Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Dev Biol ; 511: 76-83, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38614285

RESUMO

This paper introduces a single-cell atlas for pivotal developmental stages in Xenopus, encompassing gastrulation, neurulation, and early tailbud. Notably surpassing its predecessors, the new atlas enhances gene mapping, read counts, and gene/cell type nomenclature. Leveraging the latest Xenopus tropicalis genome version, alongside advanced alignment pipelines and machine learning for cell type assignment, this release maintains consistency with previous cell type annotations while rectifying nomenclature issues. Employing an unbiased approach for cell type assignment proves especially apt for embryonic contexts, given the considerable number of non-terminally differentiated cell types. An alternative cell type attribution here adopts a fuzzy, non-deterministic stance, capturing the transient nature of early embryo progenitor cells by presenting an ensemble of types in superposition. The value of the new resource is emphasized through numerous examples, with a focus on previously unexplored germ cell populations where we uncover novel transcription onset features. Offering interactive exploration via a user-friendly web portal and facilitating complete data downloads, this atlas serves as a comprehensive and accessible reference.


Assuntos
Xenopus , Animais , Xenopus/embriologia , Xenopus/genética , Gastrulação , Embrião não Mamífero/citologia , Neurulação/genética , Neurulação/fisiologia , Análise de Célula Única/métodos , Regulação da Expressão Gênica no Desenvolvimento
2.
Proc Natl Acad Sci U S A ; 121(19): e2311685121, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38683994

RESUMO

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.


Assuntos
Crista Neural , Análise de Célula Única , Xenopus laevis , Animais , Crista Neural/citologia , Crista Neural/metabolismo , Análise de Célula Única/métodos , Xenopus laevis/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Movimento Celular , Redes Reguladoras de Genes , Transcriptoma , Gastrulação , Placa Neural/metabolismo , Placa Neural/embriologia , Placa Neural/citologia , Transição Epitelial-Mesenquimal/genética , Embrião não Mamífero/metabolismo , Embrião não Mamífero/citologia , Neurulação/genética , Neurulação/fisiologia , Diferenciação Celular
3.
Brain Behav Evol ; 99(1): 45-68, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38342091

RESUMO

BACKGROUND: The phylotypic or intermediate stages are thought to be the most evolutionary conserved stages throughout embryonic development. The contrast with divergent early and later stages derived from the concept of the evo-devo hourglass model. Nonetheless, this developmental constraint has been studied as a whole embryo process, not at organ level. In this review, we explore brain development to assess the existence of an equivalent brain developmental hourglass. In the specific case of vertebrates, we propose to split the brain developmental stages into: (1) Early: Neurulation, when the neural tube arises after gastrulation. (2) Intermediate: Brain patterning and segmentation, when the neuromere identities are established. (3) Late: Neurogenesis and maturation, the stages when the neurons acquire their functionality. Moreover, we extend this analysis to other chordates brain development to unravel the evolutionary origin of this evo-devo constraint. SUMMARY: Based on the existing literature, we hypothesise that a major conservation of the phylotypic brain might be due to the pleiotropy of the inductive regulatory networks, which are predominantly expressed at this stage. In turn, earlier stages such as neurulation are rather mechanical processes, whose regulatory networks seem to adapt to environment or maternal geometries. The later stages are also controlled by inductive regulatory networks, but their effector genes are mostly tissue-specific and functional, allowing diverse developmental programs to generate current brain diversity. Nonetheless, all stages of the hourglass are highly interconnected: divergent neurulation must have a vertebrate shared end product to reproduce the vertebrate phylotypic brain, and the boundaries and transcription factor code established during the highly conserved patterning will set the bauplan for the specialised and diversified adult brain. KEY MESSAGES: The vertebrate brain is conserved at phylotypic stages, but the highly conserved mechanisms that occur during these brain mid-development stages (Inducing Regulatory Networks) are also present during other stages. Oppositely, other processes as cell interactions and functional neuronal genes are more diverse and majoritarian in early and late stages of development, respectively. These phenomena create an hourglass of transcriptomic diversity during embryonic development and evolution, with a really conserved bottleneck that set the bauplan for the adult brain around the phylotypic stage.


Assuntos
Evolução Biológica , Encéfalo , Tubo Neural , Vertebrados , Animais , Vertebrados/embriologia , Vertebrados/crescimento & desenvolvimento , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Tubo Neural/embriologia , Neurogênese/fisiologia , Neurulação/fisiologia
4.
Emerg Top Life Sci ; 7(4): 423-437, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38087891

RESUMO

Neurulation is a critical step in early embryonic development, giving rise to the neural tube, the primordium of the central nervous system in amniotes. Understanding this complex, multi-scale, multi-tissue morphogenetic process is essential to provide insights into normal development and the etiology of neural tube defects. Innovations in tissue engineering have fostered the generation of pluripotent stem cell-based in vitro models, including organoids, that are emerging as unique tools for delving into neurulation mechanisms, especially in the context of human development. Each model captures specific aspects of neural tube morphogenesis, from epithelialization to neural tissue elongation, folding and cavitation. In particular, the recent models of human and mouse trunk morphogenesis, such as gastruloids, that form a spinal neural plate-like or neural tube-like structure are opening new avenues to study normal and pathological neurulation. Here, we review the morphogenetic events generating the neural tube in the mammalian embryo and questions that remain unanswered. We discuss the advantages and limitations of existing in vitro models of neurulation and possible future technical developments.


Assuntos
Defeitos do Tubo Neural , Neurulação , Camundongos , Animais , Humanos , Neurulação/fisiologia , Tubo Neural , Placa Neural , Células-Tronco , Mamíferos
5.
Dev Biol ; 491: 105-112, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36113571

RESUMO

During neural tube closure, neural ectoderm cells constrict their apical surfaces to bend and fold the tissue into a tube that will become the central nervous system. Recent data from mice and humans with neural tube defects suggest that key genes required for neural tube closure can exert non-cell autonomous effects on cell behavior, but the nature of these effects remains obscure. Here, we coupled tissue-scale, high-resolution time-lapse imaging of the closing neural tube of Xenopus to multivariate regression modeling, and we show that medial actin accumulation drives apical constriction non-autonomously in neighborhoods of cells, rather than solely in individual cells. To further explore this effect, we examined mosaic crispant embryos and identified both autonomous and non-autonomous effects of the apical constriction protein Shroom3.


Assuntos
Actinas , Tubo Neural , Actinas/metabolismo , Animais , Humanos , Camundongos , Proteínas dos Microfilamentos/metabolismo , Morfogênese , Tubo Neural/metabolismo , Neurulação/fisiologia , Análise de Regressão
6.
Development ; 149(13)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35662330

RESUMO

Neural tube closure (NTC) is a fundamental process during vertebrate development and is indispensable for the formation of the central nervous system. Here, using Xenopus laevis embryos, live imaging, single-cell tracking, optogenetics and loss-of-function experiments, we examine the roles of convergent extension and apical constriction, and define the role of the surface ectoderm during NTC. We show that NTC is a two-stage process with distinct spatiotemporal contributions of convergent extension and apical constriction at each stage. Convergent extension takes place during the first stage and is spatially restricted at the posterior tissue, whereas apical constriction occurs during the second stage throughout the neural plate. We also show that the surface ectoderm is mechanically coupled with the neural plate and its movement during NTC is driven by neural plate morphogenesis. Finally, we show that an increase in surface ectoderm resistive forces is detrimental for neural plate morphogenesis.


Assuntos
Tubo Neural , Neurulação , Animais , Morfogênese/fisiologia , Placa Neural , Neurulação/fisiologia , Xenopus laevis
7.
Proc Natl Acad Sci U S A ; 119(20): e2117075119, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35561223

RESUMO

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.


Assuntos
Tubo Neural , Neurulação , Animais , Ectoderma/embriologia , Humanos , Camundongos , Placa Neural/embriologia , Tubo Neural/embriologia , Neurulação/fisiologia , Notocorda/embriologia
8.
Nat Biomed Eng ; 6(4): 435-448, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35347276

RESUMO

Human spinal-cord-like tissues induced from human pluripotent stem cells are typically insufficiently mature and do not mimic the morphological features of neurulation. Here, we report a three-dimensional culture system and protocol for the production of human spinal-cord-like organoids (hSCOs) recapitulating the neurulation-like tube-forming morphogenesis of the early spinal cord. The hSCOs exhibited neurulation-like tube-forming morphogenesis, cellular differentiation into the major types of spinal-cord neurons as well as glial cells, and mature synaptic functional activities, among other features of the development of the spinal cord. We used the hSCOs to screen for antiepileptic drugs that can cause neural-tube defects. hSCOs may also facilitate the study of the development of the human spinal cord and the modelling of diseases associated with neural-tube defects.


Assuntos
Defeitos do Tubo Neural , Neurulação , Humanos , Morfogênese/fisiologia , Neurulação/fisiologia , Organoides , Medula Espinal
9.
Elife ; 112022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35088714

RESUMO

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.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/embriologia , Placa Neural/embriologia , Neurulação/fisiologia , Animais , Embrião de Galinha/citologia , Galinhas/fisiologia , Camadas Germinativas/fisiologia , Hibridização in Situ Fluorescente , Fator de Transcrição PAX7/análise
10.
Nat Commun ; 12(1): 1159, 2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33608529

RESUMO

Post-zygotic mutations that generate tissue mosaicism are increasingly associated with severe congenital defects, including those arising from failed neural tube closure. Here we report that neural fold elevation during mouse spinal neurulation is vulnerable to deletion of the VANGL planar cell polarity protein 2 (Vangl2) gene in as few as 16% of neuroepithelial cells. Vangl2-deleted cells are typically dispersed throughout the neuroepithelium, and each non-autonomously prevents apical constriction by an average of five Vangl2-replete neighbours. This inhibition of apical constriction involves diminished myosin-II localisation on neighbour cell borders and shortening of basally-extending microtubule tails, which are known to facilitate apical constriction. Vangl2-deleted neuroepithelial cells themselves continue to apically constrict and preferentially recruit myosin-II to their apical cell cortex rather than to apical cap localisations. Such non-autonomous effects can explain how post-zygotic mutations affecting a minority of cells can cause catastrophic failure of morphogenesis leading to clinically important birth defects.


Assuntos
Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Defeitos do Tubo Neural/genética , Neurulação/genética , Neurulação/fisiologia , Citoesqueleto de Actina/metabolismo , Animais , Polaridade Celular/fisiologia , Modelos Animais de Doenças , Deleção de Genes , Camundongos , Morfogênese/genética , Morfogênese/fisiologia , Mutação , Miosina Tipo II/metabolismo , Crista Neural/metabolismo , Células Neuroepiteliais/metabolismo , Células Neuroepiteliais/patologia , Transcriptoma
11.
Dev Dyn ; 250(7): 955-973, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33501723

RESUMO

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.


Assuntos
Células Epiteliais/fisiologia , Tubo Neural/embriologia , Neurulação/fisiologia , Cicatrização/fisiologia , Animais , Fusão Celular , Células Cultivadas , Embrião de Mamíferos , Desenvolvimento Embrionário/fisiologia , Células Epiteliais/citologia , Feminino , Feto/embriologia , Ácido Hialurônico/metabolismo , Masculino , Camundongos , Crista Neural/embriologia , Crista Neural/fisiologia , Placa Neural/embriologia , Placa Neural/fisiologia , Defeitos do Tubo Neural/embriologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Gravidez
12.
Genome Biol ; 22(1): 43, 2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33482885

RESUMO

BACKGROUND: Microexons, exons that are ≤ 30 nucleotides, are a highly conserved and dynamically regulated set of cassette exons. They have key roles in nervous system development and function, as evidenced by recent results demonstrating the impact of microexons on behaviour and cognition. However, microexons are often overlooked due to the difficulty of detecting them using standard RNA-seq aligners. RESULTS: Here, we present MicroExonator, a novel pipeline for reproducible de novo discovery and quantification of microexons. We process 289 RNA-seq datasets from eighteen mouse tissues corresponding to nine embryonic and postnatal stages, providing the most comprehensive survey of microexons available for mice. We detect 2984 microexons, 332 of which are differentially spliced throughout mouse embryonic brain development, including 29 that are not present in mouse transcript annotation databases. Unsupervised clustering of microexons based on their inclusion patterns segregates brain tissues by developmental time, and further analysis suggests a key function for microexons in axon growth and synapse formation. Finally, we analyse single-cell RNA-seq data from the mouse visual cortex, and for the first time, we report differential inclusion between neuronal subpopulations, suggesting that some microexons could be cell type-specific. CONCLUSIONS: MicroExonator facilitates the investigation of microexons in transcriptome studies, particularly when analysing large volumes of data. As a proof of principle, we use MicroExonator to analyse a large collection of both mouse bulk and single-cell RNA-seq datasets. The analyses enabled the discovery of previously uncharacterized microexons, and our study provides a comprehensive microexon inclusion catalogue during mouse development.


Assuntos
Desenvolvimento Embrionário/genética , Éxons , Neurônios/metabolismo , Animais , Sequência de Bases , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Camundongos , Neurogênese/genética , Neurulação/genética , Neurulação/fisiologia , Splicing de RNA , Análise de Sequência de RNA , Análise de Célula Única , Software , Transcriptoma , Córtex Visual , Peixe-Zebra
13.
Neurotoxicol Teratol ; 83: 106941, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33212164

RESUMO

Nuclear factor kappa B (NF-κB) is a heterodimer of protein subunits p65 and p50, that regulates the expression of a large number of genes related to cell growth and proliferation. The p65 subunit is activated after phosphorylation by Pim-1, while the p50 subunit is the cleaved product of its precursor molecule p105. Valproic acid (VPA), an antiepileptic drug, is a known teratogen and its exposure during pregnancy is associated with 1-2% of neural tube defects in the offspring. The current study aimed at investigating the effects of in utero VPA exposure on the key components of the NF-κB signaling pathway including p65, p50, and Pim-1 in CD-1 mouse embryos during the critical period of neural tube closure. Here we report that p65, Pim-1 and p105/p50 mRNA were significantly (p < 0.05) downregulated at 1 and 3 h following in utero exposure to a teratogenic dose (400 mg/kg) of VPA in gestational day (GD)9 exposed embryos. At GD13 heads of control, non-exencephalic and exencephalic embryos were used for analysis and we found significant upregulation of p65 protein expression in non-exencephalic GD13 heads while p50 protein levels were significantly downregulated in both non-exencephalic and exencephalic groups. On the other hand, p65 and p50 protein levels remained unchanged in the nuclear extracts of the VPA-exposed non-exencephalic and exencephalic GD13 embryo heads. The reported results suggest that VPA exposure perturbates p65, p105/p50, Pim-1 transcript and p65/p50 protein levels in mouse embryos.


Assuntos
NF-kappa B/metabolismo , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Ácido Valproico/toxicidade , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/toxicidade , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Troca Materno-Fetal , Camundongos , NF-kappa B/genética , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Tubo Neural/metabolismo , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/metabolismo , Neurotoxinas/administração & dosagem , Neurotoxinas/toxicidade , Neurulação/efeitos dos fármacos , Neurulação/fisiologia , Gravidez , Proteínas Proto-Oncogênicas c-pim-1/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Ácido Valproico/administração & dosagem
14.
Sci Rep ; 10(1): 13752, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32792568

RESUMO

Sproutys are negative regulators of the Ras/Raf/MAPK signaling pathway and involved in regulation of organogenesis, differentiation, cell migration and proliferation. Although the function of Sproutys have been extensively studied during embryonic development, their role and mode of action during eye formation in vertebrate embryonic development is still unknown. Here we show that Xenopus sprouty2 is expressed in the optic vesicle at late neurula stage and knockdown of Sprouty2 prevents retinal progenitors from populating the retina, which in turn gives rise to small eyes. In the absence of Sprouty2, progenitor cell population of the retina can be restored by blocking the MAPK signaling pathway through overexpression of DN-Ras or DN-Raf. In contrast, activation of the MAPK pathway through overexpression of a constitutively active form of c-Raf (ca-Raf) inhibits progenitor population of the retina, similar to the Sprouty2 loss-of-function phenotype. Moreover, we present evidence that the retinal defect observed in Sprouty2 morphants is attributed to the failure of proper movement of retinal progenitors into the optic vesicle, rather than an effect on progenitor cell survival. These results suggest that Sprouty2 is required for the positioning of retinal progenitors within the optic vesicle through suppressing Ras/Raf/MAPK signaling pathway.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Retina/embriologia , Células-Tronco/citologia , Proteínas de Xenopus/genética , Xenopus laevis/embriologia , Animais , Diferenciação Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurulação/fisiologia , Proteínas Proto-Oncogênicas c-raf/metabolismo , Retina/citologia , Proteínas de Xenopus/metabolismo
15.
Rev. medica electron ; 42(4): 2049-2065,
Artigo em Espanhol | LILACS, CUMED | ID: biblio-1139295

RESUMO

RESUMEN Las células de la cresta neural son pluripotenciales y son llamadas la cuarta hoja germinativa del embrión. Con el objetivo de estructurar los referentes teóricos actualizados que sustenten la afirmación precedente y que constituirá material de estudio para los estudiantes de las Ciencias Médicas, se realizó la revisión de 28 referencias bibliográficas, de ellas 89% actualizadas. Estas células aparecen durante la neurulación y pasado este proceso transitan de epitelial a mesenquimatosa; migran siguiendo señales de la matriz extracelular a todo el cuerpo del embrión diferenciándose en tejidos disimiles. Muy vinculados en su evolución a mecanismos epigenéticos, hacen a esta población celular vulnerables a ser dañadas invocándose en la etiología de diferentes defectos congénitos y enfermedades crónicas no trasmisibles como cáncer. Como conclusión por su pluripotencialidad y por los mecanismos moleculares que distinguen su evolución son consideradas por muchos autores la cuarta hoja germinativa del embrión (AU).


SUMMARY Neural crest cells are pluripotentials, and are called the fourth germinative leaf of the embryo. With the objective of structuring the updated theoretical referents backing up the precedent affirmation that will be study material for the students of Medical Sciences, the authors reviewed 28 bibliographic references, 89 % of them updated. These cells appear during neurulation and after this process they transit from epithelial to mesenchymal; following extracellular matrix signals, they migrate to the whole embryo body differentiating themselves in dissimilar tissues. Tightly related in their evolution to epigenetic mechanisms, this cell population is very likely to be damaged and so they are invoked in the etiology of different congenital defects and noncommunicable chronic diseases like cancer. In conclusion, due to their pluripotentiality and the molecular mechanisms distinguishing their evolution, many authors consider them the embryo´s fourth germinative leaf (AU).


Assuntos
Humanos , Masculino , Feminino , Células/metabolismo , Crista Neural/patologia , Estudantes de Medicina , Vertebrados/genética , Neurulação/fisiologia , Crista Neural/anormalidades , Crista Neural/fisiologia , Crista Neural/fisiopatologia
16.
Dev Biol ; 461(2): 160-171, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32059837

RESUMO

In amniotes, unlike primary neurulation in the anterior body, secondary neurulation (SN) proceeds along with axial elongation by the mesenchymal-to-epithelial transition of SN precursors in the tail bud. It has been under debate whether the SN is generated by neuromesodermal common progenitor cells (NMPs) or neural restricted lineage. Our direct cell labeling and serial transplantations identify uni-fated (neural) precursors in the early tail bud. The uni-fated SN precursor territory is further divided into two subpopulations, neural-differentiating and self-renewing cells, which are regulated by high- and low levels of Sox2, respectively. Unexpectedly, uni-fated SN precursors change their fate at later stages to produce both SN and mesoderm. Thus, chicken embryos adopt a previously unappreciated prolonged phase with uni-fated SN stem cells in the early tail bud, which is absent or very limited in mouse embryos.


Assuntos
Autorrenovação Celular/fisiologia , Galinhas/genética , Células-Tronco Neurais/citologia , Tubo Neural/embriologia , Neurulação/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Cauda/embriologia , Animais , Linhagem da Célula , Embrião de Galinha , Genes Reporter , Mesoderma/citologia , Tubo Neural/citologia , Neurulação/genética , Fatores de Transcrição SOXB1/antagonistas & inibidores , Fatores de Transcrição SOXB1/genética , Cauda/citologia
17.
Neurosurgery ; 86(1): 93-100, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30690520

RESUMO

BACKGROUND: Limited dorsal myeloschisis (LDM) is postulated to be a result of incomplete dysjunction in primary neurulation. However, clinical experience of LDM located below the first-second sacral (S1-S2) vertebral level, which is formed from secondary neurulation (S2-coccyx), suggested that LDM may not be entirely explained as an error of primary neurulation. OBJECTIVE: To elucidate the location and characteristics of LDM to investigate the possible relation of its pathoembryogenesis to secondary neurulation. METHODS: Twenty-eight patients were surgically treated for LDM from 2010 to 2015. Since the level where the LDM stalk penetrates the interspinous ligament is most clearly defined on the preoperative MRI and operative field, this level was assessed to find out whether the lesions can occur in the region of secondary neurulation. RESULTS: Eleven patients (39%) with typical morphology of the stalk had interspinous defect levels lower than S1-S2. These patients were not different from 17 patients with classic LDMs at a level above or at S1-S2. This result shows that other than the low level of the interspinous level, 11 patients had lesions that could be defined as LDMs. CONCLUSION: By elucidating the location of LDM lesions (in particular, the interspinous level), we propose that LDM may be caused by errors of secondary neurulation. The hypothesis seems more plausible due to the supportive fact that the process of separation between the cutaneous and neural ectoderm is present during secondary neurulation. Hence, incomplete disjunction of the two ectoderms during secondary neurulation may result in LDM, similar to the pathomechanism proposed during primary neurulation.


Assuntos
Neurulação/fisiologia , Anormalidades da Pele/diagnóstico por imagem , Anormalidades da Pele/cirurgia , Disrafismo Espinal/diagnóstico por imagem , Disrafismo Espinal/cirurgia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Ligamentos Articulares/diagnóstico por imagem , Ligamentos Articulares/embriologia , Ligamentos Articulares/cirurgia , Imageamento por Ressonância Magnética/métodos , Masculino , Sacro/diagnóstico por imagem , Sacro/embriologia , Sacro/cirurgia
18.
Birth Defects Res ; 112(2): 205-211, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31758757

RESUMO

BACKGROUND: Neural tube defects (NTDs) result from failure of neural tube closure during embryogenesis. These severe birth defects of the central nervous system include anencephaly and spina bifida, and affect 0.5-2 per 1,000 pregnancies worldwide in humans. It has been demonstrated that acetylation plays a pivotal role during neural tube closure, as animal models for defective histone acetyltransferase proteins display NTDs. Acetylation represents an important component of the complex network of posttranslational regulatory interactions, suggesting a possible fundamental role during primary neurulation events. This study aimed to assess protein acetylation contribution to early patterning of the central nervous system both in human and murine specimens. METHODS: We used both human and mouse (Cited2 -/- ) samples to analyze the dynamic acetylation of proteins during embryo development through immunohistochemistry, western blot analysis and quantitative polymerase chain reaction. RESULTS: We report the dynamic profile of histone and protein acetylation status during neural tube closure. We also report a rescue effect in an animal model by chemical p53 inhibition. CONCLUSIONS: Our data suggest that the p53-acetylation equilibrium may play a role in primary neurulation in mammals.


Assuntos
Defeitos do Tubo Neural/embriologia , Neurulação/genética , Acetilação , Anencefalia/etiologia , Anencefalia/fisiopatologia , Animais , Modelos Animais de Doenças , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Histona Acetiltransferases/metabolismo , Humanos , Mamíferos , Camundongos/embriologia , Neurulação/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Disrafismo Espinal/etiologia , Disrafismo Espinal/fisiopatologia , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Dis Model Mech ; 12(11)2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31628096

RESUMO

Encephalocele is a clinically important birth defect that can lead to severe disability in childhood and beyond. The embryonic and early fetal pathogenesis of encephalocele is poorly understood and, although usually classified as a 'neural tube defect', there is conflicting evidence on whether encephalocele results from defective neural tube closure or is a post-neurulation defect. It is also unclear whether encephalocele can result from the same causative factors as anencephaly and open spina bifida, or whether it is aetiologically distinct. This lack of information results largely from the scarce availability of animal models of encephalocele, particularly ones that resemble the commonest, nonsyndromic human defects. Here, we report a novel mouse model of occipito-parietal encephalocele, in which the small GTPase Rac1 is conditionally ablated in the (non-neural) surface ectoderm. Most mutant fetuses have open spina bifida, and some also exhibit exencephaly/anencephaly. However, a proportion of mutant fetuses exhibit brain herniation, affecting the occipito-parietal region and closely resembling encephalocele. The encephalocele phenotype does not result from defective neural tube closure, but rather from a later disruption of the surface ectoderm covering the already closed neural tube, allowing the brain to herniate. The neuroepithelium itself shows no downregulation of Rac1 and appears morphologically normal until late gestation. A large skull defect overlies the region of brain herniation. Our work provides a new genetic model of occipito-parietal encephalocele, particularly resembling nonsyndromic human cases. Although encephalocele has a different, later-arising pathogenesis than open neural tube defects, both can share the same genetic causation.


Assuntos
Modelos Animais de Doenças , Encefalocele/etiologia , Defeitos do Tubo Neural/etiologia , Neurulação/fisiologia , Animais , Proteínas de Ligação a DNA/fisiologia , Ectoderma/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/fisiologia , Fatores de Transcrição/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia
20.
Dev Cell ; 51(2): 158-172.e4, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31639367

RESUMO

Embryos control force generation at tissue boundaries, but how they do so remains poorly understood. Here we show how tissue-specific expression of the type II cadherin, Cadherin2, patterns actomyosin contractility along tissue boundaries to control zippering and neural tube closure in the basal chordate, Ciona robusta. Cadherin2 is differentially expressed and homotypically enriched in neural cells along the neural/epidermal (Ne/Epi) boundary, where RhoA and myosin are activated during zipper progression. Homotypically enriched Cadherin2 sequesters the Rho GTPase-activating protein, Gap21/23, to homotypic junctions. Gap21/23 in turn redirects RhoA/myosin activity to heterotypic Ne/Epi junctions. By activating myosin II along Ne/Epi junctions ahead of the zipper and inhibiting myosin II along newly formed Ne/Ne junctions behind the zipper, Cadherin2 promotes tissue-level contractile asymmetry to drive zipper progression. We propose that dynamic coupling of junction exchange to local changes in contractility may control fusion and separation of epithelia in many other contexts.


Assuntos
Citoesqueleto de Actina/metabolismo , Caderinas/metabolismo , Miosina Tipo II/metabolismo , Tubo Neural/metabolismo , Actomiosina/metabolismo , Junções Aderentes/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Neurulação/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...