Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(24)2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36520654

RESUMEN

Appropriate patterning of the retina during embryonic development is assumed to underlie the establishment of spatially localised specialisations that mediate the perception of specific visual features. For example, in zebrafish, an area involved in high acuity vision (HAA) is thought to be present in the ventro-temporal retina. Here, we show that the interplay of the transcription factor Rx3 with Fibroblast Growth Factor and Hedgehog signals initiates and restricts foxd1 expression to the prospective temporal retina, initiating naso-temporal regionalisation of the retina. Abrogation of Foxd1 results in the loss of temporal and expansion of nasal retinal character, and consequent absence of the HAA. These structural defects correlate with severe visual defects, as assessed in optokinetic and optomotor response assays. In contrast, optokinetic responses are unaffected in the opposite condition, in which nasal retinal character is lost at the expense of expanded temporal character. Our study indicates that the establishment of temporal retinal character during early retinal development is required for the specification of the HAA, and suggests a prominent role of the temporal retina in controlling specific visual functions.


Asunto(s)
Proteínas Hedgehog , Pez Cebra , Animales , Pez Cebra/genética , Proteínas Hedgehog/metabolismo , Estudios Prospectivos , Retina/metabolismo , Visión Ocular
2.
Development ; 143(7): 1087-98, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26893342

RESUMEN

Maintaining neurogenesis in growing tissues requires a tight balance between progenitor cell proliferation and differentiation. In the zebrafish retina, neuronal differentiation proceeds in two stages with embryonic retinal progenitor cells (RPCs) of the central retina accounting for the first rounds of differentiation, and stem cells from the ciliary marginal zone (CMZ) being responsible for late neurogenesis and growth of the eye. In this study, we analyse two mutants with small eyes that display defects during both early and late phases of retinal neurogenesis. These mutants carry lesions in gdf6a, a gene encoding a BMP family member previously implicated in dorsoventral patterning of the eye. We show that gdf6a mutant eyes exhibit expanded retinoic acid (RA) signalling and demonstrate that exogenous activation of this pathway in wild-type eyes inhibits retinal growth, generating small eyes with a reduced CMZ and fewer proliferating progenitors, similar to gdf6a mutants. We provide evidence that RA regulates the timing of RPC differentiation by promoting cell cycle exit. Furthermore, reducing RA signalling in gdf6a mutants re-establishes appropriate timing of embryonic retinal neurogenesis and restores putative stem and progenitor cell populations in the CMZ. Together, our results support a model in which dorsally expressed gdf6a limits RA pathway activity to control the transition from proliferation to differentiation in the growing eye.


Asunto(s)
Factor 6 de Diferenciación de Crecimiento/genética , Neurogénesis/genética , Retina/embriología , Tretinoina/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Ciclo Celular/genética , Proliferación Celular , Embrión no Mamífero/embriología , Neurogénesis/fisiología , Transducción de Señal/genética , Células Madre/citología
3.
Development ; 142(22): 3933-42, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26428010

RESUMEN

The earliest known determinants of retinal nasotemporal identity are the transcriptional regulators Foxg1, which is expressed in the prospective nasal optic vesicle, and Foxd1, which is expressed in the prospective temporal optic vesicle. Previous work has shown that, in zebrafish, Fgf signals from the dorsal forebrain and olfactory primordia are required to specify nasal identity in the dorsal, prospective nasal, optic vesicle. Here, we show that Hh signalling from the ventral forebrain is required for specification of temporal identity in the ventral optic vesicle and is sufficient to induce temporal character when activated in the prospective nasal retina. Consequently, the evaginating optic vesicles become partitioned into prospective nasal and temporal domains by the opposing actions of Fgfs and Shh emanating from dorsal and ventral domains of the forebrain primordium. In absence of Fgf activity, foxd1 expression is established irrespective of levels of Hh signalling, indicating that the role of Shh in promoting foxd1 expression is only required in the presence of Fgf activity. Once the spatially complementary expression of foxd1 and foxg1 is established, the boundary between expression domains is maintained by mutual repression between Foxd1 and Foxg1.


Asunto(s)
Tipificación del Cuerpo/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Retina/embriología , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Carbocianinas , Factores de Transcripción Forkhead , Procesamiento de Imagen Asistido por Computador , Microscopía Confocal , Prosencéfalo/metabolismo
4.
Development ; 142(17): 3021-32, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26209646

RESUMEN

The optic vesicle comprises a pool of bi-potential progenitor cells from which the retinal pigment epithelium (RPE) and neural retina fates segregate during ocular morphogenesis. Several transcription factors and signaling pathways have been shown to be important for RPE maintenance and differentiation, but an understanding of the initial fate specification and determination of this ocular cell type is lacking. We show that Yap/Taz-Tead activity is necessary and sufficient for optic vesicle progenitors to adopt RPE identity in zebrafish. A Tead-responsive transgene is expressed within the domain of the optic cup from which RPE arises, and Yap immunoreactivity localizes to the nuclei of prospective RPE cells. yap (yap1) mutants lack a subset of RPE cells and/or exhibit coloboma. Loss of RPE in yap mutants is exacerbated in combination with taz (wwtr1) mutant alleles such that, when Yap and Taz are both absent, optic vesicle progenitor cells completely lose their ability to form RPE. The mechanism of Yap-dependent RPE cell type determination is reliant on both nuclear localization of Yap and interaction with a Tead co-factor. In contrast to loss of Yap and Taz, overexpression of either protein within optic vesicle progenitors leads to ectopic pigmentation in a dosage-dependent manner. Overall, this study identifies Yap and Taz as key early regulators of RPE genesis and provides a mechanistic framework for understanding the congenital ocular defects of Sveinsson's chorioretinal atrophy and congenital retinal coloboma.


Asunto(s)
Linaje de la Célula , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Epitelio Pigmentado de la Retina/citología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Alelos , Animales , Apoptosis/genética , Núcleo Celular/metabolismo , Proliferación Celular , Coloboma/patología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Morfogénesis/genética , Mutación , Fenotipo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Epitelio Pigmentado de la Retina/trasplante , Transducción de Señal/genética , Factores de Transcripción de Dominio TEA , Transactivadores/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Transgenes , Regulación hacia Arriba , Proteínas Señalizadoras YAP , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
Proc Natl Acad Sci U S A ; 108(21): 8698-703, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555593

RESUMEN

Retinoic acid receptor (RAR) signaling is required for morphogenesis of the ventral optic cup and closure of the choroid fissure, but the mechanisms by which this pathway regulates ventral eye development remain controversial and poorly understood. Although previous studies have implicated neural crest-derived periocular mesenchyme (POM) as the critical target of RA action in the eye, we show here that RAR signaling regulates choroid fissure closure in zebrafish by acting on both the ventral optic cup and the POM. We describe RAR-dependent regulation of eight genes in the neuroepithelial cells of the ventral retina and optic stalk and of six genes in the POM and show that these ventral retina/optic stalk and POM genes function independently of each other. Consequently, RAR signaling regulates ventral eye development through two independent, nonredundant mechanisms in different ocular tissues. Furthermore, the identification of two cohorts of genes implicated in ventral eye morphogenesis may help to elucidate the genetic basis of ocular coloboma in humans.


Asunto(s)
Coroides/ultraestructura , Ojo/crecimiento & desarrollo , Mesodermo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal/fisiología , Animales , Coroides/metabolismo , Coloboma , Embrión no Mamífero , Humanos , Morfogénesis , Cresta Neural/citología , Nervio Óptico/anomalías , Pez Cebra
6.
Science ; 384(6695): 573-579, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38696577

RESUMEN

Neurons on the left and right sides of the nervous system often show asymmetric properties, but how such differences arise is poorly understood. Genetic screening in zebrafish revealed that loss of function of the transmembrane protein Cachd1 resulted in right-sided habenula neurons adopting left-sided identity. Cachd1 is expressed in neuronal progenitors, functions downstream of asymmetric environmental signals, and influences timing of the normally asymmetric patterns of neurogenesis. Biochemical and structural analyses demonstrated that Cachd1 can bind simultaneously to Lrp6 and Frizzled family Wnt co-receptors. Consistent with this, lrp6 mutant zebrafish lose asymmetry in the habenulae, and epistasis experiments support a role for Cachd1 in modulating Wnt pathway activity in the brain. These studies identify Cachd1 as a conserved Wnt receptor-interacting protein that regulates lateralized neuronal identity in the zebrafish brain.


Asunto(s)
Canales de Calcio , Habénula , Neurogénesis , Neuronas , Vía de Señalización Wnt , Proteínas de Pez Cebra , Pez Cebra , Animales , Receptores Frizzled/metabolismo , Receptores Frizzled/genética , Habénula/metabolismo , Habénula/embriología , Mutación con Pérdida de Función , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Neuronas/metabolismo , Receptores Wnt/metabolismo , Receptores Wnt/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Canales de Calcio/genética , Canales de Calcio/metabolismo
7.
Blood ; 116(5): 829-40, 2010 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-20404134

RESUMEN

Blood vessel networks expand in a 2-step process that begins with vessel sprouting and is followed by vessel anastomosis. Vessel sprouting is induced by chemotactic gradients of the vascular endothelial growth factor (VEGF), which stimulates tip cell protrusion. Yet it is not known which factors promote the fusion of neighboring tip cells to add new circuits to the existing vessel network. By combining the analysis of mouse mutants defective in macrophage development or VEGF signaling with live imaging in zebrafish, we now show that macrophages promote tip cell fusion downstream of VEGF-mediated tip cell induction. Macrophages therefore play a hitherto unidentified and unexpected role as vascular fusion cells. Moreover, we show that there are striking molecular similarities between the pro-angiogenic tissue macrophages essential for vascular development and those that promote the angiogenic switch in cancer, including the expression of the cell-surface proteins TIE2 and NRP1. Our findings suggest that tissue macrophages are a target for antiangiogenic therapies, but that they could equally well be exploited to stimulate tissue vascularization in ischemic disease.


Asunto(s)
Macrófagos/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Rombencéfalo/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Polaridad Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Células Endoteliales/ultraestructura , Endotelio Vascular/crecimiento & desarrollo , Femenino , Técnicas de Sustitución del Gen , Factor Estimulante de Colonias de Macrófagos/deficiencia , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/fisiología , Masculino , Ratones , Ratones Noqueados , Neovascularización Fisiológica/fisiología , Neuropilina-1/fisiología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor TIE-2 , Vasos Retinianos/crecimiento & desarrollo , Rombencéfalo/embriología , Transactivadores/deficiencia , Factor A de Crecimiento Endotelial Vascular/deficiencia , Factor A de Crecimiento Endotelial Vascular/genética , Saco Vitelino/citología , Pez Cebra/embriología
8.
Biol Open ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34472582

RESUMEN

Zebrafish provide a unique opportunity for drug screening in living animals, with the fast-developing, transparent embryos allowing for relatively high-throughput, microscopy-based screens. However, the limited availability of rapid, flexible imaging and analysis platforms has limited the use of zebrafish in drug screens. We have developed an easy-to-use, customisable automated screening procedure suitable for high-throughput phenotype-based screens of live zebrafish. We utilised the WiScan® Hermes High Content Imaging System to rapidly acquire brightfield and fluorescent images of embryos, and the WiSoft® Athena Zebrafish Application for analysis, which harnesses an Artificial Intelligence-driven algorithm to automatically detect fish in brightfield images, identify anatomical structures, partition the animal into regions and exclusively select the desired side-oriented fish. Our initial validation combined structural analysis with fluorescence images to enumerate GFP-tagged haematopoietic stem and progenitor cells in the tails of embryos, which correlated with manual counts. We further validated this system to assess the effects of genetic mutations and X-ray irradiation in high content using a wide range of assays. Further, we performed simultaneous analysis of multiple cell types using dual fluorophores in high throughput. In summary, we demonstrate a broadly applicable and rapidly customisable platform for high-content screening in zebrafish. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Embrión no Mamífero/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Modelos Animales , Pez Cebra/embriología , Algoritmos , Animales , Fenotipo
9.
Int J Dev Biol ; 65(4-5-6): 289-299, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32930361

RESUMEN

Shaping the vertebrate eye requires evagination of the optic vesicles. These vesicles subsequently fold into optic cups prior to undergoing neurogenesis and allocating a population of late progenitors at the margin of the eye. mab21l2 encodes a protein of unknown biological function expressed in the developing optic vesicles, and loss of mab21l2 function results in malformed eyes. The bases of these defects are, however, poorly understood. To further study mab21l2 we used CRISPR/Cas9 to generate a new zebrafish mutant allele (mab21l2u517). We characterized eye morphogenesis and neurogenesis upon loss of mab21l2 function using tissue/cell-type-specific transgenes and immunostaining, in situ hybridization and bromodeoxyuridine incorporation. mab21l2u517 eyes fail to grow properly and display an excess of progenitors in the ciliary marginal zone. The expression of a transgene reporter for the vsx2 gene -a conserved marker for retinal progenitors- was delayed in mutant eyes and accompanied by disruptions in the epithelial folding that fuels optic cup morphogenesis. Mutants also displayed nasal-temporal malformations suggesting asynchronous development along that axis. Consistently, nasal retinal neurogenesis initiated but did not propagate in a timely fashion to the temporal retina. Later in development, mutant retinas did laminate and differentiate. Thus, mab21l2u517 mutants present a complex eye morphogenesis phenotype characterized by an organ-specific developmental delay. We propose that mab21l2 facilitates optic cup development with consequences both for timely neurogenesis and allocation of progenitors to the zebrafish ciliary marginal zone. These results confirm and extend previous analyses supporting the role of mab21l2 in coordinating morphogenesis and differentiation in developing eyes.


Asunto(s)
Ojo/embriología , Proteínas de Homeodominio/genética , Proteínas de Pez Cebra/genética , Pez Cebra , Animales , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Neurogénesis/genética , Retina/embriología , Pez Cebra/embriología , Pez Cebra/genética
10.
Elife ; 102021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33416493

RESUMEN

Hundreds of human genes are associated with neurological diseases, but translation into tractable biological mechanisms is lagging. Larval zebrafish are an attractive model to investigate genetic contributions to neurological diseases. However, current CRISPR-Cas9 methods are difficult to apply to large genetic screens studying behavioural phenotypes. To facilitate rapid genetic screening, we developed a simple sequencing-free tool to validate gRNAs and a highly effective CRISPR-Cas9 method capable of converting >90% of injected embryos directly into F0 biallelic knockouts. We demonstrate that F0 knockouts reliably recapitulate complex mutant phenotypes, such as altered molecular rhythms of the circadian clock, escape responses to irritants, and multi-parameter day-night locomotor behaviours. The technique is sufficiently robust to knockout multiple genes in the same animal, for example to create the transparent triple knockout crystal fish for imaging. Our F0 knockout method cuts the experimental time from gene to behavioural phenotype in zebrafish from months to one week.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Inactivación de Genes , Pruebas Genéticas/métodos , ARN Guía de Kinetoplastida/análisis , Pez Cebra/genética , Animales , Conducta Animal , Embrión no Mamífero , Fenotipo , Pez Cebra/embriología
11.
Dev Biol ; 332(2): 287-98, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19500562

RESUMEN

To gain insight into the mechanisms of Lmx1b function during ocular morphogenesis, we have studied the roles of lmx1b.1 and lmx1b.2 during zebrafish eye development. In situ hybridization and characterization of transgenic lines in which GFP is expressed under lmx1b.1 regulatory sequence show that these genes are expressed in periocular tissues and in a pattern conserved with other vertebrates. Anti-sense morpholinos against lmx1b.1 and lmx1b.2 result in defective migration of periocular mesenchymal cells around the eye and lead to apoptosis of these cells. These defects in the periocular mesenchyme are correlated with a failure in fusion of the choroid fissure or in some instances, more severe ventral optic cup morphogenesis phenotypes. Indeed, by blocking the death of the periocular mesenchyme in Lmx1b morphants, optic vesicle morphogenesis is largely restored. Within the retina of lmx1b morphants, Fgf activity is transiently up-regulated and these morphants show defective naso-temporal patterning. Epistasis experiments indicate that the increase in Fgf activity is partially responsible for the ocular anomalies caused by loss of Lmx1b function. Overall, we propose zebrafish lmx1b.1 and lmx1b.2 promote the survival of periocular mesenchymal cells that influence multiple signaling events required for proper ocular development.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/metabolismo , Mesodermo , Morfogénesis/fisiología , Retina , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra , Animales , Animales Modificados Genéticamente , Factores de Crecimiento de Fibroblastos/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Hibridación in Situ , Proteínas con Homeodominio LIM , Mesodermo/citología , Mesodermo/fisiología , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Retina/anomalías , Retina/citología , Retina/embriología , Transducción de Señal/fisiología , Factores de Transcripción/genética , Pez Cebra/anatomía & histología , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
12.
Hum Genet ; 126(6): 791-803, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19685247

RESUMEN

Mutations in the transcription factor encoding TFAP2A gene underlie branchio-oculo-facial syndrome (BOFS), a rare dominant disorder characterized by distinctive craniofacial, ocular, ectodermal and renal anomalies. To elucidate the range of ocular phenotypes caused by mutations in TFAP2A, we took three approaches. First, we screened a cohort of 37 highly selected individuals with severe ocular anomalies plus variable defects associated with BOFS for mutations or deletions in TFAP2A. We identified one individual with a de novo TFAP2A four amino acid deletion, a second individual with two non-synonymous variations in an alternative splice isoform TFAP2A2, and a sibling-pair with a paternally inherited whole gene deletion with variable phenotypic expression. Second, we determined that TFAP2A is expressed in the lens, neural retina, nasal process, and epithelial lining of the oral cavity and palatal shelves of human and mouse embryos--sites consistent with the phenotype observed in patients with BOFS. Third, we used zebrafish to examine how partial abrogation of the fish ortholog of TFAP2A affects the penetrance and expressivity of ocular phenotypes due to mutations in genes encoding bmp4 or tcf7l1a. In both cases, we observed synthetic, enhanced ocular phenotypes including coloboma and anophthalmia when tfap2a is knocked down in embryos with bmp4 or tcf7l1a mutations. These results reveal that mutations in TFAP2A are associated with a wide range of eye phenotypes and that hypomorphic tfap2a mutations can increase the risk of developmental defects arising from mutations at other loci.


Asunto(s)
Anomalías del Ojo/genética , Ojo/embriología , Retina/anomalías , Factor de Transcripción AP-2/genética , Adulto , Animales , Síndrome Branquio Oto Renal/genética , Preescolar , Femenino , Eliminación de Gen , Humanos , Lactante , Masculino , Persona de Mediana Edad , Morfogénesis/genética , Mutación , Pez Cebra , Proteínas de Pez Cebra/genética
13.
iScience ; 11: 205-223, 2019 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-30623799

RESUMEN

The transmembrane protein neuropilin-1 (NRP1) promotes vascular endothelial growth factor (VEGF) and extracellular matrix signaling in endothelial cells (ECs). Although it is established that NRP1 is essential for angiogenesis, little is known about its role in EC homeostasis. Here, we report that NRP1 promotes mitochondrial function in ECs by preventing iron accumulation and iron-induced oxidative stress through a VEGF-independent mechanism in non-angiogenic ECs. Furthermore, NRP1-deficient ECs have reduced growth and show the hallmarks of cellular senescence. We show that a subcellular pool of NRP1 localizes in mitochondria and interacts with the mitochondrial transporter ATP-binding cassette B8 (ABCB8). NRP1 loss reduces ABCB8 levels, resulting in iron accumulation, iron-induced mitochondrial superoxide production, and iron-dependent EC senescence. Treatment of NRP1-deficient ECs with the mitochondria-targeted antioxidant compound mitoTEMPO or with the iron chelator deferoxamine restores mitochondrial activity, inhibits superoxide production, and protects from cellular senescence. This finding identifies an unexpected role of NRP1 in EC homeostasis.

14.
PLoS One ; 14(1): e0211073, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30695021

RESUMEN

Through forward genetic screening for mutations affecting visual system development, we identified prominent coloboma and cell-autonomous retinal neuron differentiation, lamination and retinal axon projection defects in eisspalte (ele) mutant zebrafish. Additional axonal deficits were present, most notably at midline axon commissures. Genetic mapping and cloning of the ele mutation showed that the affected gene is slbp, which encodes a conserved RNA stem-loop binding protein involved in replication dependent histone mRNA metabolism. Cells throughout the central nervous system remained in the cell cycle in ele mutant embryos at stages when, and locations where, post-mitotic cells have differentiated in wild-type siblings. Indeed, RNAseq analysis showed down-regulation of many genes associated with neuronal differentiation. This was coincident with changes in the levels and spatial localisation of expression of various genes implicated, for instance, in axon guidance, that likely underlie specific ele phenotypes. These results suggest that many of the cell and tissue specific phenotypes in ele mutant embryos are secondary to altered expression of modules of developmental regulatory genes that characterise, or promote transitions in, cell state and require the correct function of Slbp-dependent histone and chromatin regulatory genes.


Asunto(s)
Animales Modificados Genéticamente , Orientación del Axón/genética , Diferenciación Celular , Proliferación Celular , Coloboma , Enfermedades de la Retina , Proteínas de Pez Cebra/deficiencia , Pez Cebra , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/genética , Coloboma/embriología , Coloboma/genética , Coloboma/patología , Histonas/genética , Histonas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Enfermedades de la Retina/embriología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Pez Cebra/embriología , Pez Cebra/genética
15.
Elife ; 82019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30777146

RESUMEN

The vertebrate eye originates from the eye field, a domain of cells specified by a small number of transcription factors. In this study, we show that Tcf7l1a is one such transcription factor that acts cell-autonomously to specify the eye field in zebrafish. Despite the much-reduced eye field in tcf7l1a mutants, these fish develop normal eyes revealing a striking ability of the eye to recover from a severe early phenotype. This robustness is not mediated through genetic compensation at neural plate stage; instead, the smaller optic vesicle of tcf7l1a mutants shows delayed neurogenesis and continues to grow until it achieves approximately normal size. Although the developing eye is robust to the lack of Tcf7l1a function, it is sensitised to the effects of additional mutations. In support of this, a forward genetic screen identified mutations in hesx1, cct5 and gdf6a, which give synthetically enhanced eye specification or growth phenotypes when in combination with the tcf7l1a mutation.


Asunto(s)
Ojo/crecimiento & desarrollo , Morfogénesis , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Proliferación Celular , Embrión no Mamífero/metabolismo , Ojo/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos , Cinética , Masculino , Mutación/genética , Placa Neural/embriología , Neurogénesis , Penetrancia , Fenotipo , Prosencéfalo/embriología , Proteína 1 Similar al Factor de Transcripción 7/genética , Regulación hacia Arriba/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Cigoto/metabolismo
16.
Front Cell Neurosci ; 12: 42, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29515375

RESUMEN

Coloboma is a defect in the morphogenesis of the eye that is a consequence of failure of choroid fissure fusion. It is among the most common congenital defects in humans and can significantly impact vision. However, very little is known about the cellular mechanisms that regulate choroid fissure closure. Using high-resolution confocal imaging of the zebrafish optic cup, we find that apico-basal polarity is re-modeled in cells lining the fissure in proximal to distal and inner to outer gradients during fusion. This process is accompanied by cell proliferation, displacement of vasculature, and contact between cells lining the choroid fissure and periocular mesenchyme (POM). To investigate the role of POM cells in closure of the fissure, we transplanted optic vesicles onto the yolk, allowing them to develop in a situation where they are depleted of POM. The choroid fissure forms normally in ectopic eyes but fusion fails in this condition, despite timely apposition of the nasal and temporal lips of the retina. This study resolves some of the cell behaviors underlying choroid fissure fusion and supports a role for POM in choroid fissure fusion.

17.
Curr Opin Genet Dev ; 32: 73-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25748250

RESUMEN

Vertebrate eye formation is a multistep process requiring coordinated inductive interactions between neural and non-neural ectoderm and underlying mesendoderm. The induction and shaping of the eyes involves an elaborate cellular choreography characterized by precise changes in cell shape coupled with complex cellular and epithelial movements. Consequently, the forming eye is an excellent model to study the cellular mechanisms underlying complex tissue morphogenesis. Using examples largely drawn from recent studies of optic vesicle formation in zebrafish and in cultured embryonic stem cells, in this short review, we highlight some recent advances in our understanding of the events that shape the vertebrate eye.


Asunto(s)
Ectodermo/embriología , Células Madre Embrionarias/fisiología , Ojo/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Modelos Biológicos , Organogénesis/fisiología , Vertebrados/embriología , Animales , Especificidad de la Especie , Pez Cebra
18.
Cell Rep ; 11(10): 1577-90, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26051942

RESUMEN

Sprouting blood vessels are led by filopodia-studded endothelial tip cells that respond to angiogenic signals. Mosaic lineage tracing previously revealed that NRP1 is essential for tip cell function, although its mechanistic role in tip cells remains poorly defined. Here, we show that NRP1 is dispensable for genetic tip cell identity. Instead, we find that NRP1 is essential to form the filopodial bursts that distinguish tip cells morphologically from neighboring stalk cells, because it enables the extracellular matrix (ECM)-induced activation of CDC42, a key regulator of filopodia formation. Accordingly, NRP1 knockdown and pharmacological CDC42 inhibition similarly impaired filopodia formation in vitro and in developing zebrafish in vivo. During mouse retinal angiogenesis, CDC42 inhibition impaired tip cell and vascular network formation, causing defects that resembled those due to loss of ECM-induced, but not VEGF-induced, NRP1 signaling. We conclude that NRP1 enables ECM-induced filopodia formation for tip cell function during sprouting angiogenesis.


Asunto(s)
Células Endoteliales/citología , Neuropilina-1/genética , Neuropilina-1/metabolismo , Seudópodos/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Animales , Embrión de Mamíferos , Células Endoteliales/metabolismo , Inmunohistoquímica , Ratones , Neovascularización Fisiológica/fisiología , Rombencéfalo/irrigación sanguínea , Rombencéfalo/citología , Transducción de Señal , Pez Cebra
19.
Nat Commun ; 5: 4272, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-25001599

RESUMEN

Patterning of the vertebrate optic vesicle into proximal/optic stalk and distal/neural retina involves midline-derived Hedgehog (Hh) signalling, which promotes stalk specification. In the absence of Hh signalling, the stalks are not specified, causing cyclopia. Recent studies showed that the cell adhesion molecule Cdon forms a heteromeric complex with the Hh receptor Patched 1 (Ptc1). This receptor complex binds Hh and enhances signalling activation, indicating that Cdon positively regulates the pathway. Here we show that in the developing zebrafish and chick optic vesicle, in which cdon and ptc1 are expressed with a complementary pattern, Cdon acts as a negative Hh signalling regulator. Cdon predominantly localizes to the basolateral side of neuroepithelial cells, promotes the enlargement of the neuroepithelial basal end-foot and traps Hh protein, thereby limiting its dispersion. This Ptc-independent function protects the retinal primordium from Hh activity, defines the stalk/retina boundary and thus the correct proximo-distal patterning of the eye.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Ojo/embriología , Proteínas Hedgehog/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Sitios de Unión , Tipificación del Cuerpo , Embrión de Pollo , Células HEK293 , Humanos , Proteínas de la Membrana , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/metabolismo , Pez Cebra
20.
Curr Biol ; 24(19): 2217-27, 2014 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-25201686

RESUMEN

BACKGROUND: Although left-right asymmetries are common features of nervous systems, their developmental bases are largely unknown. In the zebrafish epithalamus, dorsal habenular neurons adopt medial (dHbm) and lateral (dHbl) subnuclear character at very different frequencies on the left and right sides. The left-sided parapineal promotes the elaboration of dHbl character in the left habenula, albeit by an unknown mechanism. Likewise, the genetic pathways acting within habenular neurons to control their asymmetric differentiated character are unknown. RESULTS: In a forward genetic screen for mutations that result in loss of habenular asymmetry, we identified two mutant alleles of tcf7l2, a gene that encodes a transcriptional regulator of Wnt signaling. In tcf7l2 mutants, most neurons on both sides differentiate with dHbl identity. Consequently, the habenulae develop symmetrically, with both sides adopting a pronounced leftward character. Tcf7l2 acts cell automously in nascent equipotential neurons, and on the right side, it promotes dHbm and suppresses dHbl differentiation. On the left, the parapineal prevents this Tcf7l2-dependent process, thereby promoting dHbl differentiation. CONCLUSIONS: Tcf7l2 is essential for lateralized fate selection by habenular neurons that can differentiate along two alternative pathways, thereby leading to major neural circuit asymmetries.


Asunto(s)
Diferenciación Celular , Habénula/embriología , Neuronas/fisiología , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Embrión no Mamífero/embriología , Embrión no Mamífero/fisiología , Regulación de la Expresión Génica , Habénula/citología , Neuronas/citología , Transducción de Señal , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Pez Cebra/fisiología , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA