Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 432
Filtrar
1.
Development ; 151(20)2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39171364

RESUMEN

The first lineage differentiation in mammals gives rise to the inner cell mass and the trophectoderm (TE). In mice, TEAD4 is a master regulator of TE commitment, as it regulates the expression of other TE-specific genes and its ablation prevents blastocyst formation, but its role in other mammals remains unclear. Herein, we have observed that TEAD4 ablation in two phylogenetically distant species (bovine and rabbit) does not impede TE differentiation, blastocyst formation and the expression of TE markers, such as GATA3 and CDX2, although a reduced number of cells in the inner cell mass was observed in bovine TEAD4 knockout (KO) blastocysts. Transcriptional analysis in bovine blastocysts revealed no major transcriptional effect of the ablation, although the expression of hypoblast and Hippo signalling-related genes tended to be decreased in KO embryos. Experiments were conducted in the bovine model to determine whether TEAD4 was required for post-hatching development. TEAD4 KO spherical conceptuses showed normal development of the embryonic disc and TE, but hypoblast migration rate was reduced. At later stages of development (tubular conceptuses), no differences were observed between KO and wild-type conceptuses.


Asunto(s)
Blastocisto , Diferenciación Celular , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción de Dominio TEA , Factores de Transcripción , Animales , Bovinos , Factores de Transcripción de Dominio TEA/metabolismo , Blastocisto/metabolismo , Blastocisto/citología , Diferenciación Celular/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Conejos , Desarrollo Embrionario/genética , Ectodermo/metabolismo , Ectodermo/embriología , Ectodermo/citología , Femenino , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Trofoblastos/metabolismo , Trofoblastos/citología , Ratones , Vía de Señalización Hippo , Embrión de Mamíferos/metabolismo , Factor de Transcripción CDX2/metabolismo , Factor de Transcripción CDX2/genética , Factor de Transcripción GATA3/metabolismo , Factor de Transcripción GATA3/genética
2.
Dev Biol ; 516: 114-121, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39102935

RESUMEN

The lack of a widely accessible method for expressing genes of interest in wild-type embryos is a fundamental obstacle to understanding genetic regulation during embryonic development. In particular, only a few methods are available for introducing gene expression vectors into cells prior to neural tube closure, which is a period of drastic development for many tissues. In this study, we present a simple technique for injecting vectors into the amniotic cavity and allowing them to reach the ectodermal cells and the epithelia of endodermal organs of mouse embryos at E8.0 via in utero injection, using only a widely used optical fiber with an illuminator. Using this technique, retroviruses can be introduced to facilitate the labeling of cells in various tissues, including the brain, spinal cord, epidermis, and digestive and respiratory organs. We also demonstrated in utero electroporation of plasmid DNA into E7.0 and E8.0 embryos. Taking advantage of this method, we reveal the association between Ldb1 and the activity of the Neurog2 transcription factor in the mouse neocortex. This technique can aid in analyzing the roles of genes of interest during endo- and ectodermal development prior to neural tube closure.


Asunto(s)
Ectodermo , Electroporación , Regulación del Desarrollo de la Expresión Génica , Tubo Neural , Animales , Ectodermo/metabolismo , Ectodermo/embriología , Ratones , Tubo Neural/embriología , Tubo Neural/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Electroporación/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Embrión de Mamíferos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodermo/metabolismo , Endodermo/embriología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neurulación/genética , Vectores Genéticos/genética , Embarazo
3.
Dev Biol ; 515: 30-45, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38971398

RESUMEN

The ectoderm is the outermost of the three germ layers of the early embryo that arise during gastrulation. Once the germ layers are established, the complex interplay of cellular proliferation, differentiation, and migration results in organogenesis. The ectoderm is the progenitor of both the surface ectoderm and the neural ectoderm. Notably, the surface ectoderm develops into the epidermis and its associated appendages, nails, external exocrine glands, olfactory epithelium, and the anterior pituitary. Specification, development, and homeostasis of these organs demand a tightly orchestrated gene expression program that is often dictated by epigenetic regulation. In this review, we discuss the recent discoveries that have highlighted the importance of chromatin regulatory mechanisms mediated by transcription factors, histone and DNA modifications that aid in the development of surface ectodermal organs and maintain their homeostasis post-development.


Asunto(s)
Cromatina , Ectodermo , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Ectodermo/metabolismo , Ectodermo/embriología , Animales , Cromatina/metabolismo , Epigénesis Genética , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Diferenciación Celular/genética , Histonas/metabolismo
4.
Int J Dev Biol ; 68(2): 65-78, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39016374

RESUMEN

During embryonic development, the vertebrate embryonic epiblast is divided into two parts including neural and superficial ectoderm. The neural plate border (NPB) is a narrow transitional area which locates between these parts and contains multipotent progenitor cells. Despite its small size, the cellular heterogeneity in this region produces specific differentiated cells. Signaling pathways, transcription factors, and the expression/repression of certain genes are directly involved in these differentiation processes. Different factors such as the Wnt signaling cascade, fibroblast growth factor (FGF), bone morphogenetic protein (BMP) signaling, and Notch, which are involved in various stages of the growth, proliferation, and differentiation of embryonic cells, are also involved in the determination and differentiation of neural plate border stem cells. Therefore, it is essential to consider the interactions and temporospatial coordination related to cells, tissues, and adjacent structures. This review examines our present knowledge of the formation of the neural plate border and emphasizes the requirement for interaction between different signaling pathways, including the BMP and Wnt cascades, the expression of its special target genes and their regulations, and the precise tissue crosstalk which defines the neural crest fate in the ectoderm at the early human embryonic stages.


Asunto(s)
Proteínas Morfogenéticas Óseas , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Cresta Neural , Placa Neural , Transducción de Señal , Placa Neural/metabolismo , Placa Neural/embriología , Humanos , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Cresta Neural/metabolismo , Cresta Neural/embriología , Ectodermo/metabolismo , Ectodermo/embriología , Ectodermo/citología , Vía de Señalización Wnt/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Estratos Germinativos/metabolismo , Estratos Germinativos/citología , Proteínas Wnt/metabolismo , Proteínas Wnt/genética
5.
PLoS Biol ; 22(4): e3002611, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683880

RESUMEN

As tissues grow and change shape during animal development, they physically pull and push on each other, and these mechanical interactions can be important for morphogenesis. During Drosophila gastrulation, mesoderm invagination temporally overlaps with the convergence and extension of the ectodermal germband; the latter is caused primarily by Myosin II-driven polarised cell intercalation. Here, we investigate the impact of mesoderm invagination on ectoderm extension, examining possible mechanical and mechanotransductive effects on Myosin II recruitment and polarised cell intercalation. We find that the germband ectoderm is deformed by the mesoderm pulling in the orthogonal direction to germband extension (GBE), showing mechanical coupling between these tissues. However, we do not find a significant change in Myosin II planar polarisation in response to mesoderm invagination, nor in the rate of junction shrinkage leading to neighbour exchange events. We conclude that the main cellular mechanism of axis extension, polarised cell intercalation, is robust to the mesoderm invagination pull. We find, however, that mesoderm invagination slows down the rate of anterior-posterior cell elongation that contributes to axis extension, counteracting the tension from the endoderm invagination, which pulls along the direction of GBE.


Asunto(s)
Drosophila melanogaster , Ectodermo , Gastrulación , Mesodermo , Miosina Tipo II , Animales , Mesodermo/embriología , Mesodermo/citología , Gastrulación/fisiología , Ectodermo/citología , Ectodermo/embriología , Ectodermo/metabolismo , Miosina Tipo II/metabolismo , Drosophila melanogaster/embriología , Polaridad Celular , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrión no Mamífero , Morfogénesis , Tipificación del Cuerpo/fisiología , Drosophila/embriología
6.
Zebrafish ; 21(2): 171-176, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38621215

RESUMEN

The transgenic (TG) zebrafish allows researchers to bio-image specific biological phenomena in cells and tissues in vivo. We established TG lines to monitor changes in the ovaries of live fish. The original TG line with ovarian fluorescence was occasionally established. Although the cDNA integrated into the line was constructed for the expression of enhanced green fluorescent protein (EGFP) driven by the medaka ß-actin promoter, the expression of EGFP is restricted to the oocytes and gills in adult fish. Furthermore, we found that germinal vesicles (GVs) in oocytes of the established line can be observed by relatively strong fluorescence around the GV. In this study, we tried to capture the dynamic processes of germinal vesicle breakdown (GVBD) during meiotic cell division using the GV fluorescent oocytes. As a result, GV migration and GVBD could be monitored in real time. We also succeeded in observing actin filaments involved in the migration of GV to the animal pole. This strain can be used for education in the process of oocyte meiotic cell division.


Asunto(s)
Ectodermo/embriología , Estructuras Embrionarias , Ovario , Pez Cebra , Femenino , Animales , Oocitos , Animales Modificados Genéticamente , División Celular
7.
Nature ; 626(7998): 357-366, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38052228

RESUMEN

Recently, several studies using cultures of human embryos together with single-cell RNA-seq analyses have revealed differences between humans and mice, necessitating the study of human embryos1-8. Despite the importance of human embryology, ethical and legal restrictions have limited post-implantation-stage studies. Thus, recent efforts have focused on developing in vitro self-organizing models using human stem cells9-17. Here, we report genetic and non-genetic approaches to generate authentic hypoblast cells (naive hPSC-derived hypoblast-like cells (nHyCs))-known to give rise to one of the two extraembryonic tissues essential for embryonic development-from naive human pluripotent stem cells (hPSCs). Our nHyCs spontaneously assemble with naive hPSCs to form a three-dimensional bilaminar structure (bilaminoids) with a pro-amniotic-like cavity. In the presence of additional naive hPSC-derived analogues of the second extraembryonic tissue, the trophectoderm, the efficiency of bilaminoid formation increases from 20% to 40%, and the epiblast within the bilaminoids continues to develop in response to trophectoderm-secreted IL-6. Furthermore, we show that bilaminoids robustly recapitulate the patterning of the anterior-posterior axis and the formation of cells reflecting the pregastrula stage, the emergence of which can be shaped by genetically manipulating the DKK1/OTX2 hypoblast-like domain. We have therefore successfully modelled and identified the mechanisms by which the two extraembryonic tissues efficiently guide the stage-specific growth and progression of the epiblast as it establishes the post-implantation landmarks of human embryogenesis.


Asunto(s)
Desarrollo Embrionario , Estratos Germinativos , Células Madre Pluripotentes , Humanos , Diferenciación Celular , Implantación del Embrión , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Estratos Germinativos/citología , Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Células Madre Pluripotentes/citología , Interleucina-6/metabolismo , Gástrula/citología , Gástrula/embriología , Amnios/citología , Amnios/embriología , Amnios/metabolismo , Ectodermo/citología , Ectodermo/embriología , Ectodermo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(28): e2118938119, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35867760

RESUMEN

The vertebrate inner ear arises from a pool of progenitors with the potential to contribute to all the sense organs and cranial ganglia in the head. Here, we explore the molecular mechanisms that control ear specification from these precursors. Using a multiomics approach combined with loss-of-function experiments, we identify a core transcriptional circuit that imparts ear identity, along with a genome-wide characterization of noncoding elements that integrate this information. This analysis places the transcription factor Sox8 at the top of the ear determination network. Introducing Sox8 into the cranial ectoderm not only converts non-ear cells into ear progenitors but also activates the cellular programs for ear morphogenesis and neurogenesis. Thus, Sox8 has the unique ability to remodel transcriptional networks in the cranial ectoderm toward ear identity.


Asunto(s)
Oído Interno , Ectodermo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción SOXE , Animales , Oído Interno/embriología , Ectodermo/embriología , Factores de Transcripción SOXE/fisiología , Cráneo , Vertebrados/embriología
9.
Dev Cell ; 57(12): 1482-1495.e5, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35679863

RESUMEN

Development of primordial germ cells (PGCs) is required for reproduction. During PGC development in mammals, major epigenetic remodeling occurs, which is hypothesized to establish an epigenetic landscape for sex-specific germ cell differentiation and gametogenesis. In order to address the role of embryonic ectoderm development (EED) and histone 3 lysine 27 trimethylation (H3K27me3) in this process, we created an EED conditional knockout mouse and show that EED is essential for regulating the timing of sex-specific PGC differentiation in both ovaries and testes, as well as X chromosome dosage decompensation in testes. Integrating chromatin and whole genome bisulfite sequencing of epiblast and PGCs, we identified a poised repressive signature of H3K27me3/DNA methylation that we propose is established in the epiblast where EED and DNMT1 interact. Thus, EED joins DNMT1 in regulating the timing of sex-specific PGC differentiation during the critical window when the gonadal niche cells specialize into an ovary or testis.


Asunto(s)
Células Germinativas , Histonas , Complejo Represivo Polycomb 2 , Animales , Diferenciación Celular/genética , Metilación de ADN , Ectodermo/embriología , Femenino , Células Germinativas/metabolismo , Gónadas/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(20): e2117075119, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35561223

RESUMEN

Neurulation is the process in early vertebrate embryonic development during which the neural plate folds to form the neural tube. Spinal neural tube folding in the posterior neuropore changes over time, first showing a median hinge point, then both the median hinge point and dorsolateral hinge points, followed by dorsolateral hinge points only. The biomechanical mechanism of hinge point formation in the mammalian neural tube is poorly understood. Here we employ a mechanical finite element model to study neural tube formation. The computational model mimics the mammalian neural tube using microscopy data from mouse and human embryos. While intrinsic curvature at the neural plate midline has been hypothesized to drive neural tube folding, intrinsic curvature was not sufficient for tube closure in our simulations. We achieved neural tube closure with an alternative model combining mesoderm expansion, nonneural ectoderm expansion, and neural plate adhesion to the notochord. Dorsolateral hinge points emerged in simulations with low mesoderm expansion and zippering. We propose that zippering provides the biomechanical force for dorsolateral hinge point formation in settings where the neural plate lateral sides extend above the mesoderm. Together, these results provide a perspective on the biomechanical and molecular mechanism of mammalian spinal neurulation.


Asunto(s)
Tubo Neural , Neurulación , Animales , Ectodermo/embriología , Humanos , Ratones , Placa Neural/embriología , Tubo Neural/embriología , Neurulación/fisiología , Notocorda/embriología
11.
Dev Biol ; 483: 128-142, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35038441

RESUMEN

Brachyury is a T-box family transcription factor and plays pivotal roles in morphogenesis. In sea urchin embryos, Brachyury is expressed in the invaginating endoderm, and in the oral ectoderm of the invaginating mouth opening. The oral ectoderm is hypothesized to serve as a signaling center for oral (ventral)-aboral (dorsal) axis formation and to function as a ventral organizer. Our previous results of a single-cell RNA-seq (scRNA-seq) atlas of early Strongylocentrotus purpuratus embryos categorized the constituent cells into 22 clusters, in which the endoderm consists of three clusters and the oral ectoderm four clusters (Foster et al., 2020). Here we examined which clusters of cells expressed Brachyury in relation to the morphogenesis and the identity of the ventral organizer. Our results showed that cells of all three endoderm clusters expressed Brachyury in blastulae. Based on expression profiles of genes involved in the gene regulatory networks (GRNs) of sea urchin embryos, the three clusters are distinguishable, two likely derived from the Veg2 tier and one from the Veg1 tier. On the other hand, of the four oral-ectoderm clusters, cells of two clusters expressed Brachyury at the gastrula stage and genes that are responsible for the ventral organizer at the late blastula stage, but the other two clusters did not. At a single-cell level, most cells of the two oral-ectoderm clusters expressed organizer-related genes, nearly a half of which coincidently expressed Brachyury. This suggests that the ventral organizer contains Brachyury-positive cells which invaginate to form the stomodeum. This scRNA-seq study therefore highlights significant roles of Brachyury-expressing cells in body-plan formation of early sea urchin embryos, though cellular and molecular mechanisms for how Brachyury functions in these processes remain to be elucidated in future studies.


Asunto(s)
Ectodermo/citología , Ectodermo/metabolismo , Desarrollo Embrionario/genética , Proteínas Fetales/metabolismo , Regulación del Desarrollo de la Expresión Génica , RNA-Seq/métodos , Erizos de Mar/embriología , Erizos de Mar/genética , Análisis de la Célula Individual/métodos , Proteínas de Dominio T Box/metabolismo , Animales , Blástula/metabolismo , Ectodermo/embriología , Endodermo/embriología , Endodermo/metabolismo , Gástrula/metabolismo , Redes Reguladoras de Genes , Transducción de Señal/genética
12.
Nature ; 599(7884): 268-272, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34707290

RESUMEN

Understanding human organ formation is a scientific challenge with far-reaching medical implications1,2. Three-dimensional stem-cell cultures have provided insights into human cell differentiation3,4. However, current approaches use scaffold-free stem-cell aggregates, which develop non-reproducible tissue shapes and variable cell-fate patterns. This limits their capacity to recapitulate organ formation. Here we present a chip-based culture system that enables self-organization of micropatterned stem cells into precise three-dimensional cell-fate patterns and organ shapes. We use this system to recreate neural tube folding from human stem cells in a dish. Upon neural induction5,6, neural ectoderm folds into a millimetre-long neural tube covered with non-neural ectoderm. Folding occurs at 90% fidelity, and anatomically resembles the developing human neural tube. We find that neural and non-neural ectoderm are necessary and sufficient for folding morphogenesis. We identify two mechanisms drive folding: (1) apical contraction of neural ectoderm, and (2) basal adhesion mediated via extracellular matrix synthesis by non-neural ectoderm. Targeting these two mechanisms using drugs leads to morphological defects similar to neural tube defects. Finally, we show that neural tissue width determines neural tube shape, suggesting that morphology along the anterior-posterior axis depends on neural ectoderm geometry in addition to molecular gradients7. Our approach provides a new route to the study of human organ morphogenesis in health and disease.


Asunto(s)
Morfogénesis , Tubo Neural/anatomía & histología , Tubo Neural/embriología , Técnicas de Cultivo de Órganos/métodos , Ectodermo/citología , Ectodermo/embriología , Humanos , Modelos Biológicos , Placa Neural/citología , Placa Neural/embriología , Tubo Neural/citología , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología , Regeneración , Células Madre/citología
13.
Cell Death Dis ; 12(10): 850, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34531374

RESUMEN

PRC2-mediated epigenetic function involves the interaction with long non-coding RNAs (lncRNAs). Although the identity of some of these RNAs has been elucidated in the context of developmental programs, their counterparts in postmitotic adult tissue homeostasis remain uncharacterized. To this aim, we used terminally differentiated postmitotic skeletal muscle cells in which oxidative stress induces the dynamic activation of PRC2-Ezh1 through Embryonic Ectoderm Develpment (EED) shuttling to the nucleus. We identify lncRNA Malat-1 as a necessary partner for PRC2-Ezh1-dependent response to oxidative stress. We show that in this pathway, PRC2-EZH1 dynamic assembly, and in turn stress induced skeletal muscle targeted genes repression, depends specifically on Malat-1. Our study reports about PRC2-RNA interactions in the physiological context of adaptive oxidative stress response and identifies the first lncRNA involved in PRC2-Ezh1 function.


Asunto(s)
Epigenoma , Fibras Musculares Esqueléticas/metabolismo , Estrés Oxidativo , Complejo Represivo Polycomb 2/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Línea Celular , Ectodermo/embriología , Embrión de Mamíferos/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Histonas/metabolismo , Lisina/metabolismo , Metilación , Ratones , Modelos Biológicos , Atrofia Muscular/genética , Atrofia Muscular/patología , Estrés Oxidativo/genética , Fenotipo , Complejo Represivo Polycomb 2/genética , Unión Proteica , ARN Largo no Codificante/genética , Transcripción Genética
14.
Development ; 148(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34414417

RESUMEN

Branchio-oto-renal syndrome (BOR) is a disorder characterized by hearing loss, and craniofacial and/or renal defects. Variants in the transcription factor Six1 and its co-factor Eya1, both of which are required for otic development, are linked to BOR. We previously identified Sobp as a potential Six1 co-factor, and SOBP variants in mouse and humans cause otic phenotypes; therefore, we asked whether Sobp interacts with Six1 and thereby may contribute to BOR. Co-immunoprecipitation and immunofluorescence experiments demonstrate that Sobp binds to and colocalizes with Six1 in the cell nucleus. Luciferase assays show that Sobp interferes with the transcriptional activation of Six1+Eya1 target genes. Experiments in Xenopus embryos that either knock down or increase expression of Sobp show that it is required for formation of ectodermal domains at neural plate stages. In addition, altering Sobp levels disrupts otic vesicle development and causes craniofacial cartilage defects. Expression of Xenopus Sobp containing the human variant disrupts the pre-placodal ectoderm similar to full-length Sobp, but other changes are distinct. These results indicate that Sobp modifies Six1 function and is required for vertebrate craniofacial development, and identify Sobp as a potential candidate gene for BOR.


Asunto(s)
Desarrollo Óseo , Proteínas de Homeodominio/metabolismo , Metaloproteínas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Síndrome Branquio Oto Renal/embriología , Síndrome Branquio Oto Renal/genética , Núcleo Celular/metabolismo , Oído Interno/embriología , Oído Interno/metabolismo , Ectodermo/embriología , Ectodermo/metabolismo , Expresión Génica , Proteínas de Homeodominio/genética , Larva/crecimiento & desarrollo , Metaloproteínas/genética , Cresta Neural/embriología , Cresta Neural/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Proteínas Tirosina Fosfatasas/metabolismo , Activación Transcripcional , Proteínas de Xenopus/genética , Xenopus laevis
15.
PLoS One ; 16(7): e0254024, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34234366

RESUMEN

During embryonic development, cells differentiate into a variety of distinct cell types and subtypes with diverse transcriptional profiles. To date, transcriptomic signatures of different cell lineages that arise during development have been only partially characterized. Here we used single-cell RNA-seq to perform transcriptomic analysis of over 20,000 cells disaggregated from the trunk region of zebrafish embryos at the 30 hpf stage. Transcriptional signatures of 27 different cell types and subtypes were identified and annotated during this analysis. This dataset will be a useful resource for many researchers in the fields of developmental and cellular biology and facilitate the understanding of molecular mechanisms that regulate cell lineage choices during development.


Asunto(s)
Embrión no Mamífero/metabolismo , Perfilación de la Expresión Génica , Análisis de la Célula Individual , Torso/embriología , Pez Cebra/embriología , Pez Cebra/genética , Animales , Linaje de la Célula/genética , Ectodermo/citología , Ectodermo/embriología , Endodermo/citología , Endodermo/embriología , Endotelio Vascular/citología , Endotelio Vascular/embriología , Eritrocitos/metabolismo , Fibroblastos/citología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/citología , Mesodermo/embriología , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
Biochem Biophys Res Commun ; 567: 99-105, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34146908

RESUMEN

During vertebrate development, the formation of the central nervous system (CNS) is initiated by neural induction and patterning of the embryonic ectoderm. We previously reported that Cdc2-like kinase 2 (Clk2) promotes neural development in Xenopus embryos by regulating morphogen signaling. However, the functions of other Clk family members and their roles in early embryonic development remain unknown. Here, we show that in addition to Clk2, Clk1 and Clk3 play a role in the formation of neural tissue in Xenopus. clk1 and clk3 are co-expressed in the developing neural tissue during early Xenopus embryogenesis. We found that overexpression of clk1 and clk3 increases the expression of neural marker genes in ectodermal explants. Furthermore, knockdown experiments showed that clk3 is required for the formation of neural tissues. These results suggest that Xenopus Clk3 plays an essential role in promoting neural development during early embryogenesis.


Asunto(s)
Neurogénesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas de Xenopus/genética , Xenopus/embriología , Animales , Ectodermo/embriología , Ectodermo/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Xenopus/genética
17.
Cell Rep ; 35(12): 109289, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34161771

RESUMEN

The spatiotemporal coordination of multiple morphogens is essential for embryonic patterning yet poorly understood. During neural crest (NC) formation, dynamic bone morphogenetic protein (BMP), fibroblast growth factor (FGF), and WNT signals cooperate by acting on mesoderm and ectoderm. Here, we show that Fhl3, a scaffold LIM domain protein, modulates BMP gradient interpretation during NC induction. During gastrulation, low BMP signaling neuralizes the neural border (NB) ectoderm, while Fhl3 enhances Smad1 intracellular response in underlying paraxial mesoderm, triggering the high WNT8 signals needed to pattern the NB. During neurulation, fhl3 activation in NC ectoderm promotes simultaneous high BMP and BMP-dependent WNT activity required for specification. Mechanistically, Fhl3 interacts with Smad1 and promotes Smad1 binding to wnt8 promoter in a BMP-dependent manner. Consequently, differential Fhl3 expression in adjacent cells ensures a finely tuned coordination of BMP and WNT signaling at several stages of NC development, starting by positioning the NC-inducing mesoderm center under competent NB ectoderm.


Asunto(s)
Proteínas Morfogenéticas Óseas , Espacio Intracelular , Cresta Neural , Transducción de Señal , Proteínas Wnt , Proteínas de Xenopus , Animales , Humanos , Proteínas Morfogenéticas Óseas/metabolismo , Ectodermo/embriología , Gastrulación , Células HEK293 , Espacio Intracelular/metabolismo , Mesodermo/embriología , Cresta Neural/citología , Cresta Neural/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Wnt/metabolismo , Xenopus laevis/embriología , Proteínas de Xenopus/metabolismo
18.
Cold Spring Harb Protoc ; 2021(6)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33782096

RESUMEN

Problems of cell biology and the molecular controls underpinning them have been studied in the remarkably versatile Xenopus systems for many years. This versatility is showcased in several accompanying protocols, which are introduced here. One protocol demonstrates how the Xenopus embryonic ectoderm can be used to study the effects of mechanical cell deformation; another illustrates how the developing eye can be used as a platform for determining cell-cycle length. Two protocols show how extracts from Xenopus embryos can be exploited to characterize the behavior of specific intracellular proteins-specifically, to determine protein phosphorylation status and the ability to bind to chromatin. Finally, because specific antibodies to Xenopus proteins are pivotal reagents for cell biology and biochemistry applications, four protocols describing how to generate, purify, and assay the specificity of antibodies raised against Xenopus proteins are included in hopes of stimulating the expansion of these critical resources across the Xenopus community.


Asunto(s)
Biología Celular , Técnicas Citológicas/métodos , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Bioquímica/métodos , Cromatina/metabolismo , Ectodermo/embriología , Embrión no Mamífero/embriología , Humanos , Inmunidad/inmunología , Modelos Animales , Fosforilación , Unión Proteica , Proteínas de Xenopus/inmunología , Xenopus laevis
19.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33723076

RESUMEN

Specification of Sox2+ proneurosensory progenitors within otic ectoderm is a prerequisite for the production of sensory cells and neurons for hearing. However, the underlying molecular mechanisms driving this lineage specification remain unknown. Here, we show that the Brg1-based SWI/SNF chromatin-remodeling complex interacts with the neurosensory-specific transcriptional regulators Eya1/Six1 to induce Sox2 expression and promote proneurosensory-lineage specification. Ablation of the ATPase-subunit Brg1 or both Eya1/Six1 results in loss of Sox2 expression and lack of neurosensory identity, leading to abnormal apoptosis within the otic ectoderm. Brg1 binds to two of three distal 3' Sox2 enhancers occupied by Six1, and Brg1-binding to these regions depends on Eya1-Six1 activity. We demonstrate that the activity of these Sox2 enhancers in otic neurosensory cells specifically depends on binding to Six1. Furthermore, genome-wide and transcriptome profiling indicate that Brg1 may suppress apoptotic factor Map3k5 to inhibit apoptosis. Together, our findings reveal an essential role for Brg1, its downstream pathways, and their interactions with Six1/Eya1 in promoting proneurosensory fate induction in the otic ectoderm and subsequent neuronal lineage commitment and survival of otic cells.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/genética , Cromatina/metabolismo , Ectodermo/embriología , Ectodermo/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
20.
Development ; 148(4)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33593754

RESUMEN

The generation of the components that make up the embryonic body axis, such as the spinal cord and vertebral column, takes place in an anterior-to-posterior (head-to-tail) direction. This process is driven by the coordinated production of various cell types from a pool of posteriorly-located axial progenitors. Here, we review the key features of this process and the biology of axial progenitors, including neuromesodermal progenitors, the common precursors of the spinal cord and trunk musculature. We discuss recent developments in the in vitro production of axial progenitors and their potential implications in disease modelling and regenerative medicine.


Asunto(s)
Biología , Tipificación del Cuerpo , Gastrulación/fisiología , Estratos Germinativos/embriología , Animales , Ectodermo/embriología , Endodermo/embriología , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/inervación , Humanos , Técnicas In Vitro , Mesodermo/embriología , Mesodermo/inervación , Músculo Esquelético , Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...