Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Development ; 151(2)2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38108453

RESUMEN

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.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Animales , Xenopus laevis/metabolismo , Especies Reactivas de Oxígeno , Proliferación Celular , Proteínas Hedgehog , Retina/metabolismo , Células Madre Adultas/metabolismo , NADPH Oxidasas/genética , Vía de Señalización Wnt
2.
Glia ; 72(4): 759-776, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38225726

RESUMEN

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.


Asunto(s)
Cobalto , Degeneración Retiniana , Animales , Xenopus laevis/fisiología , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/metabolismo , Retina , Regeneración/fisiología , Proliferación Celular , Neuroglía/metabolismo
3.
Proc Natl Acad Sci U S A ; 113(36): 10103-8, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27555585

RESUMEN

Pax6 is a key transcription factor involved in eye, brain, and pancreas development. Although pax6 is expressed in the whole prospective retinal field, subsequently its expression becomes restricted to the optic cup by reciprocal transcriptional repression of pax6 and pax2 However, it remains unclear how Pax6 protein is removed from the eyestalk territory on time. Here, we report that Mid1, a member of the RBCC/TRIM E3 ligase family, which was first identified in patients with the X-chromosome-linked Opitz BBB/G (OS) syndrome, interacts with Pax6. We found that the forming eyestalk is a major domain of mid1 expression, controlled by the morphogen Sonic hedgehog (Shh). Here, Mid1 regulates the ubiquitination and proteasomal degradation of Pax6 protein. Accordantly, when Mid1 levels are knocked down, Pax6 expression is expanded and eyes are enlarged. Our findings indicate that remaining or misaddressed Pax6 protein is cleared from the eyestalk region to properly set the border between the eyestalk territory and the retina via Mid1. Thus, we identified a posttranslational mechanism, regulated by Sonic hedgehog, which is important to suppress Pax6 activity and thus breaks pax6 autoregulation at defined steps during the formation of the visual system.


Asunto(s)
Proteínas del Ojo/genética , Ojo/metabolismo , Proteínas Hedgehog/genética , Factor de Transcripción PAX6/genética , Ubiquitina-Proteína Ligasas/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Animales , Embrión no Mamífero , Ojo/crecimiento & desarrollo , Proteínas del Ojo/metabolismo , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Células HeLa , Proteínas Hedgehog/metabolismo , Humanos , Tamaño de los Órganos , Organogénesis/genética , Factor de Transcripción PAX6/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Transducción de Señal , Factores de Tiempo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas de Xenopus/metabolismo , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/metabolismo
4.
Glia ; 65(8): 1333-1349, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28548249

RESUMEN

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.


Asunto(s)
Células Ependimogliales/patología , Células Ependimogliales/fisiología , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Factores de Edad , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Proliferación Celular , Diaminas/farmacología , Modelos Animales de Enfermedad , Células Ependimogliales/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Metronidazol/farmacología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Regeneración/fisiología , Rodopsina/genética , Rodopsina/metabolismo , Factor de Transcripción SOX9/metabolismo , Tiazoles/farmacología , Urea/análogos & derivados , Urea/metabolismo , Xenopus laevis
5.
Dev Dyn ; 245(7): 727-38, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26661417

RESUMEN

Retinal dystrophies are a major cause of blindness for which there are currently no curative treatments. Transplantation of stem cell-derived neuronal progenitors to replace lost cells has been widely investigated as a therapeutic option. Another promising strategy would be to trigger self-repair mechanisms in patients, through the recruitment of endogenous cells with stemness properties. Accumulating evidence in the past 15 year0s has revealed that several retinal cell types possess neurogenic potential, thus opening new avenues for regenerative medicine. Among them, Müller glial cells have been shown to be able to undergo a reprogramming process to re-acquire a stem/progenitor state, allowing them to proliferate and generate new neurons for repair following retinal damages. Although Müller cell-dependent spontaneous regeneration is remarkable in some species such as the fish, it is extremely limited and ineffective in mammals. Understanding the cellular events and molecular mechanisms underlying Müller cell activities in species endowed with regenerative capacities could provide knowledge to unlock the restricted potential of their mammalian counterparts. In this context, the present review provides an overview of Müller cell responses to injury across vertebrate model systems and summarizes recent advances in this rapidly evolving field. Developmental Dynamics 245:727-738, 2016. © 2015 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc.


Asunto(s)
Células Ependimogliales/citología , Células Ependimogliales/fisiología , Retina/citología , Animales , Células Ependimogliales/metabolismo , Humanos , Regeneración/genética , Regeneración/fisiología , Retina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Células Madre/fisiología
6.
Dev Biol ; 386(2): 340-57, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24370451

RESUMEN

The basic helix-loop-helix (bHLH) transcriptional activator Ptf1a determines inhibitory GABAergic over excitatory glutamatergic neuronal cell fate in progenitors of the vertebrate dorsal spinal cord, cerebellum and retina. In an in situ hybridization expression survey of PR domain containing genes encoding putative chromatin-remodeling zinc finger transcription factors in Xenopus embryos, we identified Prdm13 as a histone methyltransferase belonging to the Ptf1a synexpression group. Gain and loss of Ptf1a function analyses in both frog and mice indicates that Prdm13 is positively regulated by Ptf1a and likely constitutes a direct transcriptional target. We also showed that this regulation requires the formation of the Ptf1a-Rbp-j complex. Prdm13 knockdown in Xenopus embryos and in Ptf1a overexpressing ectodermal explants lead to an upregulation of Tlx3/Hox11L2, which specifies a glutamatergic lineage and a reduction of the GABAergic neuronal marker Pax2. It also leads to an upregulation of Prdm13 transcription, suggesting an autonegative regulation. Conversely, in animal caps, Prdm13 blocks the ability of the bHLH factor Neurog2 to activate Tlx3. Additional gain of function experiments in the chick neural tube confirm that Prdm13 suppresses Tlx3(+)/glutamatergic and induces Pax2(+)/GABAergic neuronal fate. Thus, Prdm13 is a novel crucial component of the Ptf1a regulatory pathway that, by modulating the transcriptional activity of bHLH factors such as Neurog2, controls the balance between GABAergic and glutamatergic neuronal fate in the dorsal and caudal part of the vertebrate neural tube.


Asunto(s)
Diferenciación Celular/fisiología , Neuronas GABAérgicas/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Tubo Neural/embriología , Proteínas de Xenopus/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Embrión de Pollo , Cartilla de ADN/genética , Electroporación , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Ratones , Tubo Neural/citología , Factor de Transcripción PAX2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Xenopus/genética , Xenopus laevis
7.
Development ; 139(19): 3499-509, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22899850

RESUMEN

Continuous neurogenesis in the adult nervous system requires a delicate balance between proliferation and differentiation. Although Wnt/ß-catenin and Hedgehog signalling pathways are thought to share a mitogenic function in adult neural stem/progenitor cells, it remains unclear how they interact in this process. Adult amphibians produce retinal neurons from a pool of neural stem cells localised in the ciliary marginal zone (CMZ). Surprisingly, we found that perturbations of the Wnt and Hedgehog pathways result in opposite proliferative outcomes of neural stem/progenitor cells in the CMZ. Additionally, our study revealed that Wnt and Hedgehog morphogens are produced in mutually exclusive territories of the post-embryonic retina. Using genetic and pharmacological tools, we found that the Wnt and Hedgehog pathways exhibit reciprocal inhibition. Our data suggest that Sfrp-1 and Gli3 contribute to this negative cross-regulation. Altogether, our results reveal an unexpected antagonistic interplay of Wnt and Hedgehog signals that may tightly regulate the extent of neural stem/progenitor cell proliferation in the Xenopus retina.


Asunto(s)
Proliferación Celular , Proteínas Hedgehog/fisiología , Retina/embriología , Retina/crecimiento & desarrollo , Vía de Señalización Wnt/fisiología , Animales , Animales Modificados Genéticamente , Proliferación Celular/efectos de los fármacos , Antagonismo de Drogas , Embrión no Mamífero , Inhibidores Enzimáticos/farmacología , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Indoles/farmacología , Modelos Biológicos , Organogénesis/efectos de los fármacos , Organogénesis/genética , Organogénesis/fisiología , Oximas/farmacología , Receptor Cross-Talk/efectos de los fármacos , Receptor Cross-Talk/fisiología , Retina/efectos de los fármacos , Retina/metabolismo , Teratógenos/farmacología , Alcaloides de Veratrum/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Xenopus laevis/embriología
8.
Cell Death Discov ; 10(1): 48, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38272861

RESUMEN

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.

9.
Stem Cells ; 30(12): 2784-95, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22969013

RESUMEN

The retina of fish and amphibian contains genuine neural stem cells located at the most peripheral edge of the ciliary marginal zone (CMZ). However, their cell-of-origin as well as the mechanisms that sustain their maintenance during development are presently unknown. We identified Hes4 (previously named XHairy2), a gene encoding a bHLH-O transcriptional repressor, as a stem cell-specific marker of the Xenopus CMZ that is positively regulated by the canonical Wnt pathway and negatively by Hedgehog signaling. We found that during retinogenesis, Hes4 labels a small territory, located first at the pigmented epithelium (RPE)/neural retina (NR) border and later in the retinal margin, that likely gives rise to adult retinal stem cells. We next addressed whether Hes4 might impart this cell subpopulation with retinal stem cell features: inhibited RPE or NR differentiation programs, continuous proliferation, and slow cell cycle speed. We could indeed show that Hes4 overexpression cell autonomously prevents retinal precursor cells from commitment toward retinal fates and maintains them in a proliferative state. Besides, our data highlight for the first time that Hes4 may also constitute a crucial regulator of cell cycle kinetics. Hes4 gain of function indeed significantly slows down cell division, mainly through the lengthening of G1 phase. As a whole, we propose that Hes4 maintains particular stemness features in a cellular cohort dedicated to constitute the adult retinal stem cell pool, by keeping it in an undifferentiated and slowly proliferative state along embryonic retinogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Células-Madre Neurales/citología , Retina/citología , Retina/embriología , Proteínas de Xenopus/biosíntesis , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Inmunohistoquímica , Masculino , Células-Madre Neurales/metabolismo , Retina/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/embriología , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal , Vía de Señalización Wnt , Proteínas de Xenopus/genética , Xenopus laevis
10.
J Vis Exp ; (200)2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37902314

RESUMEN

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.


Asunto(s)
Retina , Retinitis Pigmentosa , Animales , Xenopus laevis , Larva , Retina/metabolismo , Animales Modificados Genéticamente , Retinitis Pigmentosa/metabolismo , Mamíferos
11.
JCI Insight ; 8(21)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37768732

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas , Degeneración Retiniana , Retinitis Pigmentosa , Humanos , Retina , Retinitis Pigmentosa/genética , Degeneración Retiniana/genética , Epitelio Pigmentado de la Retina , Proteínas Adaptadoras Transductoras de Señales
12.
Cells ; 11(18)2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36139472

RESUMEN

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.


Asunto(s)
Degeneración Retiniana , Células Ependimogliales , Glucosa/farmacología , Glucógeno Sintasa Quinasa 3/farmacología , Humanos , Retina , Degeneración Retiniana/tratamiento farmacológico
13.
Cells ; 11(5)2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35269429

RESUMEN

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.


Asunto(s)
Retinitis Pigmentosa , Rodopsina , Animales , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Células Ependimogliales/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
14.
Elife ; 112022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35838349

RESUMEN

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.


Asunto(s)
Origen de Réplica , Proteínas de Unión a Telómeros , Animales , Replicación del ADN , Momento de Replicación del ADN , Fase S/genética , Proteínas de Unión a Telómeros/genética , Xenopus laevis/genética , Xenopus laevis/metabolismo
15.
Curr Biol ; 18(15): 1156-61, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18674909

RESUMEN

The nerve cell is a eumetazoan (cnidarians and bilaterians) synapomorphy [1]; this cell type is absent in sponges, a more ancient phyletic lineage. Here, we demonstrate that despite lacking neurons, the sponge Amphimedon queenslandica expresses the Notch-Delta signaling system and a proneural basic helix loop helix (bHLH) gene in a manner that resembles the conserved molecular mechanisms of primary neurogenesis in bilaterians. During Amphimedon development, a field of subepithelial cells expresses the Notch receptor, its ligand Delta, and a sponge bHLH gene, AmqbHLH1. Cells that migrate out of this field express AmqDelta1 and give rise to putative sensory cells that populate the larval epithelium. Phylogenetic analysis suggests that AmqbHLH1 is descendent from a single ancestral bHLH gene that later duplicated to produce the atonal/neurogenin-related bHLH gene families, which include most bilaterian proneural genes [2]. By way of functional studies in Xenopus and Drosophila, we demonstrate that AmqbHLH1 has a strong proneural activity in both species with properties displayed by both neurogenin and atonal genes. From these results, we infer that the bilaterian neurogenic circuit, comprising proneural atonal-related bHLH genes coupled with Notch-Delta signaling, was functional in the very first metazoans and was used to generate an ancient sensory cell type.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Filogenia , Poríferos/genética , Transducción de Señal , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Secuencia Conservada , Drosophila , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Duplicación de Gen , Secuencias Hélice-Asa-Hélice , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Poríferos/crecimiento & desarrollo , Poríferos/metabolismo , Receptores Notch/metabolismo , Xenopus
16.
eNeuro ; 8(5)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34518365

RESUMEN

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.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Células Ganglionares de la Retina , Animales , Axones , Glucógeno Sintasa Quinasa 3/genética , Ratones , Retina
17.
Dev Biol ; 327(2): 352-65, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19135436

RESUMEN

Members of the Rx (retinal homeobox) gene family play vital roles during eye development. In Xenopus, as in most other vertebrates, two Rx-type genes have been described. While Rx1 deficiency led to loss of optic vesicles and impaired the proper development of ventral forebrain structures, a recently isolated second Rx-gene, Rx-L, seems to function in late retinogenesis. Here, we report that the specific suppression of Xenopus Rx-L function impaired the formation of the photoreceptor layer and reduced the expression of photoreceptor specific genes. Overexpression of Xenopus Rx-L induced ectopic expression of photoreceptor specific genes, but did only marginally promote the proliferation of retinal progenitor cells. Targeted overexpression of Rx-L in developing retinoblasts in vivo led to an increased fraction of photoreceptor cells at the expense of amacrine and bipolar cells and revealed that Rx-L acts as a transcription activator. A phylogenic analysis of all reported Rx-type genes revealed that they could be grouped into three categories, including an "invertebrate Rx" group, a "classical vertebrate Rx" group, and a "vertebrate Qrx/Rx-L" group. Taken together, Rx-L, unlike Rx1, is required for the determination of retinal cell types, especially photoreceptors, rather than for proliferation of retinal progenitors.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Fotorreceptoras/fisiología , Proteínas de Xenopus/metabolismo , Xenopus laevis , Secuencia de Aminoácidos , Animales , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Datos de Secuencia Molecular , Células Fotorreceptoras/citología , Retina/citología , Retina/embriología , Rodopsina/genética , Rodopsina/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas de Xenopus/genética , Xenopus laevis/anatomía & histología , Xenopus laevis/embriología
18.
Curr Opin Genet Dev ; 64: 52-59, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32619816

RESUMEN

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.


Asunto(s)
Reprogramación Celular , Células Ependimogliales/citología , Regeneración Nerviosa , Neuroglía/citología , Retina/fisiología , Cicatrización de Heridas , Animales , Proliferación Celular , Mamíferos , Retina/lesiones
19.
Cell Death Dis ; 11(8): 631, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32801350

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Distrofia del Cono/patología , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Haploinsuficiencia/genética , Animales , Animales Recién Nacidos , Proteínas Portadoras/metabolismo , Ciclo Celular , Proliferación Celular , Distrofia del Cono/genética , Receptores ErbB/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Homeostasis , Ratones Noqueados , Modelos Biológicos , Opsinas/metabolismo , Fenotipo , Retina/patología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Células Madre/metabolismo , Transactivadores/metabolismo , Proteínas Señalizadoras YAP
20.
Stem Cells ; 26(8): 2063-74, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18556512

RESUMEN

Vertebrate retinal stem cells, which reside quiescently within the ciliary margin, may offer a possibility for treatment of degenerative retinopathies. The highly proliferative retinal precursor cells in Xenopus eyes are confined to the most peripheral region, called the ciliary marginal zone (CMZ). Although the canonical Wnt pathway has been implicated in the developing retina of different species, little is known about its involvement in postembryonic retinas. Using a green fluorescent protein-based Wnt-responsive reporter, we show that in transgenic Xenopus tadpoles, the canonical Wnt signaling is activated in the postembryonic CMZ. To further investigate the functional implications of this, we generated transgenic, hormone-inducible canonical Wnt pathway activating and repressing systems, which are directed to specifically intersect at the nuclear endpoint of transcriptional Wnt target gene activation. We found that postembryonic induction of the canonical Wnt pathway in transgenic retinas resulted in increased proliferation in the CMZ compartment. This is most likely due to delayed cell cycle exit, as inferred from a pulse-chase experiment on 5-bromo-2'-deoxyuridine-labeled retinal precursors. Conversely, repression of the canonical Wnt pathway inhibited proliferation of CMZ cells. Neither activation nor repression of the Wnt pathway affected the differentiated cells in the central retina. We conclude that even at postembryonic stages, the canonical Wnt signaling pathway continues to have a major function in promoting proliferation and maintaining retinal stem cells. These findings may contribute to the eventual design of vertebrate, stem cell-based retinal therapies. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Ojo/metabolismo , Retina/metabolismo , Células Madre/citología , Proteínas Wnt/metabolismo , Xenopus laevis/metabolismo , Animales , Animales Modificados Genéticamente , Proliferación Celular , Ojo/crecimiento & desarrollo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Modelos Genéticos , Retina/crecimiento & desarrollo , Transducción de Señal , Factores de Tiempo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA