Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 308
Filtrar
1.
J Cell Sci ; 136(16)2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37589341

RESUMO

Bioenergetic metabolism is a key regulator of cellular function and signaling, but how it can instruct the behavior of cells and their fate during embryonic development remains largely unknown. Here, we investigated the role of glucose metabolism in the development of avian trunk neural crest cells (NCCs), a migratory stem cell population of the vertebrate embryo. We uncovered that trunk NCCs display glucose oxidation as a prominent metabolic phenotype, in contrast to what is seen for cranial NCCs, which instead rely on aerobic glycolysis. In addition, only one pathway downstream of glucose uptake is not sufficient for trunk NCC development. Indeed, glycolysis, mitochondrial respiration and the pentose phosphate pathway are all mobilized and integrated for the coordinated execution of diverse cellular programs, epithelial-to-mesenchymal transition, adhesion, locomotion, proliferation and differentiation, through regulation of specific gene expression. In the absence of glucose, the OXPHOS pathway fueled by pyruvate failed to promote trunk NCC adaptation to environmental stiffness, stemness maintenance and fate-decision making. These findings highlight the need for trunk NCCs to make the most of the glucose pathway potential to meet the high metabolic demands appropriate for their development.


Assuntos
Glucose , Crista Neural , Codorniz , Codorniz/crescimento & desenvolvimento , Codorniz/metabolismo , Animais , Crista Neural/crescimento & desenvolvimento , Crista Neural/metabolismo , Glucose/metabolismo , Tubo Neural/citologia , Células Cultivadas , Técnicas In Vitro , Fosforilação Oxidativa , Redes e Vias Metabólicas , Adesão Celular
2.
PLoS Genet ; 18(1): e1010012, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35041640

RESUMO

Ribosomes are essential nanomachines responsible for protein production. Although ribosomes are present in every living cell, ribosome biogenesis dysfunction diseases, called ribosomopathies, impact particular tissues specifically. Here, we evaluate the importance of the box C/D snoRNA-associated ribosomal RNA methyltransferase fibrillarin (Fbl) in the early embryonic development of Xenopus laevis. We report that in developing embryos, the neural plate, neural crest cells (NCCs), and NCC derivatives are rich in fbl transcripts. Fbl knockdown leads to striking morphological defects affecting the eyes and craniofacial skeleton, due to lack of NCC survival caused by massive p53-dependent apoptosis. Fbl is required for efficient pre-rRNA processing and 18S rRNA production, which explains the early developmental defects. Using RiboMethSeq, we systematically reinvestigated ribosomal RNA 2'-O methylation in X. laevis, confirming all 89 previously mapped sites and identifying 15 novel putative positions in 18S and 28S rRNA. Twenty-three positions, including 10 of the new ones, were validated orthogonally by low dNTP primer extension. Bioinformatic screening of the X. laevis transcriptome revealed candidate box C/D snoRNAs for all methylated positions. Mapping of 2'-O methylation at six developmental stages in individual embryos indicated a trend towards reduced methylation at specific positions during development. We conclude that fibrillarin knockdown in early Xenopus embryos causes reduced production of functional ribosomal subunits, thus impairing NCC formation and migration.


Assuntos
Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Precursores de RNA/metabolismo , RNA Ribossômico 18S/metabolismo , RNA Ribossômico 28S/metabolismo , Xenopus laevis/crescimento & desenvolvimento , Animais , Olho/crescimento & desenvolvimento , Olho/metabolismo , Técnicas de Silenciamento de Genes , Metilação , Crista Neural/crescimento & desenvolvimento , Crista Neural/metabolismo , Placa Neural/crescimento & desenvolvimento , Placa Neural/metabolismo , Processamento Pós-Transcricional do RNA , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética
3.
Congenit Anom (Kyoto) ; 62(1): 27-37, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34816492

RESUMO

Palatogenesis is affected by many factors, including gene polymorphisms and exposure to toxic chemicals during sensitive developmental periods. Cleft palate is one of the most common congenital anomalies, and ongoing efforts to elucidate the molecular mechanisms underlying palatogenesis are providing useful insights to reduce the risk of this disorder. To identify novel potential regulators of palatogenesis, we analyzed public transcriptome datasets from a mouse model of cleft palate caused by selective deletion of transforming growth factor-ß (TGFß) receptor type 2 in cranial neural crest cells. We identified the homeobox transcription factor Mohawk (Mkx) as a gene downregulated in the maxilla of TGFß knockout mice compared with wild-type mice. To examine the role of mkx in palatogenesis, we used CRISPR/Cas9 editing to generate zebrafish with impaired expression of mkxa and mkxb, the zebrafish homologs of Mkx. We found that mkx crispants expressed reduced levels of gli1, a critical transcription factor in the Sonic hedgehog (SHH) signaling pathway that plays an important role in the regulation of palatogenesis. Furthermore, we found that mkxa-/- zebrafish were more susceptible than mkxa+/+ zebrafish to the deleterious effects of cyclopamine, an inhibitor of SHH signaling, on upper jaw development. These results suggest that Mkx may be involved in palatogenesis regulated by TGFß and SHH signaling, and that impairment in Mkx function may be related to the etiology of cleft palate.


Assuntos
Fissura Palatina , Proteínas de Homeodomínio , Palato/crescimento & desenvolvimento , Fatores de Transcrição , Animais , Fissura Palatina/induzido quimicamente , Fissura Palatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Proteínas Hedgehog/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Crista Neural/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
4.
Genesis ; 59(10): e23448, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34498354

RESUMO

SETD2 is a histone H3 lysine 36 (H3K36) tri-methylase that is upregulated in response to neural crest induction. Because the H3K36 di-methylase NSD3 and cytoplasmic non-histone protein methylation are necessary for neural crest development, we investigated the expression and requirement for SETD2 in the neural crest. SetD2 is expressed throughout the chick blastoderm beginning at gastrulation. Subsequently, SetD2 mRNA becomes restricted to the neural plate, where it is strongly and dynamically expressed as neural tissue is regionalized and cell fate decisions are made. This includes expression in premigratory neural crest cells, which is downregulated prior to migration. Likely due to the early onset of its expression, SETD2 morpholino knockdown does not significantly alter premigratory Sox10 expression or neural crest migration; however, both are disrupted by a methyltransferase mutant SETD2 construct. These results suggest that SETD2 activity is essential for early neural crest development, further demonstrating that lysine methylation is an important mechanism regulating the neural crest.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Crista Neural/crescimento & desenvolvimento , Neurogênese/genética , Fatores de Transcrição SOXE/genética , Animais , Movimento Celular/genética , Embrião de Galinha , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Histonas/genética , Morfolinos/genética , Crista Neural/metabolismo , Processamento de Proteína Pós-Traducional/genética , RNA Mensageiro/genética
5.
Nat Rev Neurosci ; 22(10): 616-626, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34471282

RESUMO

In their seminal 1983 paper, Gans and Northcutt proposed that evolution of the vertebrate 'new head' was made possible by the advent of the neural crest and cranial placodes. The neural crest is a stem cell population that arises adjacent to the forming CNS and contributes to important cell types, including components of the peripheral nervous system and craniofacial skeleton and elements of the cardiovascular system. In the past few years, the new head hypothesis has been challenged by the discovery in invertebrate chordates of cells with some, but not all, characteristics of vertebrate neural crest cells. Here, we discuss recent findings regarding how neural crest cells may have evolved during the course of deuterostome evolution. The results suggest that there was progressive addition of cell types to the repertoire of neural crest derivatives throughout vertebrate evolution. Novel genomic tools have enabled higher resolution insight into neural crest evolution, from both a cellular and a gene regulatory perspective. Together, these data provide clues regarding the ancestral neural crest state and how the neural crest continues to evolve to contribute to the success of vertebrates as efficient predators.


Assuntos
Evolução Biológica , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Crista Neural/crescimento & desenvolvimento , Crânio/crescimento & desenvolvimento , Animais , Humanos , Crista Neural/citologia , Crânio/citologia , Vertebrados
6.
Genesis ; 59(10): e23447, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34478234

RESUMO

The neural crest is a dynamic embryonic structure that plays a major role in the formation of the vertebrate craniofacial skeleton. Neural crest formation is regulated by a complex sequence of events directed by a network of transcription factors working in concert with chromatin modifiers. The high mobility group nucleosome binding protein 1 (Hmgn1) is a nonhistone chromatin architectural protein, associated with transcriptionally active chromatin. Here we report the expression and function of Hmgn1 during Xenopus neural crest and craniofacial development. Hmgn1 is broadly expressed at the gastrula and neurula stages, and is enriched in the head region at the tailbud stage, especially in the eyes and the pharyngeal arches. Hmgn1 knockdown affected the expression of several neural crest specifiers, including sox8, sox10, foxd3, and twist1, while other genes (sox9 and snai2) were only marginally affected. The specificity of this phenotype was confirmed by rescue, where injection of Hmgn1 mRNA was able to restore sox10 expression in morphant embryos. The reduction in neural crest gene expression at the neurula stage in Hmgn1 morphant embryos correlated with a decreased number of sox10- and twist1-positive cells in the pharyngeal arches at the tailbud stage, and hypoplastic craniofacial cartilages at the tadpole stage. These results point to a novel role for Hmgn1 in the control of gene expression essential for neural crest and craniofacial development. Future work will investigate the precise mode of action of Hmgn1 in this context.


Assuntos
Desenvolvimento Embrionário/genética , Proteína HMGN1/genética , Crista Neural/crescimento & desenvolvimento , Fatores de Transcrição SOXE/genética , Proteína 1 Relacionada a Twist/genética , Proteínas de Xenopus/genética , Animais , Cromatina/genética , Embrião não Mamífero , Fatores de Transcrição Forkhead/genética , Gástrula/crescimento & desenvolvimento , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas de Silenciamento de Genes , Proteína HMGN1/antagonistas & inibidores , Crista Neural/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição/genética , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento
7.
Nat Commun ; 12(1): 4680, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34344887

RESUMO

Craniofacial microsomia (CFM) is the second most common congenital facial anomaly, yet its genetic etiology remains unknown. We perform whole-exome or genome sequencing of 146 kindreds with sporadic (n = 138) or familial (n = 8) CFM, identifying a highly significant burden of loss of function variants in SF3B2 (P = 3.8 × 10-10), a component of the U2 small nuclear ribonucleoprotein complex, in probands. We describe twenty individuals from seven kindreds harboring de novo or transmitted haploinsufficient variants in SF3B2. Probands display mandibular hypoplasia, microtia, facial and preauricular tags, epibulbar dermoids, lateral oral clefts in addition to skeletal and cardiac abnormalities. Targeted morpholino knockdown of SF3B2 in Xenopus results in disruption of cranial neural crest precursor formation and subsequent craniofacial cartilage defects, supporting a link between spliceosome mutations and impaired neural crest development in congenital craniofacial disease. The results establish haploinsufficient variants in SF3B2 as the most prevalent genetic cause of CFM, explaining ~3% of sporadic and ~25% of familial cases.


Assuntos
Síndrome de Goldenhar/genética , Haploinsuficiência , Fatores de Processamento de RNA/genética , Adolescente , Adulto , Animais , Criança , Exoma/genética , Feminino , Estudos de Associação Genética , Síndrome de Goldenhar/patologia , Humanos , Lactente , Masculino , Mutação , Crista Neural/crescimento & desenvolvimento , Crista Neural/patologia , Linhagem , Spliceossomos/genética , Xenopus laevis
8.
Development ; 148(16)2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34338282

RESUMO

Mutations in ITPR1 cause ataxia and aniridia in individuals with Gillespie syndrome (GLSP). However, the pathogenic mechanisms underlying aniridia remain unclear. We identified a de novo GLSP mutation hotspot in the 3'-region of ITPR1 in five individuals with GLSP. Furthermore, RNA-sequencing and immunoblotting revealed an eye-specific transcript of Itpr1, encoding a 218amino acid isoform. This isoform is localized not only in the endoplasmic reticulum, but also in the nuclear and cytoplasmic membranes. Ocular-specific transcription was repressed by SOX9 and induced by MAF in the anterior eye segment (AES) tissues. Mice lacking seven base pairs of the last Itpr1 exon exhibited ataxia and aniridia, in which the iris lymphatic vessels, sphincter and dilator muscles, corneal endothelium and stroma were disrupted, but the neural crest cells persisted after completion of AES formation. Our analyses revealed that the 218-amino acid isoform regulated the directionality of actin fibers and the intensity of focal adhesion. The isoform might control the nuclear entry of transcriptional regulators, such as YAP. It is also possible that ITPR1 regulates both AES differentiation and muscle contraction in the iris.


Assuntos
Aniridia/sangue , Aniridia/genética , Segmento Anterior do Olho/crescimento & desenvolvimento , Ataxia Cerebelar/sangue , Ataxia Cerebelar/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Deficiência Intelectual/sangue , Deficiência Intelectual/genética , Mutação , Crista Neural/crescimento & desenvolvimento , Adolescente , Animais , Segmento Anterior do Olho/metabolismo , Criança , Pré-Escolar , Modelos Animais de Doenças , Éxons , Feminino , Técnicas de Introdução de Genes , Células HEK293 , Humanos , Lactente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Crista Neural/metabolismo , Isoformas de Proteínas/metabolismo , Transfecção , Adulto Jovem
9.
Cell Mol Life Sci ; 78(9): 4435-4450, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33796894

RESUMO

During early neural development, changes in signalling inform the expression of transcription factors that in turn instruct changes in cell identity. At the same time, switches in adhesion molecule expression result in cellular rearrangements that define the morphology of the emerging neural tube. It is becoming increasingly clear that these two processes influence each other; adhesion molecules do not simply operate downstream of or in parallel with changes in cell identity but rather actively feed into cell fate decisions. Why are differentiation and adhesion so tightly linked? It is now over 60 years since Conrad Waddington noted the remarkable "Constancy of the Wild Type" (Waddington in Nature 183: 1654-1655, 1959) yet we still do not fully understand the mechanisms that make development so reproducible. Conversely, we do not understand why directed differentiation of cells in a dish is sometimes unpredictable and difficult to control. It has long been suggested that cells make decisions as 'local cooperatives' rather than as individuals (Gurdon in Nature 336: 772-774, 1988; Lander in Cell 144: 955-969, 2011). Given that the cadherin family of adhesion molecules can simultaneously influence morphogenesis and signalling, it is tempting to speculate that they may help coordinate cell fate decisions between neighbouring cells in the embryo to ensure fidelity of patterning, and that the uncoupling of these processes in a culture dish might underlie some of the problems with controlling cell fate decisions ex-vivo. Here we review the expression and function of cadherins during early neural development and discuss how and why they might modulate signalling and differentiation as neural tissues are formed.


Assuntos
Caderinas/metabolismo , Crista Neural/metabolismo , Animais , Caderinas/química , Caderinas/classificação , Adesão Celular , Diferenciação Celular , Evolução Molecular , Humanos , Crista Neural/citologia , Crista Neural/crescimento & desenvolvimento , Neurogênese , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Int J Mol Sci ; 22(8)2021 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-33920095

RESUMO

Research on the development of the dorsal neural tube is particularly challenging. In this highly dynamic domain, a temporal transition occurs between early neural crest progenitors that undergo an epithelial-to-mesenchymal transition and exit the neural primordium, and the subsequent roof plate, a resident epithelial group of cells that constitutes the dorsal midline of the central nervous system. Among other functions, the roof plate behaves as an organizing center for the generation of dorsal interneurons. Despite extensive knowledge of the formation, emigration and migration of neural crest progenitors, little is known about the mechanisms leading to the end of neural crest production and the transition into a roof plate stage. Are these two mutually dependent or autonomously regulated processes? Is the generation of roof plate and dorsal interneurons induced by neural tube-derived factors throughout both crest and roof plate stages, respectively, or are there differences in signaling properties and responsiveness as a function of time? In this review, we discuss distinctive characteristics of each population and possible mechanisms leading to the shift between the above cell types.


Assuntos
Diferenciação Celular/genética , Sistema Nervoso Central/crescimento & desenvolvimento , Crista Neural/crescimento & desenvolvimento , Tubo Neural/crescimento & desenvolvimento , Animais , Proteínas Morfogenéticas Ósseas/genética , Sistema Nervoso Central/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Interneurônios/metabolismo , Transdução de Sinais/genética , Proteínas Wnt/genética
12.
Elife ; 102021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33591267

RESUMO

Neural crest cells (NCCs) are vertebrate stem cells that give rise to various cell types throughout the developing body in early life. Here, we utilized single-cell transcriptomic analyses to delineate NCC-derivatives along the posterior developing vertebrate, zebrafish, during the late embryonic to early larval stage, a period when NCCs are actively differentiating into distinct cellular lineages. We identified several major NCC/NCC-derived cell-types including mesenchyme, neural crest, neural, neuronal, glial, and pigment, from which we resolved over three dozen cellular subtypes. We dissected gene expression signatures of pigment progenitors delineating into chromatophore lineages, mesenchyme cells, and enteric NCCs transforming into enteric neurons. Global analysis of NCC derivatives revealed they were demarcated by combinatorial hox gene codes, with distinct profiles within neuronal cells. From these analyses, we present a comprehensive cell-type atlas that can be utilized as a valuable resource for further mechanistic and evolutionary investigations of NCC differentiation.


Assuntos
Linhagem da Célula , Crista Neural/crescimento & desenvolvimento , Peixe-Zebra/crescimento & desenvolvimento , Animais , Diferenciação Celular , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Larva/crescimento & desenvolvimento
13.
PLoS Genet ; 17(1): e1009296, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33465092

RESUMO

The process of cell fate commitment involves sequential changes in the gene expression profiles of embryonic progenitors. This is exemplified in the development of the neural crest, a migratory stem cell population derived from the ectoderm of vertebrate embryos. During neural crest formation, cells transition through distinct transcriptional states in a stepwise manner. The mechanisms underpinning these shifts in cell identity are still poorly understood. Here we employ enhancer analysis to identify a genetic sub-circuit that controls developmental transitions in the nascent neural crest. This sub-circuit links Wnt target genes in an incoherent feedforward loop that controls the sequential activation of genes in the neural crest lineage. By examining the cis-regulatory apparatus of Wnt effector gene AXUD1, we found that multipotency factor SP5 directly promotes neural plate border identity, while inhibiting premature expression of specification genes. Our results highlight the importance of repressive interactions in the neural crest gene regulatory network and illustrate how genes activated by the same upstream signal become temporally segregated during progressive fate restriction.


Assuntos
Elementos Facilitadores Genéticos/genética , Crista Neural/crescimento & desenvolvimento , Placa Neural/crescimento & desenvolvimento , Fatores de Transcrição/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Diferenciação Celular/genética , Movimento Celular/genética , Embrião de Galinha , Proteínas de Ligação a DNA/genética , Ectoderma/crescimento & desenvolvimento , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Humanos , Hibridização In Situ , Crista Neural/metabolismo , Placa Neural/metabolismo , Via de Sinalização Wnt/genética
14.
PLoS One ; 16(1): e0244794, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33439865

RESUMO

Understanding how fate specification of distinct cell-types from multipotent progenitors occurs is a fundamental question in embryology. Neural crest stem cells (NCSCs) generate extraordinarily diverse derivatives, including multiple neural, skeletogenic and pigment cell fates. Key transcription factors and extracellular signals specifying NCSC lineages remain to be identified, and we have only a little idea of how and when they function together to control fate. Zebrafish have three neural crest-derived pigment cell types, black melanocytes, light-reflecting iridophores and yellow xanthophores, which offer a powerful model for studying the molecular and cellular mechanisms of fate segregation. Mitfa has been identified as the master regulator of melanocyte fate. Here, we show that an Mitf-related transcription factor, Tfec, functions as master regulator of the iridophore fate. Surprisingly, our phenotypic analysis of tfec mutants demonstrates that Tfec also functions in the initial specification of all three pigment cell-types, although the melanocyte and xanthophore lineages recover later. We show that Mitfa represses tfec expression, revealing a likely mechanism contributing to the decision between melanocyte and iridophore fate. Our data are consistent with the long-standing proposal of a tripotent progenitor restricted to pigment cell fates. Moreover, we investigate activation, maintenance and function of tfec in multipotent NCSCs, demonstrating for the first time its role in the gene regulatory network forming and maintaining early neural crest cells. In summary, we build on our previous work to characterise the gene regulatory network governing iridophore development, establishing Tfec as the master regulator driving iridophore specification from multipotent progenitors, while shedding light on possible cellular mechanisms of progressive fate restriction.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Crista Neural/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular , Linhagem da Célula , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Melanócitos/citologia , Melanócitos/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Mutagênese , Crista Neural/citologia , Pigmentação/genética , RNA Guia de Cinetoplastídeos/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
15.
Dev Growth Differ ; 63(1): 93-99, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33326593

RESUMO

Neural crest (NC) cells give rise to a wide variety of cell types and tissues, such as neurons and glial cells in the peripheral nervous system. Sox2, which encodes an HMG-box transcription factor, is known to mediate pluripotency of primordial germ cells and embryonic stem (ES)/induced pluripotent stem (iPS) cells, and to regulate central nervous system development. Previous studies have revealed that Sox2 is also an important regulator of NC development. This review summarizes the well-established inhibitory roles of Sox2 in NC formation and subsequent neuronal differentiation of NC-derived cells. This review also covers recent studies suggesting additional roles for Sox2 in early NC development, neurogenesis, and glial differentiation of NC-derived cells.


Assuntos
Crista Neural/crescimento & desenvolvimento , Fatores de Transcrição SOXB1/metabolismo , Humanos , Crista Neural/citologia , Crista Neural/metabolismo
16.
Methods Mol Biol ; 2230: 3-16, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33197005

RESUMO

Development of cartilage and bone, the core components of the mouse skeletal system, depends on coordinated proliferation and differentiation of skeletogenic cells, including chondrocytes and osteoblasts. These cells differentiate from common progenitor cells originating in the mesoderm and neural crest. Multiple signaling pathways and transcription factors tightly regulate differentiation and proliferation of skeletal cells. In this chapter, we overview the process of mouse skeletal development and discuss major regulators of skeletal cells at each developmental stage.


Assuntos
Desenvolvimento Ósseo/genética , Mesoderma/crescimento & desenvolvimento , Osteogênese/genética , Células-Tronco/citologia , Animais , Cartilagem/crescimento & desenvolvimento , Diferenciação Celular/genética , Condrócitos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Crista Neural/crescimento & desenvolvimento , Osteoblastos/metabolismo , Transdução de Sinais/genética
17.
Development ; 148(22)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-35020872

RESUMO

Neural crest cells are crucial in development, not least because of their remarkable multipotency. Early findings stimulated two hypotheses for how fate specification and commitment from fully multipotent neural crest cells might occur, progressive fate restriction (PFR) and direct fate restriction, differing in whether partially restricted intermediates were involved. Initially hotly debated, they remain unreconciled, although PFR has become favoured. However, testing of a PFR hypothesis of zebrafish pigment cell development refutes this view. We propose a novel 'cyclical fate restriction' hypothesis, based upon a more dynamic view of transcriptional states, reconciling the experimental evidence underpinning the traditional hypotheses.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Crista Neural/crescimento & desenvolvimento , Peixe-Zebra/crescimento & desenvolvimento , Animais , Linhagem da Célula/fisiologia , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Melanócitos/metabolismo , Pigmentação/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
Development ; 148(22)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-35020873

RESUMO

The dynamics of multipotent neural crest cell differentiation and invasion as cells travel throughout the vertebrate embryo remain unclear. Here, we preserve spatial information to derive the transcriptional states of migrating neural crest cells and the cellular landscape of the first four chick cranial to cardiac branchial arches (BA1-4) using label-free, unsorted single-cell RNA sequencing. The faithful capture of branchial arch-specific genes led to identification of novel markers of migrating neural crest cells and 266 invasion genes common to all BA1-4 streams. Perturbation analysis of a small subset of invasion genes and time-lapse imaging identified their functional role to regulate neural crest cell behaviors. Comparison of the neural crest invasion signature to other cell invasion phenomena revealed a shared set of 45 genes, a subset of which showed direct relevance to human neuroblastoma cell lines analyzed after exposure to the in vivo chick embryonic neural crest microenvironment. Our data define an important spatio-temporal reference resource to address patterning of the vertebrate head and neck, and previously unidentified cell invasion genes with the potential for broad impact.


Assuntos
Região Branquial/crescimento & desenvolvimento , Cabeça/crescimento & desenvolvimento , Pescoço/crescimento & desenvolvimento , Crista Neural/crescimento & desenvolvimento , Animais , Padronização Corporal/genética , Região Branquial/embriologia , Diferenciação Celular/genética , Movimento Celular/genética , Microambiente Celular/genética , Embrião de Galinha , Embrião de Mamíferos , Embrião não Mamífero , Desenvolvimento Embrionário/genética , Cabeça/embriologia , Humanos , Mesoderma/crescimento & desenvolvimento , Células-Tronco Multipotentes/citologia , Pescoço/embriologia , Crista Neural/metabolismo , Neuroblastoma/genética , Neuroblastoma/patologia , Organogênese/genética , Microambiente Tumoral/genética , Vertebrados/genética , Vertebrados/crescimento & desenvolvimento
19.
Biomolecules ; 11(1)2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33375093

RESUMO

The neural crest hypothesis states that the phenotypic features of the domestication syndrome are due to a reduced number or disruption of neural crest cells (NCCs) migration, as these cells differentiate at their final destinations and proliferate into different tissues whose activity is reduced by domestication. Comparing the phenotypic characteristics of modern and prehistoric man, it is clear that during their recent evolutionary past, humans also went through a process of self-domestication with a simultaneous prolongation of the period of socialization. This has led to the development of social abilities and skills, especially language, as well as neoteny. Disorders of neural crest cell development and migration lead to many different conditions such as Waardenburg syndrome, Hirschsprung disease, fetal alcohol syndrome, DiGeorge and Treacher-Collins syndrome, for which the mechanisms are already relatively well-known. However, for others, such as Williams-Beuren syndrome and schizophrenia that have the characteristics of hyperdomestication, and autism spectrum disorders, and 7dupASD syndrome that have the characteristics of hypodomestication, much less is known. Thus, deciphering the biological determinants of disordered self-domestication has great potential for elucidating the normal and disturbed ontogenesis of humans, as well as for the understanding of evolution of mammals in general.


Assuntos
Evolução Biológica , Domesticação , Idioma , Crista Neural/fisiopatologia , Movimento Celular/genética , Proliferação de Células/genética , Transtornos do Espectro Alcoólico Fetal/genética , Doença de Hirschsprung/genética , Humanos , Disostose Mandibulofacial/genética , Crista Neural/crescimento & desenvolvimento , Crista Neural/metabolismo , Fenótipo , Esquizofrenia/genética , Habilidades Sociais , Síndrome de Waardenburg/genética , Síndrome de Williams/genética
20.
Elife ; 92020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33198887

RESUMO

Paired fins are a defining feature of the jawed vertebrate body plan, but their evolutionary origin remains unresolved. Gegenbaur proposed that paired fins evolved as gill arch serial homologues, but this hypothesis is now widely discounted, owing largely to the presumed distinct embryonic origins of these structures from mesoderm and neural crest, respectively. Here, we use cell lineage tracing to test the embryonic origin of the pharyngeal and paired fin skeleton in the skate (Leucoraja erinacea). We find that while the jaw and hyoid arch skeleton derive from neural crest, and the pectoral fin skeleton from mesoderm, the gill arches are of dual origin, receiving contributions from both germ layers. We propose that gill arches and paired fins are serially homologous as derivatives of a continuous, dual-origin mesenchyme with common skeletogenic competence, and that this serial homology accounts for their parallel anatomical organization and shared responses to axial patterning signals.


A common way to evolve new body parts is to copy existing ones and to remodel them. In insects for example, the antennae, mouth parts and legs all follow the same basic body plan, with modifications that adapt them for different uses. In the late 19th century, anatomist Karl Gegenbaur noticed a similar pattern in fish. He saw similarities between pairs of fins and pairs of gills, suggesting that one evolved from the other. But there is currently no fossil evidence documenting such a transformation. Modern research has shown that the development of both gill and fin skeletons shares common genetic pathways. But the cells that form the two structures do not come from the same place. Gill skeletons develop from a part of the embryo called the neural crest, while fin skeletons come from a region called the mesoderm. One way to test Gegenbaur's idea is to look more closely at the cells that form gill and fin skeletons as fish embryos develop. Here, Sleight and Gillis examined the gills and fins of a cartilaginous fish called Leucoraja erinacea, also known as the little skate. Sleight and Gillis labelled the cells from the neural crest and mesoderm of little skate embryos with a fluorescent dye and then tracked the cells over several weeks. While the fins did form from mesoderm cells, the gills did not develop as expected. The first gill contained only neural crest cells, but the rest were a mixture of both cell types. This suggests that fins and gills develop from a common pool of cells that consists of both neural crest and mesoderm cells, which have the potential to develop into either body part. This previously unrecognised embryonic continuity between gills and fins explains why these structures respond in the same way to the same genetic cues, regardless of what cell type they develop from. Based on this new evidence, Sleight and Gillis believe that Gegenbaur was right, and that fins and gills do indeed share an evolutionary history. While firm evidence for the transformation of gills into fins remains elusive, this work suggests it is possible. A deeper understanding of the process could shed light on the development of other repeated structures in nature. Research shows that animals use a relatively small number of genetic cues to set out their body plans. This can make it hard to use genetics alone to study their evolutionary history. But, looking at how different cell types respond to those cues to build anatomical features, like fins and gills, could help to fill in the gaps.


Assuntos
Nadadeiras de Animais/embriologia , Brânquias/embriologia , Crista Neural/crescimento & desenvolvimento , Rajidae/embriologia , Animais , Embrião não Mamífero , Desenvolvimento Embrionário , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esqueleto/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA