Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 241
Filtrar
1.
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
2.
PLoS One ; 19(7): e0293852, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39083515

RESUMEN

The zinc finger and BTB domain-containing 11 gene (zbtb11) is expressed in the Xenopus anterior neuroectoderm, but the molecular nature of the Zbtb11 protein during embryonic development remains to be elucidated. Here, we show the role of Zbtb11 in anterior patterning of the neuroectoderm and the cooperative action with the transcription factor Otx2. Both overexpression and knockdown of zbtb11 caused similar phenotypes: expanded expression of the posterior gene gbx2 in the neural plate, and later microcephaly with reduced eyes, suggesting that a proper level of zbtb11 expression is necessary for normal patterning of the neuroectoderm, including eye formation. Co-immunoprecipitation assays showed that Zbtb11 formed a complex with itself and with a phosphomimetic and repressive form of Otx2, suggesting that Zbtb11 forms a dimer or oligomer and interacts with Otx2 in a phosphorylation-dependent manner. Reporter analysis further showed that Zbtb11 enhanced the activity of the phosphomimetic Otx2 to repress a silencer element of the posterior gene meis3. These data suggest that Zbtb11 coordinates with phosphorylated Otx2 to specify the anterior neuroectoderm by repressing posterior genes.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Placa Neural , Factores de Transcripción Otx , Proteínas de Xenopus , Animales , Factores de Transcripción Otx/metabolismo , Factores de Transcripción Otx/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Placa Neural/metabolismo , Placa Neural/embriología , Xenopus laevis , Unión Proteica , Fosforilación , Tipificación del Cuerpo/genética
3.
Dev Biol ; 514: 1-11, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38878991

RESUMEN

In chordates, the central nervous system arises from precursors that have distinct developmental and transcriptional trajectories. Anterior nervous systems are ontogenically associated with ectodermal lineages while posterior nervous systems are associated with mesoderm. Taking advantage of the well-documented cell lineage of ascidian embryos, we asked to what extent the transcriptional states of the different neural lineages become similar during the course of progressive lineage restriction. We performed single-cell RNA sequencing (scRNA-seq) analyses on hand-dissected neural precursor cells of the two distinct lineages, together with those of their sister cell lineages, with a high temporal resolution covering five successive cell cycles from the 16-cell to neural plate stages. A transcription factor binding site enrichment analysis of neural specific genes at the neural plate stage revealed limited evidence for shared transcriptional control between the two neural lineages, consistent with their different ontogenies. Nevertheless, PCA analysis and hierarchical clustering showed that, by neural plate stages, the two neural lineages cluster together. Consistent with this, we identified a set of genes enriched in both neural lineages at the neural plate stage, including miR-124, Celf3.a, Zic.r-b, and Ets1/2. Altogether, the current study has revealed genome-wide transcriptional dynamics of neural progenitor cells of two distinct developmental origins. Our scRNA-seq dataset is unique and provides a valuable resource for future analyses, enabling a precise temporal resolution of cell types not previously described from dissociated embryos.


Asunto(s)
Linaje de la Célula , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Animales , Linaje de la Célula/genética , Desarrollo Embrionario/genética , Placa Neural/embriología , Placa Neural/metabolismo , Placa Neural/citología , Ciona intestinalis/embriología , Ciona intestinalis/genética , Urocordados/embriología , Urocordados/genética , Análisis de la Célula Individual , Embrión no Mamífero/metabolismo , Embrión no Mamífero/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología
4.
Development ; 151(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38828854

RESUMEN

The neural plate border (NPB) of vertebrate embryos is segregated from the neural plate (NP) and epidermal regions, and comprises an intermingled group of progenitors with multiple fate potential. Recent studies have shown that, during the gastrula stage, TFAP2A acts as a pioneer factor in remodeling the epigenetic landscape required to activate components of the NPB induction program. Here, we show that chick Tfap2a has two highly conserved binding sites for miR-137, and both display a reciprocal expression pattern at the NPB and NP, respectively. In addition, ectopic miR-137 expression reduced TFAP2A, whereas its functional inhibition expanded their territorial distribution overlapping with PAX7. Furthermore, we demonstrate that loss of the de novo DNA methyltransferase DNMT3A expanded miR-137 expression to the NPB. Bisulfite sequencing revealed a markedly elevated presence of non-canonical CpH methylation within the miR-137 promoter region when comparing NPB and NP samples. Our findings show that miR-137 contributes to the robustness of NPB territorial restriction in vertebrate development.


Asunto(s)
Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , MicroARNs , Placa Neural , Factor de Transcripción AP-2 , Animales , MicroARNs/genética , MicroARNs/metabolismo , Embrión de Pollo , Metilación de ADN/genética , Placa Neural/metabolismo , Placa Neural/embriología , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A/metabolismo , Regiones Promotoras Genéticas/genética , Sitios de Unión
5.
Mol Cells ; 47(6): 100076, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38825188

RESUMEN

The actin-based cytoskeleton is considered a fundamental driving force for cell differentiation and development. Destrin (Dstn), a member of the actin-depolymerizing factor family, regulates actin dynamics by treadmilling actin filaments and increasing globular actin pools. However, the specific developmental roles of dstn have yet to be fully elucidated. Here, we investigated the physiological functions of dstn during early embryonic development using Xenopus laevis as an experimental model organism. dstn is expressed in anterior neural tissue and neural plate during Xenopus embryogenesis. Depleting dstn promoted morphants with short body axes and small heads. Moreover, dstn inhibition extended the neural plate region, impairing cell migration and distribution during neurulation. In addition to the neural plate, dstn knockdown perturbed neural crest cell migration. Our data suggest new insights for understanding the roles of actin dynamics in embryonic neural development, simultaneously presenting a new challenge for studying the complex networks governing cell migration involving actin dynamics.


Asunto(s)
Movimiento Celular , Destrina , Desarrollo Embrionario , Xenopus laevis , Animales , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Destrina/metabolismo , Destrina/genética , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Cresta Neural/metabolismo , Cresta Neural/embriología , Cresta Neural/citología , Neurogénesis , Placa Neural/metabolismo , Placa Neural/embriología , Actinas/metabolismo , Regulación del Desarrollo de la Expresión Génica
6.
Dev Growth Differ ; 66(5): 320-328, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38925637

RESUMEN

During the formation of the neural tube, the primordium of the vertebrate central nervous system, the actomyosin activity of cells in different regions drives neural plate bending. However, how the stiffness of the neural plate and surrounding tissues is regulated and mechanically influences neural plate bending has not been elucidated. Here, we used atomic force microscopy to reveal the relationship between the stiffness of the neural plate and the mesoderm during Xenopus neural tube formation. Measurements with intact embryos revealed that the stiffness of the neural plate was consistently higher compared with the non-neural ectoderm and that it increased in an actomyosin activity-dependent manner during neural plate bending. Interestingly, measurements of isolated tissue explants also revealed that the relationship between the stiffness of the apical and basal sides of the neural plate was reversed during bending and that the stiffness of the mesoderm was lower than that of the basal side of the neural plate. The experimental elevation of mesoderm stiffness delayed neural plate bending, suggesting that low mesoderm stiffness mechanically supports neural tube closure. This study provides an example of mechanical interactions between tissues during large-scale morphogenetic movements.


Asunto(s)
Placa Neural , Tubo Neural , Xenopus laevis , Animales , Tubo Neural/embriología , Tubo Neural/citología , Tubo Neural/metabolismo , Placa Neural/embriología , Placa Neural/metabolismo , Placa Neural/citología , Xenopus laevis/embriología , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ectodermo/citología , Ectodermo/metabolismo , Microscopía de Fuerza Atómica , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Embrión no Mamífero/embriología
7.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38851297

RESUMEN

The development of the human central nervous system initiates in the early embryonic period until long after delivery. It has been shown that several neurological and neuropsychiatric diseases originate from prenatal incidents. Mathematical models offer a direct way to understand neurodevelopmental processes better. Mathematical modelling of neurodevelopment during the embryonic period is challenging in terms of how to 'Approach', how to initiate modelling and how to propose the appropriate equations that fit the underlying dynamics of neurodevelopment during the embryonic period while including the variety of elements that are built-in naturally during the process of neurodevelopment. It is imperative to answer where and how to start modelling; in other words, what is the appropriate 'Approach'? Therefore, one objective of this study was to tackle the mathematical issue broadly from different aspects and approaches. The approaches were divided into three embryonic categories: cell division, neural tube growth and neural plate growth. We concluded that the neural plate growth approach provides a suitable platform for simulation of brain formation/neurodevelopment compared to cell division and neural tube growth. We devised a novel equation and designed algorithms that include geometrical and topological algorithms that could fit most of the necessary elements of the neurodevelopmental process during the embryonic period. Hence, the proposed equations and defined mathematical structure would be a platform to generate an artificial neural network that autonomously grows and develops.


Asunto(s)
Modelos Biológicos , Tubo Neural , Animales , Humanos , Algoritmos , División Celular , Desarrollo Embrionario , Modelos Neurológicos , Redes Neurales de la Computación , Placa Neural/citología , Placa Neural/embriología , Tubo Neural/embriología , Neurogénesis , Neuronas/citología
8.
Development ; 151(14)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38940470

RESUMEN

SoxB1 transcription factors (Sox2/3) are well known for their role in early neural fate specification in the embryo, but little is known about functional roles for SoxB1 factors in non-neural ectodermal cell types, such as the neural plate border (NPB). Using Xenopus laevis, we set out to determine whether SoxB1 transcription factors have a regulatory function in NPB formation. Here, we show that SoxB1 factors are necessary for NPB formation, and that prolonged SoxB1 factor activity blocks the transition from a NPB to a neural crest state. Using ChIP-seq, we demonstrate that Sox3 is enriched upstream of NPB genes in early NPB cells and in blastula stem cells. Depletion of SoxB1 factors in blastula stem cells results in downregulation of NPB genes. Finally, we identify Pou5f3 factors as potential Sox3 partners in regulating the formation of the NPB and show that their combined activity is needed for normal NPB gene expression. Together, these data identify a role for SoxB1 factors in the establishment and maintenance of the NPB, in part through partnership with Pou5f3 factors.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Cresta Neural , Placa Neural , Factores de Transcripción SOXB1 , Proteínas de Xenopus , Xenopus laevis , Animales , Placa Neural/metabolismo , Placa Neural/embriología , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXB1/genética , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Cresta Neural/metabolismo , Cresta Neural/citología , Blástula/metabolismo , Embrión no Mamífero/metabolismo
9.
Proc Natl Acad Sci U S A ; 121(19): e2311685121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38683994

RESUMEN

Neural crest cells exemplify cellular diversification from a multipotent progenitor population. However, the full sequence of early molecular choices orchestrating the emergence of neural crest heterogeneity from the embryonic ectoderm remains elusive. Gene-regulatory-networks (GRN) govern early development and cell specification toward definitive neural crest. Here, we combine ultradense single-cell transcriptomes with machine-learning and large-scale transcriptomic and epigenomic experimental validation of selected trajectories, to provide the general principles and highlight specific features of the GRN underlying neural crest fate diversification from induction to early migration stages using Xenopus frog embryos as a model. During gastrulation, a transient neural border zone state precedes the choice between neural crest and placodes which includes multiple converging gene programs. During neurulation, transcription factor connectome, and bifurcation analyses demonstrate the early emergence of neural crest fates at the neural plate stage, alongside an unbiased multipotent-like lineage persisting until epithelial-mesenchymal transition stage. We also decipher circuits driving cranial and vagal neural crest formation and provide a broadly applicable high-throughput validation strategy for investigating single-cell transcriptomes in vertebrate GRNs in development, evolution, and disease.


Asunto(s)
Cresta Neural , Análisis de la Célula Individual , Xenopus laevis , Animales , Cresta Neural/citología , Cresta Neural/metabolismo , Análisis de la Célula Individual/métodos , Xenopus laevis/embriología , Regulación del Desarrollo de la Expresión Génica , Movimiento Celular , Redes Reguladoras de Genes , Transcriptoma , Gastrulación , Placa Neural/metabolismo , Placa Neural/embriología , Placa Neural/citología , Transición Epitelial-Mesenquimal/genética , Embrión no Mamífero/metabolismo , Embrión no Mamífero/citología , Neurulación/genética , Neurulación/fisiología , Diferenciación Celular
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.
Elife ; 112022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35088714

RESUMEN

The epiblast of vertebrate embryos is comprised of neural and non-neural ectoderm, with the border territory at their intersection harboring neural crest and cranial placode progenitors. Here, we a generate single-cell atlas of the developing chick epiblast from late gastrulation through early neurulation stages to define transcriptional changes in the emerging 'neural plate border' as well as other regions of the epiblast. Focusing on the border territory, the results reveal gradual establishment of heterogeneous neural plate border signatures, including novel genes that we validate by fluorescent in situ hybridization. Developmental trajectory analysis infers that segregation of neural plate border lineages only commences at early neurulation, rather than at gastrulation as previously predicted. We find that cells expressing the prospective neural crest marker Pax7 contribute to multiple lineages, and a subset of premigratory neural crest cells shares a transcriptional signature with their border precursors. Together, our results suggest that cells at the neural plate border remain heterogeneous until early neurulation, at which time progenitors become progressively allocated toward defined neural crest and placode lineages. The data also can be mined to reveal changes throughout the developing epiblast.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Cresta Neural/embriología , Placa Neural/embriología , Neurulación/fisiología , Animales , Embrión de Pollo/citología , Pollos/fisiología , Estratos Germinativos/fisiología , Hibridación Fluorescente in Situ , Factor de Transcripción PAX7/análisis
12.
Dev Biol ; 483: 66-75, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34968443

RESUMEN

In recent years CRISPR-Cas9 knockouts (KO) have become increasingly ultilised to study gene function. MicroRNAs (miRNAs) are short non-coding RNAs, 20-22 nucleotides long, which affect gene expression through post-transcriptional repression. We previously identified miRNAs-196a and -219 as implicated in the development of Xenopus neural crest (NC). The NC is a multipotent stem-cell population, specified during early neurulation. Following EMT, NC cells migrate to various points in the developing embryo where they give rise to a number of tissues including parts of the peripheral nervous system, pigment cells and craniofacial skeleton. Dysregulation of NC development results in many diseases grouped under the term neurocristopathies. As miRNAs are so small, it is difficult to design CRISPR sgRNAs that reproducibly lead to a KO. We have therefore designed a novel approach using two guide RNAs to effectively 'drop out' a miRNA. We have knocked out miR-196a and miR-219 and compared the results to morpholino knockdowns (KD) of the same miRNAs. Validation of efficient CRISPR miRNA KO and phenotype analysis included use of whole-mount in situ hybridization of key NC and neural plate border markers such as Pax3, Xhe2, Sox10 and Snail2, q-RT-PCR and Sanger sequencing. To show specificity we have also rescued the knockout phenotype using miRNA mimics. MiRNA-219 and miR-196a KO's both show loss of NC, altered neural plate and hatching gland phenotypes. Tadpoles show gross craniofacial and pigment phenotypes.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Inactivación de Genes/métodos , MicroARNs/genética , Xenopus laevis/embriología , Xenopus laevis/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Hibridación in Situ/métodos , Morfolinos/genética , Cresta Neural/embriología , Cresta Neural/metabolismo , Placa Neural/embriología , Placa Neural/metabolismo , Neurulación/genética , Fenotipo , ARN Guía de Kinetoplastida/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
13.
Mol Cells ; 44(10): 723-735, 2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34711690

RESUMEN

Spemann organizer is a center of dorsal mesoderm and itself retains the mesoderm character, but it has a stimulatory role for neighboring ectoderm cells in becoming neuroectoderm in gastrula embryos. Goosecoid (Gsc) overexpression in ventral region promotes secondary axis formation including neural tissues, but the role of gsc in neural specification could be indirect. We examined the neural inhibitory and stimulatory roles of gsc in the same cell and neighboring cells contexts. In the animal cap explant system, Gsc overexpression inhibited expression of neural specific genes including foxd4l1.1, zic3, ncam, and neurod. Genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) and promoter analysis of early neural genes of foxd4l1.1 and zic3 were performed to show that the neural inhibitory mode of gsc was direct. Site-directed mutagenesis and serially deleted construct studies of foxd4l1.1 promoter revealed that Gsc directly binds within the foxd4l1.1 promoter to repress its expression. Conjugation assay of animal cap explants was also performed to demonstrate an indirect neural stimulatory role for gsc. The genes for secretory molecules, Chordin and Noggin, were up-regulated in gsc injected cells with the neural fate only achieved in gsc uninjected neighboring cells. These experiments suggested that gsc regulates neuroectoderm formation negatively when expressed in the same cell and positively in neighboring cells via soluble factors. One is a direct suppressive circuit of neural genes in gsc expressing mesoderm cells and the other is an indirect stimulatory circuit for neurogenesis in neighboring ectoderm cells via secreted BMP antagonizers.


Asunto(s)
Proteína Goosecoide/metabolismo , Placa Neural/embriología , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales
14.
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
15.
Biol Open ; 10(7)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34259326

RESUMEN

Coordinated polarization of cells in the tissue plane, known as planar cell polarity (PCP), is associated with a signaling pathway critical for the control of morphogenetic processes. Although the segregation of PCP components to opposite cell borders is believed to play a critical role in this pathway, whether PCP derives from egg polarity or preexistent long-range gradient, or forms in response to a localized cue, remains a challenging question. Here we investigate the Xenopus neural plate, a tissue that has been previously shown to exhibit PCP. By imaging Vangl2 and Prickle3, we show that PCP is progressively acquired in the neural plate and requires a signal from the posterior region of the embryo. Tissue transplantations indicated that PCP is triggered in the neural plate by a planar cue from the dorsal blastopore lip. The PCP cue did not depend on the orientation of the graft and was distinct from neural inducers. These observations suggest that neuroectodermal PCP is not instructed by a preexisting molecular gradient but induced by a signal from the dorsal blastopore lip.


Asunto(s)
Polaridad Celular/fisiología , Gástrula/embriología , Morfogénesis/fisiología , Placa Neural/embriología , Xenopus/embriología , Animales , Transducción de Señal
16.
Dev Biol ; 478: 59-75, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34029538

RESUMEN

Morphogenesis of the vertebrate neural tube occurs by elongation and bending of the neural plate, tissue shape changes that are driven at the cellular level by polarized cell intercalation and cell shape changes, notably apical constriction and cell wedging. Coordinated cell intercalation, apical constriction, and wedging undoubtedly require complex underlying cytoskeletal dynamics and remodeling of adhesions. Mutations of the gene encoding Scribble result in neural tube defects in mice, however the cellular and molecular mechanisms by which Scrib regulates neural cell behavior remain unknown. Analysis of Scribble mutants revealed defects in neural tissue shape changes, and live cell imaging of mouse embryos showed that the Scrib mutation results in defects in polarized cell intercalation, particularly in rosette resolution, and failure of both cell apical constriction and cell wedging. Scrib mutant embryos displayed aberrant expression of the junctional proteins ZO-1, Par3, Par6, E- and N-cadherins, and the cytoskeletal proteins actin and myosin. These findings show that Scribble has a central role in organizing the molecular complexes regulating the morphomechanical neural cell behaviors underlying vertebrate neurulation, and they advance our understanding of the molecular mechanisms involved in mammalian neural tube closure.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Defectos del Tubo Neural/embriología , Tubo Neural/embriología , Animales , Polaridad Celular , Forma de la Célula , Proteínas del Citoesqueleto , Expresión Génica , Uniones Intercelulares/metabolismo , Uniones Intercelulares/ultraestructura , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Morfogénesis , Mutación , Proteínas del Tejido Nervioso/genética , Placa Neural/citología , Placa Neural/embriología , Tubo Neural/citología , Defectos del Tubo Neural/genética , Células Neuroepiteliales/citología , Células Neuroepiteliales/metabolismo , Células Neuroepiteliales/ultraestructura , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
17.
Nat Commun ; 12(1): 2058, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824332

RESUMEN

Wnt signaling regulates cell proliferation and cell differentiation as well as migration and polarity during development. However, it is still unclear how the Wnt ligand distribution is precisely controlled to fulfil these functions. Here, we show that the planar cell polarity protein Vangl2 regulates the distribution of Wnt by cytonemes. In zebrafish epiblast cells, mouse intestinal telocytes and human gastric cancer cells, Vangl2 activation generates extremely long cytonemes, which branch and deliver Wnt protein to multiple cells. The Vangl2-activated cytonemes increase Wnt/ß-catenin signaling in the surrounding cells. Concordantly, Vangl2 inhibition causes fewer and shorter cytonemes to be formed and reduces paracrine Wnt/ß-catenin signaling. A mathematical model simulating these Vangl2 functions on cytonemes in zebrafish gastrulation predicts a shift of the signaling gradient, altered tissue patterning, and a loss of tissue domain sharpness. We confirmed these predictions during anteroposterior patterning in the zebrafish neural plate. In summary, we demonstrate that Vangl2 is fundamental to paracrine Wnt/ß-catenin signaling by controlling cytoneme behaviour.


Asunto(s)
Proteínas de la Membrana/metabolismo , Seudópodos/metabolismo , Vía de Señalización Wnt , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo , Embrión no Mamífero/metabolismo , Activación Enzimática , Fibroblastos/metabolismo , Gastrulación , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones Endogámicos C57BL , Placa Neural/embriología , Placa Neural/metabolismo , Neurogénesis , Comunicación Paracrina , Análisis de Sistemas , Telocitos/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo
18.
Dev Dyn ; 250(7): 955-973, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33501723

RESUMEN

BACKGROUND: Neural tube (NT) closure is a complex developmental process that takes place in the early stages of embryogenesis and that is a key step in neurulation. In mammals, the process by which the neural plate generates the NT requires organized cell movements and tissue folding, and it terminates with the fusion of the apposed ends of the neural folds. RESULTS: Here we describe how almost identical cellular and molecular machinery is used to fuse the spinal neural folds as that involved in the repair of epithelial injury in the same area of the embryo. For both natural and wound activated closure of caudal neural tissue, hyaluronic acid and platelet-derived growth factor signaling appear to be crucial for the final fusion step. CONCLUSIONS: There seems to be no general wound healing machinery for all tissues but rather, a tissue-specific epithelial fusion machinery that embryos activate when necessary after abnormal epithelial opening.


Asunto(s)
Células Epiteliales/fisiología , Tubo Neural/embriología , Neurulación/fisiología , Cicatrización de Heridas/fisiología , Animales , Fusión Celular , Células Cultivadas , Embrión de Mamíferos , Desarrollo Embrionario/fisiología , Células Epiteliales/citología , Femenino , Feto/embriología , Ácido Hialurónico/metabolismo , Masculino , Ratones , Cresta Neural/embriología , Cresta Neural/fisiología , Placa Neural/embriología , Placa Neural/fisiología , Defectos del Tubo Neural/embriología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Embarazo
19.
Commun Biol ; 4(1): 147, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514864

RESUMEN

Primary neurulation is the process by which the neural tube, the central nervous system precursor, is formed from the neural plate. Incomplete neural tube closure occurs frequently, yet underlying causes remain poorly understood. Developmental studies in amniotes and amphibians have identified hingepoint and neural fold formation as key morphogenetic events and hallmarks of primary neurulation, the disruption of which causes neural tube defects. In contrast, the mode of neurulation in teleosts has remained highly debated. Teleosts are thought to have evolved a unique mode of neurulation, whereby the neural plate infolds in absence of hingepoints and neural folds, at least in the hindbrain/trunk where it has been studied. Using high-resolution imaging and time-lapse microscopy, we show here the presence of these morphological landmarks in the zebrafish anterior neural plate. These results reveal similarities between neurulation in teleosts and other vertebrates and hence the suitability of zebrafish to understand human neurulation.


Asunto(s)
Células Epiteliales/fisiología , Placa Neural/embriología , Tubo Neural/embriología , Neurulación , Prosencéfalo/embriología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Movimiento Celular , Forma de la Célula , Células Epiteliales/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Morfogénesis , Placa Neural/metabolismo , Tubo Neural/metabolismo , Defectos del Tubo Neural/embriología , Prosencéfalo/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Imagen de Lapso de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
20.
Int J Dev Biol ; 65(4-5-6): 263-273, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32930349

RESUMEN

The internalization of multi-cellular tissues is a key morphogenetic process during animal development and organ formation. A good example of this is the initial stages of vertebrate central nervous system formation whereby a transient embryonic structure called the neural plate is able to undergo collective cell rearrangements within the dorsal midline. Despite the fact that defects in neural plate midline internalization may result in a series of severe clinical conditions, such as spina bifida and anencephaly, the biochemical and biomechanical details of this process remain only partially characterized. Here we review the main cellular and molecular mechanisms underlying midline cell and tissue internalization during vertebrate neural tube formation. We discuss the contribution of collective cell mechanisms including convergence and extension, as well as apical constriction facilitating midline neural plate shaping. Furthermore, we summarize recent studies that shed light on how the interplay of signaling pathways and cell biomechanics modulate neural plate internalization. In addition, we discuss how adhesion-dependent cell-cell contact appears to be a critical component during midline cell convergence and surface cell contraction via cell-cell mechanical coupling. We envision that more detailed high-resolution quantitative data at both cell and tissue levels will be required to properly model the mechanisms of vertebrate neural plate internalization with the hope of preventing human neural tube defects.


Asunto(s)
Placa Neural , Tubo Neural , Vertebrados/embriología , Animales , Morfogénesis , Placa Neural/embriología , Tubo Neural/embriología , Neurulación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA