci in retinal stem cells, as we previously showed for Yap. Altogether, our findings unveil a non-transcriptional role for Yap in regulating replication dynamics. We propose that Yap and Rif1 function as brakes to control the DNA replication program in early embryos and post-embryonic stem cells.


Subject(s)
Replication Origin , Telomere-Binding Proteins , Animals , DNA Replication , DNA Replication Timing , S Phase/genetics , Telomere-Binding Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
8.
Cells ; 11(5)2022 02 25.
Article in English | MEDLINE | ID: mdl-35269429

ABSTRACT

Retinitis pigmentosa is an inherited retinal dystrophy that ultimately leads to blindness due to the progressive degeneration of rod photoreceptors and the subsequent non-cell autonomous death of cones. Rhodopsin is the most frequently mutated gene in this disease. We here developed rhodopsin gene editing-based models of retinitis pigmentosa in two Xenopus species, Xenopus laevis and Xenopus tropicalis, by using CRISPR/Cas9 technology. In both of them, loss of rhodopsin function results in massive rod cell degeneration characterized by progressive shortening of outer segments and occasional cell death. This is followed by cone morphology deterioration. Despite these apparently similar degenerative environments, we found that Müller glial cells behave differently in Xenopus laevis and Xenopus tropicalis. While a significant proportion of Müller cells re-enter into the cell cycle in Xenopus laevis, their proliferation remains extremely limited in Xenopus tropicalis. This work thus reveals divergent responses to retinal injury in closely related species. These models should help in the future to deepen our understanding of the mechanisms that have shaped regeneration during evolution, with tremendous differences across vertebrates.


Subject(s)
Retinitis Pigmentosa , Rhodopsin , Animals , CRISPR-Cas Systems/genetics , Disease Models, Animal , Ependymoglial Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism
9.
eNeuro ; 8(5)2021.
Article in English | MEDLINE | ID: mdl-34518365

ABSTRACT

Glycogen synthase kinase 3 (GSK3) proteins (GSK3α and GSK3ß) are key mediators of signaling pathways, with crucial roles in coordinating fundamental biological processes during neural development. Here we show that the complete loss of GSK3 signaling in mouse retinal progenitors leads to microphthalmia with broad morphologic defects. A single wild-type allele of either Gsk3α or Gsk3ß is able to rescue this phenotype. In this genetic context, all cell types are present in a functional retina. However, we unexpectedly detected a large number of cells in the inner nuclear layer expressing retinal ganglion cell (RGC)-specific markers (called displaced RGCs, dRGCs) when at least one allele of Gsk3α is expressed. The excess of dRGCs leads to an increased number of axons projecting into the ipsilateral medial terminal nucleus, an area of the brain belonging to the non-image-forming visual circuit and poorly targeted by RGCs in wild-type retina. Transcriptome analysis and optomotor response assay suggest that at least a subset of dRGCs in Gsk3 mutant mice are direction-selective RGCs. Our study thus uncovers a unique role of GSK3 in controlling the production of ganglion cells in the inner nuclear layer, which correspond to dRGCs, a rare and poorly characterized retinal cell type.


Subject(s)
Glycogen Synthase Kinase 3 , Retinal Ganglion Cells , Animals , Axons , Glycogen Synthase Kinase 3/genetics , Mice , Retina
10.
Cell Death Dis ; 11(8): 631, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32801350

ABSTRACT

Hippo signalling regulates eye growth during embryogenesis through its effectors YAP and TAZ. Taking advantage of a Yap heterozygous mouse line, we here sought to examine its function in adult neural retina, where YAP expression is restricted to Müller glia. We first discovered an unexpected temporal dynamic of gene compensation. At postnatal stages, Taz upregulation occurs, leading to a gain of function-like phenotype characterised by EGFR signalling potentiation and delayed cell-cycle exit of retinal progenitors. In contrast, Yap+/- adult retinas no longer exhibit TAZ-dependent dosage compensation. In this context, Yap haploinsufficiency in aged individuals results in Müller glia dysfunction, late-onset cone degeneration, and reduced cone-mediated visual response. Alteration of glial homeostasis and altered patterns of cone opsins were also observed in Müller cell-specific conditional Yap-knockout aged mice. Together, this study highlights a novel YAP function in Müller cells for the maintenance of retinal tissue homeostasis and the preservation of cone integrity. It also suggests that YAP haploinsufficiency should be considered and explored as a cause of cone dystrophies in human.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cone Dystrophy/pathology , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Haploinsufficiency/genetics , Animals , Animals, Newborn , Carrier Proteins/metabolism , Cell Cycle , Cell Proliferation , Cone Dystrophy/genetics , ErbB Receptors/metabolism , Gene Deletion , Gene Expression Regulation , Homeostasis , Mice, Knockout , Models, Biological , Opsins/metabolism , Phenotype , Retina/pathology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Stem Cells/metabolism , Trans-Activators/metabolism , YAP-Signaling Proteins
11.
Curr Opin Genet Dev ; 64: 52-59, 2020 10.
Article in English | MEDLINE | ID: mdl-32619816

ABSTRACT

Retinal regeneration efficiency from Müller glia varies tremendously among vertebrate species, being extremely limited in mammals. Efforts towards the identification of molecular mechanisms underlying Müller cell proliferative and neurogenic potential should help finding strategies to awake them and ensure regeneration in mammals. We provide here an update on the most recent and original progresses made in the field. These include remarkable discoveries regarding (i) unprecedented cross-species comparison of Müller cell transcriptome using single-cell technologies, (ii) the identification of new strategies to promote both the proliferative and the neurogenic potential of mammalian Müller cells, (iii) the role of the epigenome in regulating Müller glia plasticity, (iv) miRNA-based regulatory mechanisms of Müller cell response to injury, and (v) the influence of inflammatory signals on the regenerative process.


Subject(s)
Cellular Reprogramming , Ependymoglial Cells/cytology , Nerve Regeneration , Neuroglia/cytology , Retina/physiology , Wound Healing , Animals , Cell Proliferation , Mammals , Retina/injuries
12.
Cold Spring Harb Protoc ; 2019(8)2019 08 01.
Article in English | MEDLINE | ID: mdl-31147394

ABSTRACT

Cell cycle progression is intimately linked to cell fate commitment during development. In addition, adult stem cells show specific proliferative behaviors compared to progenitors. Exploring cell cycle dynamics and regulation is therefore of utmost importance, but constitutes a great challenge in vivo. Here we provide a protocol for evaluating in vivo the length of all cell cycle phases of neural stem and progenitor cells in the post-embryonic Xenopus retina. These cells are localized in the ciliary marginal zone (CMZ), a peripheral region of the retina that sustains continuous neurogenesis throughout the animal's life. The CMZ bears two tremendous advantages for cell cycle kinetics analyses. First, this region, where proliferative cells are sequestered, can be easily delineated. Second, the spatial organization of the CMZ mirrors the temporal sequence of retinal development, allowing for topological distinction between retinal stem cells (residing in the most peripheral margin), and amplifying progenitors (located more centrally). We describe herein how to determine CMZ cell cycle parameters using a combination of (i) a cumulative labeling assay, (ii) the percentage of labeled mitosis calculation, and (iii) the mitotic index measurement. Taken together, these techniques allow us to estimate total cell cycle length (TC) as well as the duration of all cell cycle phases (TS/G2/M/G1). Although the method presented here was adapted to the particular system of the CMZ, it should be applicable to other tissues and developmental stages as well.


Subject(s)
Cell Cycle , Cytological Techniques , Neural Stem Cells/cytology , Retina/cytology , Xenopus laevis/metabolism , Animals , Cell Nucleus/metabolism , Kinetics , Larva/cytology , Staining and Labeling , Tissue Fixation
13.
Cell Rep ; 27(6): 1712-1725.e6, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31067458

ABSTRACT

Contrasting with fish or amphibian, retinal regeneration from Müller glia is largely limited in mammals. In our quest toward the identification of molecular cues that may boost their stemness potential, we investigated the involvement of the Hippo pathway effector YAP (Yes-associated protein), which is upregulated in Müller cells following retinal injury. Conditional Yap deletion in mouse Müller cells prevents cell-cycle gene upregulation that normally accompanies reactive gliosis upon photoreceptor cell death. We further show that, in Xenopus, a species endowed with efficient regenerative capacity, YAP is required for their injury-dependent proliferative response. In the mouse retina, where Müller cells do not spontaneously proliferate, YAP overactivation is sufficient to induce their reprogramming into highly proliferative cells. Overall, we unravel a pivotal role for YAP in tuning Müller cell proliferative response to injury and highlight a YAP-EGFR (epidermal growth factor receptor) axis by which Müller cells exit their quiescence state, a critical step toward regeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle , Ependymoglial Cells/pathology , Neuroglia/pathology , Retinal Degeneration/pathology , Trans-Activators/metabolism , Xenopus Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Proliferation , Ependymoglial Cells/metabolism , Epidermal Growth Factor/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Signal Transduction , Transcription, Genetic , Up-Regulation/genetics , Xenopus laevis , YAP-Signaling Proteins
14.
Methods Mol Biol ; 1865: 133-146, 2018.
Article in English | MEDLINE | ID: mdl-30151764

ABSTRACT

Xenopus is an attractive model system for regeneration studies, as it exhibits an extraordinary regenerative capacity compared to mammals. It is commonly used to study body part regeneration following amputation, for instance of the limb, the tail, or the retina. Models with more subtle injuries are also needed for human degenerative disease modeling, allowing for the study of stem cell recruitment for the regeneration of a given cellular subtype. We present here a model to ablate photoreceptor cells in the Xenopus retina. This method is based on the nitroreductase/metronidazole (NTR/MTZ) system, a combination of chemical and genetic tools, allowing for the conditional ablation of targeted cells. This type of approach establishes Xenopus as a powerful model to study cellular regeneration and stem cell regulation.


Subject(s)
Metronidazole/pharmacology , Nitroreductases/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Xenopus/metabolism , Animals , Animals, Genetically Modified , Green Fluorescent Proteins/metabolism , Photoreceptor Cells, Vertebrate/drug effects , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/metabolism , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/genetics , Transgenes
15.
PLoS One ; 13(4): e0193606, 2018.
Article in English | MEDLINE | ID: mdl-29672592

ABSTRACT

Wnt proteins form a family of highly conserved secreted molecules that are critical mediators of cell-cell signaling during embryogenesis. Partial data on Wnt activity in different tissues and at different stages have been reported in frog embryos. Our objective here is to provide a coherent and detailed description of Wnt activity throughout embryo development. Using a transgenic Xenopus tropicalis line carrying a Wnt-responsive reporter sequence, we depict the spatial and temporal dynamics of canonical Wnt activity during embryogenesis. We provide a comprehensive series of in situ hybridization in whole-mount embryos and in cross-sections, from gastrula to tadpole stages, with special focus on neural tube, retina and neural crest cell development. This collection of patterns will thus constitute a valuable resource for developmental biologists to picture the dynamics of Wnt activity during development.


Subject(s)
Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , Gastrula/metabolism , In Situ Hybridization , Neural Crest/metabolism , Neural Tube/metabolism , Wnt Proteins/genetics , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/genetics
16.
Neural Dev ; 12(1): 16, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28863786

ABSTRACT

BACKGROUND: Amacrine interneurons that modulate synaptic plasticity between bipolar and ganglion cells constitute the most diverse cell type in the retina. Most are inhibitory neurons using either GABA or glycine as neurotransmitters. Although several transcription factors involved in amacrine cell fate determination have been identified, mechanisms underlying amacrine cell subtype specification remain to be further understood. The Prdm13 histone methyltransferase encoding gene is a target of the transcription factor Ptf1a, an essential regulator of inhibitory neuron cell fate in the retina. Here, we have deepened our knowledge on its interaction with Ptf1a and investigated its role in amacrine cell subtype determination in the developing Xenopus retina. METHODS: We performed prdm13 gain and loss of function in Xenopus and assessed the impact on retinal cell fate determination using RT-qPCR, in situ hybridization and immunohistochemistry. RESULTS: We found that prdm13 in the amphibian Xenopus is expressed in few retinal progenitors and in about 40% of mature amacrine cells, predominantly in glycinergic ones. Clonal analysis in the retina reveals that prdm13 overexpression favours amacrine cell fate determination, with a bias towards glycinergic cells. Conversely, knockdown of prdm13 specifically inhibits glycinergic amacrine cell genesis. We also showed that, as in the neural tube, prdm13 is subjected to a negative autoregulation in the retina. Our data suggest that this is likely due to its ability to repress the expression of its inducer, ptf1a. CONCLUSIONS: Our results demonstrate that Prdm13, downstream of Ptf1a, acts as an important regulator of glycinergic amacrine subtype specification in the Xenopus retina. We also reveal that Prdm13 regulates ptf1a expression through a negative feedback loop.


Subject(s)
Amacrine Cells/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neurogenesis/physiology , Retina/embryology , Xenopus Proteins/metabolism , Amacrine Cells/cytology , Animals , Feedback, Physiological/physiology , Gene Expression Regulation, Developmental , Glycine/metabolism , Retina/metabolism , Xenopus laevis
17.
J Cell Biol ; 216(6): 1849-1864, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28495838

ABSTRACT

Usher syndrome type 1 (USH1) causes combined hearing and sight defects, but how mutations in USH1 genes lead to retinal dystrophy in patients remains elusive. The USH1 protein complex is associated with calyceal processes, which are microvilli of unknown function surrounding the base of the photoreceptor outer segment. We show that in Xenopus tropicalis, these processes are connected to the outer-segment membrane by links composed of protocadherin-15 (USH1F protein). Protocadherin-15 deficiency, obtained by a knockdown approach, leads to impaired photoreceptor function and abnormally shaped photoreceptor outer segments. Rod basal outer disks displayed excessive outgrowth, and cone outer segments were curved, with lamellae of heterogeneous sizes, defects also observed upon knockdown of Cdh23, encoding cadherin-23 (USH1D protein). The calyceal processes were virtually absent in cones and displayed markedly reduced F-actin content in rods, suggesting that protocadherin-15-containing links are essential for their development and/or maintenance. We propose that calyceal processes, together with their associated links, control the sizing of rod disks and cone lamellae throughout their daily renewal.


Subject(s)
Cadherins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Rod Cell Outer Segment/metabolism , Usher Syndromes/metabolism , Xenopus Proteins/metabolism , Xenopus/metabolism , Actin Cytoskeleton/metabolism , Animals , Cadherins/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Larva/genetics , Larva/metabolism , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Rod Cell Outer Segment/ultrastructure , Usher Syndromes/genetics , Usher Syndromes/pathology , Xenopus/embryology , Xenopus/genetics , Xenopus Proteins/genetics
18.
Glia ; 65(8): 1333-1349, 2017 08.
Article in English | MEDLINE | ID: mdl-28548249

ABSTRACT

A striking aspect of tissue regeneration is its uneven distribution among different animal classes, both in terms of modalities and efficiency. The retina does not escape the rule, exhibiting extraordinary self-repair properties in anamniote species but extremely limited ones in mammals. Among cellular sources prone to contribute to retinal regeneration are Müller glial cells, which in teleosts have been known for a decade to re-acquire a stem/progenitor state and regenerate retinal neurons following injury. As their regenerative potential was hitherto unexplored in amphibians, we tackled this issue using two Xenopus retinal injury paradigms we implemented: a mechanical needle poke injury and a transgenic model allowing for conditional photoreceptor cell ablation. These models revealed that Müller cells are indeed able to proliferate and replace lost cells following damage/degeneration in the retina. Interestingly, the extent of cell cycle re-entry appears dependent on the age of the animal, with a refractory period in early tadpole stages. Our findings pave the way for future studies aimed at identifying the molecular cues that either sustain or constrain the recruitment of Müller glia, an issue of utmost importance to set up therapeutic strategies for eye regenerative medicine.


Subject(s)
Ependymoglial Cells/pathology , Ependymoglial Cells/physiology , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Age Factors , Animals , Animals, Genetically Modified , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Proliferation , Diamines/pharmacology , Disease Models, Animal , Ependymoglial Cells/metabolism , Gene Expression Regulation/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Metronidazole/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Radiation-Sensitizing Agents/pharmacology , Regeneration/physiology , Rhodopsin/genetics , Rhodopsin/metabolism , SOX9 Transcription Factor/metabolism , Thiazoles/pharmacology , Urea/analogs & derivatives , Urea/metabolism , Xenopus laevis
19.
Invest Ophthalmol Vis Sci ; 58(4): 1941-1953, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28384715

ABSTRACT

Purpose: During retinal degeneration, Müller glia cells respond to photoreceptor loss by undergoing reactive gliosis, with both detrimental and beneficial effects. Increasing our knowledge of the complex molecular response of Müller cells to retinal degeneration is thus essential for the development of new therapeutic strategies. The purpose of this work was to identify new factors involved in Müller cell response to photoreceptor cell death. Methods: Whole transcriptome sequencing was performed from wild-type and degenerating rd10 mouse retinas at P30. The changes in mRNA abundance for several differentially expressed genes were assessed by quantitative RT-PCR (RT-qPCR). Protein expression level and retinal cellular localization were determined by western blot and immunohistochemistry, respectively. Results: Pathway-level analysis from whole transcriptomic data revealed the Hippo/YAP pathway as one of the main signaling pathways altered in response to photoreceptor degeneration in rd10 retinas. We found that downstream effectors of this pathway, YAP and TEAD1, are specifically expressed in Müller cells and that their expression, at both the mRNA and protein levels, is increased in rd10 reactive Müller glia after the onset of photoreceptor degeneration. The expression of Ctgf and Cyr61, two target genes of the transcriptional YAP/TEAD complex, is also upregulated following photoreceptor loss. Conclusions: This work reveals for the first time that YAP and TEAD1, key downstream effectors of the Hippo pathway, are specifically expressed in Müller cells. We also uncovered a deregulation of the expression and activity of Hippo/YAP pathway components in reactive Müller cells under pathologic conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/genetics , Ependymoglial Cells/metabolism , Gene Expression Regulation , Phosphoproteins/genetics , Photoreceptor Cells/metabolism , RNA, Messenger/genetics , Retinal Degeneration/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Blotting, Western , Cell Cycle Proteins , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Ependymoglial Cells/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nuclear Proteins , Phosphoproteins/biosynthesis , Photoreceptor Cells/pathology , Real-Time Polymerase Chain Reaction , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/biosynthesis , YAP-Signaling Proteins
20.
Curr Pathobiol Rep ; 5(1): 67-78, 2017.
Article in English | MEDLINE | ID: mdl-28255526

ABSTRACT

PURPOSE OF REVIEW: Retinal degenerative diseases have immense socio-economic impact. Studying animal models that recapitulate human eye pathologies aids in understanding the pathogenesis of diseases and allows for the discovery of novel therapeutic strategies. Some non-mammalian species are known to have remarkable regenerative abilities and may provide the basis to develop strategies to stimulate self-repair in patients suffering from these retinal diseases. RECENT FINDINGS: Non-mammalian organisms, such as zebrafish and Xenopus, have become attractive model systems to study retinal diseases. Additionally, many fish and amphibian models of retinal cell ablation and cell lineage analysis have been developed to study regeneration. These investigations highlighted several cellular sources for retinal repair in different fish and amphibian species. Moreover, major differences in repair mechanisms have been reported in these animal models. SUMMARY: This review aims to emphasize first on the importance of zebrafish and Xenopus models in studying the pathogenesis of retinal diseases and, second, on the different modes of regeneration processes in these model organisms.

SELECTION OF CITATIONS
SEARCH DETAIL