Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
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
2.
Dev Biol ; 511: 76-83, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38614285

RESUMEN

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.


Asunto(s)
Xenopus , Animales , Xenopus/embriología , Xenopus/genética , Gastrulación , Embrión no Mamífero/citología , Neurulación/genética , Neurulación/fisiología , Análisis de la Célula Individual/métodos , Regulación del Desarrollo de la Expresión Génica
3.
Development ; 144(22): 4183-4194, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29038306

RESUMEN

Neural crest (NC) specification comprises an early phase, initiating immature NC progenitors formation at neural plate stage, and a later phase at neural fold stage, resulting in a functional premigratory NC that is able to delaminate and migrate. We found that the NC gene regulatory network triggers upregulation of pfkfb4 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4) during this late specification phase. As shown in previous studies, PFKFB4 controls AKT signaling in gastrulas and glycolysis rate in adult cells. Here, we focus on PFKFB4 function in NC during and after neurulation, using time-controlled or hypomorph depletions in vivo We find that PFKFB4 is essential both for specification of functional premigratory NC and for its migration. PFKFB4-depleted embryos fail to activate n-cadherin and late NC specifiers, and exhibit severe migration defects resulting in craniofacial defects. AKT signaling mediates PFKFB4 function in NC late specification, whereas both AKT signaling and glycolysis regulate migration. These findings highlight novel and essential roles of PFKFB4 activity in later stages of NC development that are wired into the NC gene regulatory network.


Asunto(s)
Movimiento Celular , Cresta Neural/citología , Fosfofructoquinasa-2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Transición Epitelial-Mesenquimal , Cara/embriología , Glucólisis , Larva , Modelos Biológicos , Neuronas/citología , Neuronas/metabolismo , Neurulación , Cráneo/embriología , Xenopus laevis/embriología
4.
PLoS Biol ; 15(10): e2004045, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29049289

RESUMEN

During vertebrate neurulation, the embryonic ectoderm is patterned into lineage progenitors for neural plate, neural crest, placodes and epidermis. Here, we use Xenopus laevis embryos to analyze the spatial and temporal transcriptome of distinct ectodermal domains in the course of neurulation, during the establishment of cell lineages. In order to define the transcriptome of small groups of cells from a single germ layer and to retain spatial information, dorsal and ventral ectoderm was subdivided along the anterior-posterior and medial-lateral axes by microdissections. Principal component analysis on the transcriptomes of these ectoderm fragments primarily identifies embryonic axes and temporal dynamics. This provides a genetic code to define positional information of any ectoderm sample along the anterior-posterior and dorsal-ventral axes directly from its transcriptome. In parallel, we use nonnegative matrix factorization to predict enhanced gene expression maps onto early and mid-neurula embryos, and specific signatures for each ectoderm area. The clustering of spatial and temporal datasets allowed detection of multiple biologically relevant groups (e.g., Wnt signaling, neural crest development, sensory placode specification, ciliogenesis, germ layer specification). We provide an interactive network interface, EctoMap, for exploring synexpression relationships among genes expressed in the neurula, and suggest several strategies to use this comprehensive dataset to address questions in developmental biology as well as stem cell or cancer research.


Asunto(s)
Ectodermo/embriología , Cresta Neural/embriología , Neuronas/citología , Células Madre/metabolismo , Xenopus laevis/embriología , Algoritmos , Animales , Análisis por Conglomerados , Bases de Datos Genéticas , Ectodermo/metabolismo , Gastrulación/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Internet , Microdisección , Neoplasias/genética , Cresta Neural/metabolismo , Neurulación/genética , Análisis de Componente Principal , Factores de Tiempo , Transcriptoma/genética , Proteínas Wnt/metabolismo , Xenopus laevis/genética
5.
Dev Biol ; 444 Suppl 1: S144-S155, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29859890

RESUMEN

AKT signaling is an essential intracellular pathway controlling cell homeostasis, cell proliferation and survival, as well as cell migration and differentiation in adults. Alterations impacting the AKT pathway are involved in many pathological conditions in human disease. Similarly, during development, multiple transmembrane molecules, such as FGF receptors, PDGF receptors or integrins, activate AKT to control embryonic cell proliferation, migration, differentiation, and also cell fate decisions. While many studies in mouse embryos have clearly implicated AKT signaling in the differentiation of several neural crest derivatives, information on AKT functions during the earliest steps of neural crest development had remained relatively scarce until recently. However, recent studies on known and novel regulators of AKT signaling demonstrate that this pathway plays critical roles throughout the development of neural crest progenitors. Non-mammalian models such as fish and frog embryos have been instrumental to our understanding of AKT functions in neural crest development, both in neural crest progenitors and in the neighboring tissues. This review combines current knowledge acquired from all these different vertebrate animal models to describe the various roles of AKT signaling related to neural crest development in vivo. We first describe the importance of AKT signaling in patterning the tissues involved in neural crest induction, namely the dorsal mesoderm and the ectoderm. We then focus on AKT signaling functions in neural crest migration and differentiation.


Asunto(s)
Desarrollo Embrionario/fisiología , Cresta Neural/embriología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Tipificación del Cuerpo/fisiología , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Mesodermo/metabolismo , Modelos Animales , Cresta Neural/citología , Cresta Neural/patología , Neurogénesis , Transducción de Señal , Vertebrados/embriología
6.
Dev Biol ; 444 Suppl 1: S36-S46, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29852131

RESUMEN

The neural crest is induced at the edge between the neural plate and the nonneural ectoderm, in an area called the neural (plate) border, during gastrulation and neurulation. In recent years, many studies have explored how this domain is patterned, and how the neural crest is induced within this territory, that also participates to the prospective dorsal neural tube, the dorsalmost nonneural ectoderm, as well as placode derivatives in the anterior area. This review highlights the tissue interactions, the cell-cell signaling and the molecular mechanisms involved in this dynamic spatiotemporal patterning, resulting in the induction of the premigratory neural crest. Collectively, these studies allow building a complex neural border and early neural crest gene regulatory network, mostly composed by transcriptional regulations but also, more recently, including novel signaling interactions.


Asunto(s)
Cresta Neural/citología , Cresta Neural/metabolismo , Cresta Neural/fisiología , Animales , Evolución Biológica , Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/fisiología , Movimiento Celular , Embrión de Pollo , Ectodermo/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Melanocitos/citología , Sistema Nervioso/metabolismo , Placa Neural/metabolismo , Placa Neural/fisiología , Neurogénesis/fisiología , Neurulación/fisiología , Transducción de Señal , Vía de Señalización Wnt/fisiología , Proteínas de Xenopus/genética , Xenopus laevis/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
7.
Semin Cell Dev Biol ; 44: 87-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26410165

RESUMEN

The nine vertebrate PAX transcription factors (PAX1-PAX9) play essential roles during early development and organogenesis. Pax genes were identified in vertebrates using their homology with the Drosophila melanogaster paired gene DNA-binding domain. PAX1-9 functions are largely conserved throughout vertebrate evolution, in particular during central nervous system and neural crest development. The neural crest is a vertebrate invention, which gives rise to numerous derivatives during organogenesis, including neurons and glia of the peripheral nervous system, craniofacial skeleton and mesenchyme, the heart outflow tract, endocrine and pigment cells. Human and mouse spontaneous mutations as well as experimental analyses have evidenced the critical and diverse functions of PAX factors during neural crest development. Recent studies have highlighted the role of PAX3 and PAX7 in neural crest induction. Additionally, several PAX proteins - PAX1, 3, 7, 9 - regulate cell proliferation, migration and determination in multiple neural crest-derived lineages, such as cardiac, sensory, and enteric neural crest, pigment cells, glia, craniofacial skeleton and teeth, or in organs developing in close relationship with the neural crest such as the thymus and parathyroids. The diverse PAX molecular functions during neural crest formation rely on fine-tuned modulations of their transcriptional transactivation properties. These modulations are generated by multiple means, such as different roles for the various isoforms (formed by alternative splicing), or posttranslational modifications which alter protein-DNA binding, or carefully orchestrated protein-protein interactions with various co-factors which control PAX proteins activity. Understanding these regulations is the key to decipher the versatile roles of PAX transcription factors in neural crest development, differentiation and disease.


Asunto(s)
Cresta Neural/fisiología , Factores de Transcripción Paired Box/fisiología , Animales , Humanos , Cresta Neural/embriología , Cresta Neural/metabolismo , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo
8.
Dev Biol ; 386(2): 461-72, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24360906

RESUMEN

Neural crest development is orchestrated by a complex and still poorly understood gene regulatory network. Premigratory neural crest is induced at the lateral border of the neural plate by the combined action of signaling molecules and transcription factors such as AP2, Gbx2, Pax3 and Zic1. Among them, Pax3 and Zic1 are both necessary and sufficient to trigger a complete neural crest developmental program. However, their gene targets in the neural crest regulatory network remain unknown. Here, through a transcriptome analysis of frog microdissected neural border, we identified an extended gene signature for the premigratory neural crest, and we defined novel potential members of the regulatory network. This signature includes 34 novel genes, as well as 44 known genes expressed at the neural border. Using another microarray analysis which combined Pax3 and Zic1 gain-of-function and protein translation blockade, we uncovered 25 Pax3 and Zic1 direct targets within this signature. We demonstrated that the neural border specifiers Pax3 and Zic1 are direct upstream regulators of neural crest specifiers Snail1/2, Foxd3, Twist1, and Tfap2b. In addition, they may modulate the transcriptional output of multiple signaling pathways involved in neural crest development (Wnt, Retinoic Acid) through the induction of key pathway regulators (Axin2 and Cyp26c1). We also found that Pax3 could maintain its own expression through a positive autoregulatory feedback loop. These hierarchical inductions, feedback loops, and pathway modulations provide novel tools to understand the neural crest induction network.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Cresta Neural/embriología , Factores de Transcripción Paired Box/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Ensayo de Cambio de Movilidad Electroforética , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Hibridación in Situ , Análisis por Micromatrices , Factor de Transcripción PAX3 , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Xenopus laevis/genética
9.
Proc Natl Acad Sci U S A ; 108(1): 155-60, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21169220

RESUMEN

The neural crest (NC) emerges from combinatorial inductive events occurring within its progenitor domain, the neural border (NB). Several transcription factors act early at the NB, but the initiating molecular events remain elusive. Recent data from basal vertebrates suggest that ap2 might have been critical for NC emergence; however, the role of AP2 factors at the NB remains unclear. We show here that AP2a initiates NB patterning and is sufficient to elicit a NB-like pattern in neuralized ectoderm. In contrast, the other early regulators do not participate in ap2a initiation at the NB, but cooperate to further establish a robust NB pattern. The NC regulatory network uses a multistep cascade of secreted inducers and transcription factors, first at the NB and then within the NC progenitors. Here we report that AP2a acts at two distinct steps of this cascade. As the earliest known NB specifier, AP2a mediates Wnt signals to initiate the NB and activate pax3; as a NC specifier, AP2a regulates further NC development independent of and downstream of NB patterning. Our findings reconcile conflicting observations from various vertebrate organisms. AP2a provides a paradigm for the reiterated use of multifunctional molecules, thereby facilitating emergence of the NC in vertebrates.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/genética , Modelos Biológicos , Cresta Neural/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Ensayo de Cambio de Movilidad Electroforética , Epistasis Genética , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Cresta Neural/embriología , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Front Cell Dev Biol ; 12: 1370905, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39071803

RESUMEN

Neurodevelopmental proteasomopathies constitute a recently defined class of rare Mendelian disorders, arising from genomic alterations in proteasome-related genes. These alterations result in the dysfunction of proteasomes, which are multi-subunit protein complexes essential for maintaining cellular protein homeostasis. The clinical phenotype of these diseases manifests as a syndromic association involving impaired neural development and multisystem abnormalities, notably craniofacial anomalies and malformations of the cardiac outflow tract (OFT). These observations suggest that proteasome loss-of-function variants primarily affect specific embryonic cell types which serve as origins for both craniofacial structures and the conotruncal portion of the heart. In this hypothesis article, we propose that neural crest cells (NCCs), a highly multipotent cell population, which generates craniofacial skeleton, mesenchyme as well as the OFT of the heart, in addition to many other derivatives, would exhibit a distinctive vulnerability to protein homeostasis perturbations. Herein, we introduce the diverse cellular compensatory pathways activated in response to protein homeostasis disruption and explore their potential implications for NCC physiology. Altogether, the paper advocates for investigating proteasome biology within NCCs and their early cranial and cardiac derivatives, offering a rationale for future exploration and laying the initial groundwork for therapeutic considerations.

11.
Dev Biol ; 366(1): 22-33, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22305800

RESUMEN

The neural crest is a transient and multipotent cell population arising at the edge of the neural plate in vertebrates. Recent findings highlight that neural crest patterning is initiated during gastrulation, i.e. earlier than classically described, in a progenitor domain named the neural border. This chapter reviews the dynamic and complex molecular interactions underlying neural border formation and neural crest emergence.


Asunto(s)
Inducción Embrionaria , Cresta Neural , Placa Neural , Animales , Biomarcadores , Tipificación del Cuerpo , Linaje de la Célula , Ectodermo/citología , Ectodermo/embriología , Ectodermo/fisiología , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/fisiología , Placa Neural/citología , Placa Neural/embriología , Placa Neural/fisiología , Vertebrados
12.
Dev Biol ; 366(1): 96-9, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22306197

RESUMEN

Controling embryonic stem cell fate in vitro has been a major challenge in the past decade. Several protocols have been developed to obtain neural crest derivatives in culture, using more or less defined conditions. Here, we present various strategies used to date to obtain neural crest specification and the markers that can be used to identify human neural crest cells.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Embrionarias/citología , Cresta Neural/citología , Animales , Biomarcadores , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Separación Celular/métodos , Células Cultivadas , Células Nutrientes , Humanos , Cresta Neural/embriología
13.
Stem Cell Res ; 69: 103074, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36989619

RESUMEN

Waardenburg syndrome type 1 (WS1), a rare genetic disease characterized by pigmentation defects and mild craniofacial anomalies often associated with congenital deafness is caused by heterozygous mutations in the PAX3 gene (2q36.1). We have generated two induced pluripotent stem cell lines (PCli029-A and PCli031-A) from two patients from the same family both carrying the same heterozygous deletion in PAX3 exon 1 (c.-70_85 + 366del). These cells are pluripotent as they can differentiate into ectoderm, mesoderm and endoderm. They also can activate the early neural crest marker SNAI2. These cells will be useful for studying the human neural crest-derived pigment cells.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome de Waardenburg , Humanos , Síndrome de Waardenburg/genética , Cresta Neural , Factor de Transcripción PAX3/genética , Mutación
14.
Life Sci Alliance ; 5(12)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35914811

RESUMEN

Cell migration is a complex process, tightly regulated during embryonic development and abnormally activated during cancer metastasis. RAS-dependent signaling is a major nexus controlling essential cell parameters including proliferation, survival, and migration, utilizing downstream effectors such as the PI3K/AKT signaling pathway. In melanoma, oncogenic mutations frequently enhance RAS, PI3K/AKT, or MAP kinase signaling and trigger other cancer hallmarks among which the activation of metabolism regulators. PFKFB4 is one of these critical regulators of glycolysis and of the Warburg effect. Here, however, we explore a novel function of PFKFB4 in melanoma cell migration. We find that PFKFB4 interacts with ICMT, a posttranslational modifier of RAS. PFKFB4 promotes ICMT/RAS interaction, controls RAS localization at the plasma membrane, activates AKT signaling and enhances cell migration. We thus provide evidence of a novel and glycolysis-independent function of PFKFB4 in human cancer cells. This unconventional activity links the metabolic regulator PFKFB4 to RAS-AKT signaling and impacts melanoma cell migration.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas c-akt , Línea Celular Tumoral , Movimiento Celular/fisiología , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo , Proteína Metiltransferasas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
15.
Dev Biol ; 340(2): 381-96, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20116373

RESUMEN

Pax3 and Pax7 paralogous genes have functionally diverged in vertebrate evolution, creating opportunity for a new distribution of roles between the two genes and the evolution of novel functions. Here we focus on the regulation and function of Pax7 in the brain and neural crest of amphibian embryos, which display a different pax7 expression pattern, compared to the other vertebrates already described. Pax7 expression is restricted to the midbrain, hindbrain and anterior spinal cord, and Pax7 activity is important for maintaining the fates of these regions, by restricting otx2 expression anteriorly. In contrast, pax3 displays broader expression along the entire neuraxis and Pax3 function is important for posterior brain patterning without acting on otx2 expression. Moreover, while both genes are essential for neural crest patterning, we show that they do so using two distinct mechanisms: Pax3 acts within the ectoderm which will be induced into neural crest, while Pax7 is essential for the inducing activity of the paraxial mesoderm towards the prospective neural crest.


Asunto(s)
Cresta Neural/metabolismo , Factor de Transcripción PAX7/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Blastómeros/metabolismo , Tipificación del Cuerpo/genética , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Embrión no Mamífero/fisiología , Inmunohistoquímica , Hibridación in Situ , Mesodermo/metabolismo , Microinyecciones , Modelos Biológicos , Sistema Nervioso/metabolismo , Oligonucleótidos Antisentido/farmacología , Técnicas de Cultivo de Órganos , Factor de Transcripción PAX3 , Factor de Transcripción PAX7/genética , Factores de Transcripción Paired Box/genética , ARN Mensajero/metabolismo , Proteínas de Xenopus/genética
16.
Fac Rev ; 10: 38, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34046642

RESUMEN

The neural crest is a unique population of multipotent cells forming in vertebrate embryos. Their vast cell fate potential enables the generation of a diverse array of differentiated cell types in vivo. These include, among others, connective tissue, cartilage and bone of the face and skull, neurons and glia of the peripheral nervous system (including enteric nervous system), and melanocytes. Following migration, these derivatives extensively populate multiple germ layers. Within the competent neural border ectoderm, an area located at the junction between the neural and non-neural ectoderm during embryonic development, neural crest cells form in response to a series of inductive secreted cues including BMP, Wnt, and FGF signals. As cells become progressively specified, they express transcriptional modules conducive with their stage of fate determination or cell state. Those sequential states include the neural border state, the premigratory neural crest state, the epithelium-to-mesenchyme transitional state, and the migratory state to end with post-migratory and differentiation states. However, despite the extensive knowledge accumulated over 150 years of neural crest biology, many key questions remain open, in particular the timing of neural crest lineage determination, the control of potency during early developmental stages, and the lineage relationships between different subpopulations of neural crest cells. In this review, we discuss the recent advances in understanding early neural crest formation using cutting-edge high-throughput single cell sequencing approaches. We will discuss how this new transcriptomic data, from 2017 to 2021, has advanced our knowledge of the steps in neural crest cell lineage commitment and specification, the mechanisms driving multipotency, and diversification. We will then discuss the questions that remain to be resolved and how these approaches may continue to unveil the biology of these fascinating cells.

17.
Curr Top Dev Biol ; 145: 313-348, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34074534

RESUMEN

Neural crest cells are a multipotent embryonic stem cell population that emerges from the lateral border of the neural plate after an epithelium-to-mesenchyme transition. These cells then migrate extensively in the embryo and generate a large variety of differentiated cell types and tissues. Alterations in almost any of the processes involved in neural crest development can cause severe congenital defects in humans. Moreover, the malignant transformation of one of the many neural crest derivatives, during childhood or in adults, can cause the development of aggressive tumors prone to metastasis such as melanoma and neuroblastoma. Collectively these diseases are called neurocristopathies. Here we review how a variety of approaches implemented using the amphibian Xenopus as an experimental model have shed light on the molecular basis of numerous neurocristopathies, and how this versatile yet underused vertebrate animal model could help accelerate discoveries in the field. Using the current framework of the neural crest gene regulatory network, we review the pathologies linked to defects at each step of neural crest formation and highlight studies that have used the Xenopus model to decipher the cellular and molecular aspects of neurocristopathies.


Asunto(s)
Anomalías Congénitas/patología , Modelos Animales de Enfermedad , Cresta Neural/patología , Xenopus laevis , Animales , Humanos , Cresta Neural/metabolismo
18.
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
19.
Dev Biol ; 333(1): 26-36, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19555680

RESUMEN

The organization of the embryonic neural plate requires coordination of multiple signal transduction pathways, including fibroblast growth factors (FGFs), bone morphogenetic proteins (BMPs), and WNTs. Many studies have suggested that a critical component of this process is the patterning of posterior neural tissues by an FGF-caudal signaling cascade. Here, we have identified a novel player, Dazap2, and show that it is required in vivo for posterior neural fate. Loss of Dazap2 in embryos resulted in diminished expression of hoxb9 with a concurrent increase in the anterior marker otx2. Furthermore, we found that Dazap2 is required for FGF dependent posterior patterning; surprisingly, this is independent of Cdx activity. Furthermore, in contrast to FGF activity, Dazap2 induction of hoxb9 is not blocked by loss of canonical Wnt signaling. Functionally, we found that increasing Dazap2 levels alters neural patterning and induces posterior neural markers. This activity overcomes the anteriorizing effects of noggin, and is downstream of FGF receptor activation. Our results strongly suggest that Dazap2 is a novel and essential branch of FGF-induced neural patterning.


Asunto(s)
Proteínas Fetales/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Sistema Nervioso/embriología , Proteínas Wnt/metabolismo , Proteínas de Xenopus/fisiología , Xenopus laevis/embriología , Animales , Tipificación del Cuerpo/fisiología , Proteínas de Homeodominio/biosíntesis , Proteínas de Xenopus/biosíntesis , Proteínas de Xenopus/genética , Xenopus laevis/metabolismo
20.
Front Physiol ; 11: 608812, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324244

RESUMEN

The neural crest (NC) cells and cranial placodes are two ectoderm-derived innovations in vertebrates that led to the acquisition of a complex head structure required for a predatory lifestyle. They both originate from the neural border (NB), a portion of the ectoderm located between the neural plate (NP), and the lateral non-neural ectoderm. The NC gives rise to a vast array of tissues and cell types such as peripheral neurons and glial cells, melanocytes, secretory cells, and cranial skeletal and connective cells. Together with cells derived from the cranial placodes, which contribute to sensory organs in the head, the NC also forms the cranial sensory ganglia. Multiple in vivo studies in different model systems have uncovered the signaling pathways and genetic factors that govern the positioning, development, and differentiation of these tissues. In this literature review, we give an overview of NC and placode development, focusing on the early gene regulatory network that controls the formation of the NB during early embryonic stages, and later dictates the choice between the NC and placode progenitor fates.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA