Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3365, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664376

RESUMEN

Hedgehog (Hh) signaling relies on the primary cilium, a cell surface organelle that serves as a signaling hub for the cell. Using proximity labeling and quantitative proteomics, we identify Numb as a ciliary protein that positively regulates Hh signaling. Numb localizes to the ciliary pocket and acts as an endocytic adaptor to incorporate Ptch1 into clathrin-coated vesicles, thereby promoting Ptch1 exit from the cilium, a key step in Hh signaling activation. Numb loss impedes Sonic hedgehog (Shh)-induced Ptch1 exit from the cilium, resulting in reduced Hh signaling. Numb loss in spinal neural progenitors reduces Shh-induced differentiation into cell fates reliant on high Hh activity. Genetic ablation of Numb in the developing cerebellum impairs the proliferation of granule cell precursors, a Hh-dependent process, resulting in reduced cerebellar size. This study highlights Numb as a regulator of ciliary Ptch1 levels during Hh signal activation and demonstrates the key role of ciliary pocket-mediated endocytosis in cell signaling.


Asunto(s)
Cerebelo , Cilios , Proteínas Hedgehog , Proteínas del Tejido Nervioso , Receptor Patched-1 , Transducción de Señal , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Cilios/metabolismo , Animales , Receptor Patched-1/metabolismo , Receptor Patched-1/genética , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Cerebelo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Humanos , Endocitosis , Diferenciación Celular , Proliferación Celular , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Ratones Noqueados
2.
Cell Rep ; 43(4): 114005, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38551961

RESUMEN

The retina is exquisitely patterned, with neuronal somata positioned at regular intervals to completely sample the visual field. Here, we show that phosphatase and tensin homolog (Pten) controls starburst amacrine cell spacing by modulating vesicular trafficking of cell adhesion molecules and Wnt proteins. Single-cell transcriptomics and double-mutant analyses revealed that Pten and Down syndrome cell adhesion molecule Dscam) are co-expressed and function additively to pattern starburst amacrine cell mosaics. Mechanistically, Pten loss accelerates the endocytic trafficking of DSCAM, FAT3, and MEGF10 off the cell membrane and into endocytic vesicles in amacrine cells. Accordingly, the vesicular proteome, a molecular signature of the cell of origin, is enriched in exocytosis, vesicle-mediated transport, and receptor internalization proteins in Pten conditional knockout (PtencKO) retinas. Wnt signaling molecules are also enriched in PtencKO retinal vesicles, and the genetic or pharmacological disruption of Wnt signaling phenocopies amacrine cell patterning defects. Pten thus controls vesicular trafficking of cell adhesion and signaling molecules to establish retinal amacrine cell mosaics.


Asunto(s)
Células Amacrinas , Adhesión Celular , Endocitosis , Fosfohidrolasa PTEN , Retina , Vía de Señalización Wnt , Animales , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Retina/metabolismo , Ratones , Células Amacrinas/metabolismo , Ratones Noqueados , Transporte de Proteínas , Proteínas Wnt/metabolismo , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/genética
3.
Cell Rep ; 42(8): 112985, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37590135

RESUMEN

The balance of contralateral and ipsilateral retinogeniculate projections is critical for binocular vision, but the transcriptional programs regulating this process remain ill defined. Here we show that the Pou class homeobox protein POU3F1 is expressed in nascent mouse contralateral retinal ganglion cells (cRGCs) but not ipsilateral RGCs (iRGCs). Upon Pou3f1 inactivation, the proportion of cRGCs is reduced in favor of iRGCs, leading to abnormal projection ratios at the optic chiasm. Conversely, misexpression of Pou3f1 in progenitors increases the production of cRGCs. Using CUT&RUN and RNA sequencing in gain- and loss-of-function assays, we demonstrate that POU3F1 regulates expression of several key members of the cRGC gene regulatory network. Finally, we report that POU3F1 is sufficient to induce RGC-like cell production, even in late-stage retinal progenitors of Atoh7 knockout mice. This work uncovers POU3F1 as a regulator of the cRGC transcriptional program, opening possibilities for optic nerve regenerative therapies.

4.
Proc Natl Acad Sci U S A ; 120(19): e2122168120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126716

RESUMEN

Temporal identity factors are sufficient to reprogram developmental competence of neural progenitors and shift cell fate output, but whether they can also reprogram the identity of terminally differentiated cells is unknown. To address this question, we designed a conditional gene expression system that allows rapid screening of potential reprogramming factors in mouse retinal glial cells combined with genetic lineage tracing. Using this assay, we found that coexpression of the early temporal identity transcription factors Ikzf1 and Ikzf4 is sufficient to directly convert Müller glial (MG) cells into cells that translocate to the outer nuclear layer (ONL), where photoreceptor cells normally reside. We name these "induced ONL (iONL)" cells. Using genetic lineage tracing, histological, immunohistochemical, and single-cell transcriptome and multiome analyses, we show that expression of Ikzf1/4 in MG in vivo, without retinal injury, mostly generates iONL cells that share molecular characteristics with bipolar cells, although a fraction of them stain for Rxrg, a cone photoreceptor marker. Furthermore, we show that coexpression of Ikzf1 and Ikzf4 can reprogram mouse embryonic fibroblasts to induced neurons in culture by rapidly remodeling chromatin and activating a neuronal gene expression program. This work uncovers general neuronal reprogramming properties for temporal identity factors in terminally differentiated cells.


Asunto(s)
Fibroblastos , Retina , Animales , Ratones , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/fisiología , Factores de Transcripción/metabolismo , Diferenciación Celular/fisiología , Reprogramación Celular
5.
Semin Cell Dev Biol ; 142: 36-42, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35760728

RESUMEN

Understanding how retinal progenitor cells (RPCs) give rise to the variety of neural cell types of the retina has been a question of major interest over the last few decades. While environmental cues and transcription factor networks have been shown to control specific cell fate decisions, how RPCs alter fate output over time to control proper histogenesis remains poorly understood. In recent years, the identification of "temporal identity factors (TIFs)", which control RPC competence states to ensure that the right cell types are produced at the right time, has contributed to increasing our understanding of temporal patterning in the retina. Here, we review the different TIFs identified to date in the mammalian retina and discuss the underlying mechanisms by which they are thought to operate. We conclude by speculating on how identification of temporal patterning mechanisms might support the development of new therapeutic approaches against visual impairments.


Asunto(s)
Retina , Células Madre , Animales , Células Madre/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Diferenciación Celular , Mamíferos
6.
Development ; 150(2)2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36537580

RESUMEN

Temporal identity factors regulate competence of neural progenitors to generate specific cell types in a time-dependent manner, but how they operate remains poorly defined. In the developing mouse retina, the Ikaros zinc-finger transcription factor Ikzf1 regulates production of early-born cell types, except cone photoreceptors. In this study we show that, during early stages of retinal development, another Ikaros family protein, Ikzf4, functions redundantly with Ikzf1 to regulate cone photoreceptor production. Using CUT&RUN and functional assays, we show that Ikzf4 binds and represses genes involved in late-born rod photoreceptor specification, hence favoring cone production. At late stages, when Ikzf1 is no longer expressed in progenitors, we show that Ikzf4 re-localizes to target genes involved in gliogenesis and is required for Müller glia production. We report that Ikzf4 regulates Notch signaling genes and is sufficient to activate the Hes1 promoter through two Ikzf GGAA-binding motifs, suggesting a mechanism by which Ikzf4 may influence gliogenesis. These results uncover a combinatorial role for Ikaros family members during nervous system development and provide mechanistic insights on how they temporally regulate cell fate output.


Asunto(s)
Factor de Transcripción Ikaros , Retina , Ratones , Animales , Retina/metabolismo , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Diferenciación Celular/genética
7.
Sci Adv ; 8(42): eabm4295, 2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-36260685

RESUMEN

Accumulation of the microtubule-associated protein Tau is linked to neuronal cell death in tauopathies, but how intraneuronal Tau levels are regulated in health and disease remains unclear. Here, we show that conditional inactivation of the trafficking adaptor protein Numb in retinal ganglion cells (RGCs) increases Tau levels and leads to axonal blebbing, which is followed by neuronal cell loss in aged mice. In the TauP301S mouse model of tauopathy, conditional inactivation of Numb in RGCs and spinal motoneurons accelerates neurodegeneration, and loss of Numb in motoneurons also leads to precocious hindlimb paralysis. Conversely, overexpression of the long isoform of Numb (Numb-72) decreases intracellular Tau levels and reduces axonal blebbing in TauP301S RGCs, leading to improved electrical activity in cultured neurons and improves performance in a visually guided behavior test in vivo. These results uncover Numb as a key regulator of intracellular Tau levels and identify Numb-72 as a potential therapeutic factor for tauopathies.


Asunto(s)
Tauopatías , Ratones , Animales , Tauopatías/genética , Tauopatías/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Modelos Animales de Enfermedad , Células Ganglionares de la Retina/metabolismo , Axones/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo
8.
Sci Adv ; 8(36): eabh2868, 2022 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-36070393

RESUMEN

Many transcription factors regulating the production, survival, and function of photoreceptor cells have been identified, but little is known about transcriptional co-regulators in retinal health and disease. Here, we show that BCL6 co-repressor (BCOR), a Polycomb repressive complex 1 factor mutated in various cancers, is involved in photoreceptor degenerative diseases. Using proteomics and transcription assays, we report that BCOR interacts with the transcription factors CRX and OTX2 and reduces their ability to activate the promoters of photoreceptor-specific genes. CUT&RUN sequencing further shows that BCOR shares genome-wide binding profiles with CRX/OTX2, consistent with a general co-repression activity. We also identify missense mutations in human BCOR in five families that have no evidence of cancer but present severe early-onset X-linked retinal degeneration. Last, we show that the human BCOR mutants cause degeneration when expressed in the mouse retina and have enhanced repressive activity on OTX2. These results uncover a role for BCOR in photoreceptors in both health and disease.

9.
Curr Biol ; 31(21): R1434-R1437, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34752770

RESUMEN

Cell reprogramming is generally considered an artificially induced event. Excitingly, a new study shows that post-mitotic cell reprogramming occurs naturally in the developing fish retina, uncovering a mechanism involved in the generation of cell diversity.


Asunto(s)
Reprogramación Celular , Retina , Animales
10.
Sci Adv ; 7(11)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33712461

RESUMEN

Retinal ganglion cells (RGCs) relay visual information from the eye to the brain. RGCs are the first cell type generated during retinal neurogenesis. Loss of function of the transcription factor Atoh7, expressed in multipotent early neurogenic retinal progenitors leads to a selective and essentially complete loss of RGCs. Therefore, Atoh7 is considered essential for conferring competence on progenitors to generate RGCs. Despite the importance of Atoh7 in RGC specification, we find that inhibiting apoptosis in Atoh7-deficient mice by loss of function of Bax only modestly reduces RGC numbers. Single-cell RNA sequencing of Atoh7;Bax-deficient retinas shows that RGC differentiation is delayed but that the gene expression profile of RGC precursors is grossly normal. Atoh7;Bax-deficient RGCs eventually mature, fire action potentials, and incorporate into retinal circuitry but exhibit severe axonal guidance defects. This study reveals an essential role for Atoh7 in RGC survival and demonstrates Atoh7-dependent and Atoh7-independent mechanisms for RGC specification.

11.
Sci Rep ; 11(1): 3858, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594190

RESUMEN

Neural progenitor cells undergo identity transitions during development to ensure the generation different types of neurons and glia in the correct sequence and proportions. A number of temporal identity factors that control these transitions in progenitor competence have been identified, but the molecular mechanisms underlying their function remain unclear. Here, we asked how Casz1, the mammalian orthologue of Drosophila castor, regulates competence during retinal development. We show that Casz1 is required to control the transition between neurogenesis and gliogenesis. Using BioID proteomics, we reveal that Casz1 interacts with the nucleosome remodeling and deacetylase (NuRD) complex in retinal cells. Finally, we show that both the NuRD and the polycomb repressor complexes are required for Casz1 to promote the rod fate and suppress gliogenesis. As additional temporal identity factors have been found to interact with the NuRD complex in other contexts, we propose that these factors might act through this common biochemical process to regulate neurogenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Células-Madre Neurales/fisiología , Neurogénesis , Retina/embriología , Factores de Transcripción/metabolismo , Animales , Células Ependimogliales , Ratones , Ratones Noqueados , Proteínas del Grupo Polycomb/metabolismo , Retina/citología
12.
Development ; 147(18)2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32878923

RESUMEN

Multipotent retinal progenitor cells (RPCs) generate various cell types in a precise chronological order, but how exactly cone photoreceptor production is restricted to early stages remains unclear. Here, we show that the POU-homeodomain factors Pou2f1/Pou2f2, the homologs of Drosophila temporal identity factors nub/pdm2, regulate the timely production of cones in mice. Forcing sustained expression of Pou2f1 or Pou2f2 in RPCs expands the period of cone production, whereas misexpression in late-stage RPCs triggers ectopic cone production at the expense of late-born fates. Mechanistically, we report that Pou2f1 induces Pou2f2 expression, which binds to a POU motif in the promoter of the rod-inducing factor Nrl to repress its expression. Conversely, conditional inactivation of Pou2f2 in RPCs increases Nrl expression and reduces cone production. Finally, we provide evidence that Pou2f1 is part of a cross-regulatory cascade with the other temporal identity factors Ikzf1 and Casz1. These results uncover Pou2f1/2 as regulators of the temporal window for cone genesis and, given their widespread expression in the nervous system, raise the possibility of a general role in temporal patterning.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Proteínas del Ojo/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Animales , Drosophila/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Madre/metabolismo
13.
Neuron ; 102(6): 1157-1171.e5, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31054872

RESUMEN

During development, Shh attracts commissural axons toward the floor plate through a non-canonical, transcription-independent signaling pathway that requires the receptor Boc. Here, we find that Shh induces Boc internalization into early endosomes and that endocytosis is required for Shh-mediated growth-cone turning. Numb, an endocytic adaptor, binds to Boc and is required for Boc internalization, Shh-mediated growth-cone turning in vitro, and commissural axon guidance in vivo. Similar to Boc, Ptch1 is also internalized by Shh in a Numb-dependent manner; however, the binding of Shh to Ptch1 alone is not sufficient to induce Ptch1 internalization nor growth-cone turning. Therefore, the binding of Shh to Boc is required for Ptch1 internalization and growth-cone turning. Our data support a model where Boc endocytosis via Numb is required for Ptch1 internalization and Shh signaling in axon guidance. Thus, Boc acts as a Shh-dependent endocytic platform gating Ptch1 internalization and Shh signaling.


Asunto(s)
Orientación del Axón/genética , Endocitosis/genética , Conos de Crecimiento/metabolismo , Proteínas Hedgehog/metabolismo , Inmunoglobulina G/metabolismo , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Receptor Patched-1/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo
14.
Proc Natl Acad Sci U S A ; 115(34): E7987-E7996, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30072429

RESUMEN

Genome organization plays a fundamental role in the gene-expression programs of numerous cell types, but determinants of higher-order genome organization are poorly understood. In the developing mouse retina, rod photoreceptors represent a good model to study this question. They undergo a process called "chromatin inversion" during differentiation, in which, as opposed to classic nuclear organization, heterochromatin becomes localized to the center of the nucleus and euchromatin is restricted to the periphery. While previous studies showed that the lamin B receptor participates in this process, the molecular mechanisms regulating lamina function during differentiation remain elusive. Here, using conditional genetics, we show that the zinc finger transcription factor Casz1 is required to establish and maintain the inverted chromatin organization of rod photoreceptors and to safeguard their gene-expression profile and long-term survival. At the mechanistic level, we show that Casz1 interacts with the polycomb repressor complex in a splice variant-specific manner and that both are required to suppress the expression of the nuclear envelope intermediate filament lamin A/C in rods. Lamin A is in turn sufficient to regulate heterochromatin organization and nuclear position. Furthermore, we show that Casz1 is sufficient to expand and centralize the heterochromatin of fibroblasts, suggesting a general role for Casz1 in nuclear organization. Together, these data support a model in which Casz1 cooperates with polycomb to control rod genome organization, in part by silencing lamin A/C.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Heterocromatina/metabolismo , Lamina Tipo A/metabolismo , Modelos Biológicos , Proteínas del Grupo Polycomb/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Silenciador del Gen/fisiología , Heterocromatina/genética , Lamina Tipo A/genética , Ratones , Ratones Transgénicos , Proteínas del Grupo Polycomb/genética , Células Fotorreceptoras Retinianas Bastones/citología , Factores de Transcripción/genética
15.
Cell Rep ; 23(8): 2416-2428, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29791852

RESUMEN

Newborn neurons follow molecular cues to reach their final destination, but whether early life experience influences lamination remains largely unexplored. As light is among the first stimuli to reach the developing nervous system via intrinsically photosensitive retinal ganglion cells (ipRGCs), we asked whether ipRGCs could affect lamination in the developing mouse retina. We show here that ablation of ipRGCs causes cone photoreceptors to mislocalize at different apicobasal positions in the retina. This effect is partly mediated by light-evoked activity in ipRGCs, as dark rearing or silencing of ipRGCs leads a subset of cones to mislocalize. Furthermore, ablation of ipRGCs alters the cone transcriptome and decreases expression of the dopamine receptor D4, while injection of L-DOPA or D4 receptor agonist rescues the displaced cone phenotype observed in dark-reared animals. These results show that early light-mediated activity in ipRGCs influences neuronal lamination and identify ipRGC-elicited dopamine release as a mechanism influencing cone position.


Asunto(s)
Células Fotorreceptoras Retinianas Conos/metabolismo , Células Ganglionares de la Retina/metabolismo , Opsinas de Bastones/metabolismo , Animales , Dopamina/administración & dosificación , Dopamina/metabolismo , Luz , Fototransducción , Ratones Endogámicos C57BL , Transcripción Genética , Transcriptoma/genética
16.
Development ; 145(6)2018 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-29549107

RESUMEN

Ben Barres changed our view of glial cell function and impacted the lives of many people who interacted with him. Remembering an outstanding scientist and mentor.


Asunto(s)
Neurobiología , Neuroglía , Historia del Siglo XX , Humanos , Masculino , Estados Unidos
17.
Dev Dyn ; 247(1): 10-17, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28643368

RESUMEN

Recent studies reported the transfer of fluorescent labels between grafted and host cells after transplantation of photoreceptor precursor cells in the mouse retina. While clearly impacting the interpretation of transplantation studies in the retina, the potential impact of material transfer in other experimental paradigms using cell-specific labels remains uncertain. Here, we briefly review the evidence supporting material transfer in transplantation studies and discuss whether it might influence retinal cell lineage tracing experiments in developmental and regeneration studies. We also propose ways to control for the possible confounding occurrence of label exchange in such experiments. Developmental Dynamics 247:10-17, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Neuronas/citología , Retina/citología , Animales , Ratones
18.
Dev Cell ; 42(1): 3-5, 2017 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-28697330

RESUMEN

Specialized areas in the vertebrate retina are critical for high-acuity vision, yet the molecular mechanisms driving the development of high-acuity areas (HAAs) remain largely unknown. In Developmental Cell, da Silva and Cepko (2017) show that restricted degradation of retinoic acid and elevated FGF8 signaling give rise to the chick HAA.


Asunto(s)
Retina , Tretinoina , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal
19.
Elife ; 62017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28617242

RESUMEN

The visual system consists of two major subsystems, image-forming circuits that drive conscious vision and non-image-forming circuits for behaviors such as circadian photoentrainment. While historically considered non-overlapping, recent evidence has uncovered crosstalk between these subsystems. Here, we investigated shared developmental mechanisms. We revealed an unprecedented role for light in the maturation of the circadian clock and discovered that intrinsically photosensitive retinal ganglion cells (ipRGCs) are critical for this refinement process. In addition, ipRGCs regulate retinal waves independent of light, and developmental ablation of a subset of ipRGCs disrupts eye-specific segregation of retinogeniculate projections. Specifically, a subset of ipRGCs, comprising ~200 cells and which project intraretinally and to circadian centers in the brain, are sufficient to mediate both of these developmental processes. Thus, this subset of ipRGCs constitute a shared node in the neural networks that mediate light-dependent maturation of the circadian clock and light-independent refinement of retinogeniculate projections.


Asunto(s)
Relojes Circadianos , Luz , Retina/fisiología , Retina/efectos de la radiación , Células Ganglionares de la Retina/fisiología , Células Ganglionares de la Retina/efectos de la radiación , Vías Visuales/fisiología , Animales , Ratones , Ratones Noqueados
20.
Front Neural Circuits ; 11: 105, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29375321

RESUMEN

Retinal degenerative diseases, which lead to the death of rod and cone photoreceptor cells, are the leading cause of inherited vision loss worldwide. Induced pluripotent or embryonic stem cells (iPSCs/ESCs) have been proposed as a possible source of new photoreceptors to restore vision in these conditions. The proof of concept studies carried out in mouse models of retinal degeneration over the past decade have highlighted several limitations for cell replacement in the retina, such as the low efficiency of cone photoreceptor production from stem cell cultures and the poor integration of grafted cells in the host retina. Current protocols to generate photoreceptors from stem cells are largely based on the use of extracellular factors. Although these factors are essential to induce the retinal progenitor cell (RPC) fate from iPSCs/ESCs, developmental studies have shown that RPCs alter fate output as a function of time (i.e., their temporal identity) to generate the seven major classes of retinal cell types, rather than spatial position. Surprisingly, current stem cell differentiation protocols largely ignore the intrinsic temporal identity of dividing RPCs, which we argue likely explains the low efficiency of cone production in such cultures. In this article, we briefly review the mechanisms regulating temporal identity in RPCs and discuss how they could be exploited to improve cone photoreceptor production for cell replacement therapies.


Asunto(s)
Células-Madre Neurales/fisiología , Retina/fisiología , Animales , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Retina/citología , Retina/fisiopatología , Retina/cirugía , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/fisiología , Enfermedades de la Retina/fisiopatología , Enfermedades de la Retina/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA