Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genesis ; 62(1): e23553, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37735882

RESUMO

The neural crest is a stem cell population that originates from the ectoderm during the initial steps of nervous system development. Neural crest cells delaminate from the neuroepithelium by undergoing a spatiotemporally regulated epithelial-mesenchymal transition (EMT) that proceeds in a coordinated wave head-to-tail to exit from the neural tube. While much is known about the transcriptional programs and membrane changes that promote EMT, there are additional levels of gene expression control that neural crest cells exert at the level of RNA to control EMT and migration. Yet, the role of post-transcriptional regulation, and how it drives and contributes to neural crest EMT, is not well understood. In this mini-review, we explore recent advances in our understanding of the role of post-transcriptional regulation during neural crest EMT.


Assuntos
Transição Epitelial-Mesenquimal , Crista Neural , Crista Neural/metabolismo , Transição Epitelial-Mesenquimal/genética , Ectoderma , Tubo Neural , Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento
2.
Proc Natl Acad Sci U S A ; 119(51): e2212879119, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36508654

RESUMO

Epithelial-to-mesenchymal transition (EMT) is a dramatic change in cellular physiology during development and metastasis, which requires coordination between cell signaling, adhesion, and membrane protrusions. These processes all involve dynamic changes in the plasma membrane; yet, how membrane lipid content regulates membrane function during EMT remains incompletely understood. By screening for differential expression of lipid-modifying genes over the course of EMT in the avian neural crest, we have identified the ceramide-producing enzyme neutral sphingomyelinase 2 (nSMase2) as a critical regulator of a developmental EMT. nSMase2 expression begins at the onset of EMT, and in vivo knockdown experiments demonstrate that nSMase2 is necessary for neural crest migration. We find that nSMase2 promotes Wnt and BMP signaling and is required to activate the mesenchymal gene expression program. Mechanistically, we show that nSMase2-dependent ceramide production is necessary for and sufficient to up-regulate endocytosis and is required for Wnt co-receptor internalization. Finally, inhibition of endocytosis in the neural crest mimics the loss of migration and Wnt signaling observed following nSMase2 knockdown. Our results support a model in which nSMase2 is expressed at the onset of neural crest EMT to produce ceramide and facilitate receptor-mediated endocytosis of Wnt and BMP signaling complexes, thereby activating promigratory gene expression. These results highlight the critical role of plasma membrane lipid metabolism in regulating transcriptional changes during developmental EMT programs.


Assuntos
Transição Epitelial-Mesenquimal , Crista Neural , Transição Epitelial-Mesenquimal/genética , Via de Sinalização Wnt , Membrana Celular/metabolismo , Lipídeos de Membrana/metabolismo , Ceramidas/metabolismo , Movimento Celular/genética
3.
Elife ; 112022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-36189921

RESUMO

While neural crest development is known to be transcriptionally controlled via sequential activation of gene regulatory networks (GRNs), recent evidence increasingly implicates a role for post-transcriptional regulation in modulating the output of these regulatory circuits. Using available single-cell RNA-sequencing datasets from avian embryos to identify potential post-transcriptional regulators, we found that Elavl1, which encodes for an RNA-binding protein with roles in transcript stability, was enriched in the premigratory cranial neural crest. Perturbation of Elavl1 resulted in premature neural crest delamination from the neural tube as well as significant reduction in transcripts associated with the neural crest specification GRN, phenotypes that are also observed with downregulation of the canonical Wnt inhibitor Draxin. That Draxin is the primary target for stabilization by Elavl1 during cranial neural crest specification was shown by RNA-sequencing, RNA immunoprecipitation, RNA decay measurement, and proximity ligation assays, further supporting the idea that the downregulation of neural crest specifier expression upon Elavl1 knockdown was largely due to loss of Draxin. Importantly, exogenous Draxin rescued cranial neural crest specification defects observed with Elavl1 knockdown. Thus, Elavl1 plays a critical a role in the maintenance of cranial neural crest specification via Draxin mRNA stabilization. Together, these data highlight an important intersection of post-transcriptional regulation with modulation of the neural crest specification GRN.


As an embryo develops, different genetic programs become activated to give cell populations a specific biological identity that will shape their fate. For instance, when certain sets of genes get switched on, cells from the outermost layer of the embryo start to migrate to their final destination within the body. There, these 'neural crest cells' will contribute to bones and cartilage in the face, pigmented skin spots, and muscles or nerves in the gut. When genes responsible for the neural crest identity are active, their instructions are copied into an 'RNA molecule' which will then relay this information to protein-building structures. How well the RNA can pass on the message depends on how long it persists within the cell. Certain RNA-binding proteins can control this process, but it is unclear whether and how this regulation takes place in neural crest cells. In their work, Hutchins et al. therefore focused on identifying RNA-binding proteins involved in neural crest identity. Exploratory searches of genetic data from chick embryos revealed that, even before they started to migrate, neural crest cells which have recently acquired their identity produced large amounts of the RNA-binding protein Elavl1. In addition, these cells did not behave normally when embryos were deprived of the protein: they left the outer layer too soon and then switched off genes important for their identity. Genetic studies of neural crest cells lacking Elavl1 revealed that this effect was due to having lost the RNA molecule produced from the Draxin gene. Introducing an additional source of Draxin into mutant embryos missing Elavl1 was enough to restore normal neural crest behaviour. Further biochemical experiments then showed that the RNA for Draxin decayed quickly in the absence of Elavl1. This suggests that the protein normally allows Draxin's RNA to persist long enough to pass on its message. These results reveal a new mechanism controlling the identity and behaviour of the neural crest. Since many cancers in adulthood arise from the descendants of neural crest cells, Hutchins et al. hope that this knowledge could lead to improved therapies in the future.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Crista Neural , Crista Neural/fisiologia , RNA/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
4.
Dev Biol ; 477: 251-261, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34102166

RESUMO

BMP signaling plays iterative roles during vertebrate neural crest development from induction through craniofacial morphogenesis. However, far less is known about the role of BMP activity in cranial neural crest epithelial-to-mesenchymal transition and delamination. By measuring canonical BMP signaling activity as a function of time from specification through early migration of avian midbrain neural crest cells, we found elevated BMP signaling during delamination stages. Moreover, inhibition of canonical BMP activity via a dominant negative mutant Type I BMP receptor showed that BMP signaling is required for neural crest migration from the midbrain, independent from an effect on EMT and delamination. Transcriptome profiling on control compared to BMP-inhibited cranial neural crest cells identified novel BMP targets during neural crest delamination and early migration including targets of the Notch pathway that are upregulated following BMP inhibition. These results suggest potential crosstalk between the BMP and Notch pathways in early migrating cranial neural crest and provide novel insight into mechanisms regulated by BMP signaling during early craniofacial development.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Mesencéfalo/embriologia , Crista Neural/metabolismo , Transdução de Sinais , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Embrião de Galinha , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Mesencéfalo/metabolismo , Crista Neural/embriologia , Crânio/embriologia , Crânio/metabolismo , Técnicas de Cultura de Tecidos
5.
Front Physiol ; 12: 624037, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33613313

RESUMO

Canonical Wnt signaling plays an essential role in proper craniofacial morphogenesis, at least partially due to regulation of various aspects of cranial neural crest development. In an effort to gain insight into the etiology of craniofacial abnormalities resulting from Wnt signaling and/or cranial neural crest dysfunction, we sought to identify Wnt-responsive targets during chick cranial neural crest development. To this end, we leveraged overexpression of a canonical Wnt antagonist, Draxin, in conjunction with RNA-sequencing of cranial neural crest cells that have just activated their epithelial-mesenchymal transition (EMT) program. Through differential expression analysis, gene list functional annotation, hybridization chain reaction (HCR), and quantitative reverse transcription polymerase chain reaction (RT-qPCR), we validated a novel downstream target of canonical Wnt signaling in cranial neural crest - RHOB - and identified possible signaling pathway crosstalk underlying cranial neural crest migration. The results reveal novel putative targets of canonical Wnt signaling during cranial neural crest EMT and highlight important intersections across signaling pathways involved in craniofacial development.

6.
Methods Mol Biol ; 2179: 3-6, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32939707

RESUMO

The epithelial-to-mesenchymal transition (EMT) encompasses a complex cascade of events through which a cell transits to reduce its epithelial characteristics and become migratory. Classically, this transition has been considered complete upon loss of molecular markers characteristic of an "epithelial" state and acquisition of those associated with "mesenchymal" cells. Recently, however, evidence from both developmental and cancer EMT contexts suggest that cells undergoing EMT are often heterogeneous, concomitantly expressing both epithelial and mesenchymal markers to varying degrees; rather, cells frequently display a "partial" EMT phenotype and do not necessarily require full "mesenchymalization" to become migratory. Here, we offer a brief perspective on recent important advances in our fundamental understanding of the spectrum of cellular states that occur during partial EMT in the context of development and cancer metastasis.


Assuntos
Técnicas de Cultura de Células/métodos , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Células-Tronco Neoplásicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia
7.
Elife ; 92020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32965216

RESUMO

During gastrulation, neural crest cells are specified at the neural plate border, as characterized by Pax7 expression. Using single-cell RNA sequencing coupled with high-resolution in situ hybridization to identify novel transcriptional regulators, we show that chromatin remodeler Hmga1 is highly expressed prior to specification and maintained in migrating chick neural crest cells. Temporally controlled CRISPR-Cas9-mediated knockouts uncovered two distinct functions of Hmga1 in neural crest development. At the neural plate border, Hmga1 regulates Pax7-dependent neural crest lineage specification. At premigratory stages, a second role manifests where Hmga1 loss reduces cranial crest emigration from the dorsal neural tube independent of Pax7. Interestingly, this is rescued by stabilized ß-catenin, thus implicating Hmga1 as a canonical Wnt activator. Together, our results show that Hmga1 functions in a bimodal manner during neural crest development to regulate specification at the neural plate border, and subsequent emigration from the neural tube via canonical Wnt signaling.


The neural plate is a structure that serves as the basis for the brain and central nervous system during the development of animals with a backbone. In particular, the tissues at the border of the neural plate become the neural crest, a group of highly mobile cells that can specialize to form nerves and parts of the face. The exact molecular mechanisms that allow the crest to emerge are still unknown. The protein Hmga1 alters how genes are packaged and organized inside cells, which in turn influences how genes are switched on and off. Here, Gandhi et al. studied how Hmga1 helps to shape the neural crest in developing chicken embryos. To do so, they harnessed a genetic tool called CRISPR-Cas9, and deleted the gene that encodes Hmga1 at specific developmental stages. This manipulation highlighted two periods where Hmga1 is active. First, Hmga1 helped to define neural crest cells at the neural plate border by activating a gene called pax7. Then, at a later stage, Hmga1 allowed these cells to move to other parts of the body by triggering the Wnt communication system. Failure for the neural crest to develop properly causes birth defects and cancers such as melanoma and childhood neuroblastoma, highlighting the need to better understand how this structure is formed. In addition, a better grasp of the roles of Hmga1 in healthy development could help to appreciate how it participates in a range of adult cancers.


Assuntos
Proteínas Aviárias/genética , Movimento Celular , Embrião de Galinha/embriologia , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas HMGA/genética , Crista Neural/embriologia , Animais , Proteínas Aviárias/metabolismo , Galinhas/fisiologia , Proteínas HMGA/metabolismo , Via de Sinalização Wnt
8.
Dev Biol ; 446(2): 151-158, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30579765

RESUMO

Premigratory neural crest cells arise within the dorsal neural tube and subsequently undergo an epithelial-to-mesenchymal transition (EMT) to leave the neuroepithelium and initiate migration. Draxin is a Wnt modulator that has been shown to control the timing of cranial neural crest EMT. Here we show that this process is accompanied by three stages of remodeling of the basement membrane protein laminin, from regression to expansion and channel formation. Loss of Draxin results in blocking laminin remodeling at the regression stage, whereas ectopic maintenance of Draxin blocks remodeling at the expansion stage. The latter effect is rescued by addition of Snail2, previously shown to be downstream of Draxin. Our results demonstrate an essential function for the Wnt modulator Draxin in regulating basement membrane remodeling during cranial neural crest EMT.


Assuntos
Proteínas Aviárias/genética , Membrana Basal/metabolismo , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica no Desenvolvimento , Laminina/genética , Crista Neural/metabolismo , Animais , Proteínas Aviárias/metabolismo , Membrana Basal/embriologia , Movimento Celular/genética , Embrião de Galinha , Técnicas de Silenciamento de Genes , Laminina/metabolismo , Crista Neural/embriologia , Crânio/embriologia , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo
9.
J Cell Biol ; 217(10): 3683-3697, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30026247

RESUMO

Neural crest cells undergo a spatiotemporally regulated epithelial-to-mesenchymal transition (EMT) that proceeds head to tailward to exit from the neural tube. In this study, we show that the secreted molecule Draxin is expressed in a transient rostrocaudal wave that mirrors this emigration pattern, initiating after neural crest specification and being down-regulated just before delamination. Functional experiments reveal that Draxin regulates the timing of cranial neural crest EMT by transiently inhibiting canonical Wnt signaling. Ectopic maintenance of Draxin in the cranial neural tube blocks full EMT; while cells delaminate, they fail to become mesenchymal and migratory. Loss of Draxin results in premature delamination but also in failure to mesenchymalize. These results suggest that a pulse of intermediate Wnt signaling triggers EMT and is necessary for its completion. Taken together, these data show that transient secreted Draxin mediates proper levels of canonical Wnt signaling required to regulate the precise timing of initiation and completion of cranial neural crest EMT.


Assuntos
Proteínas Aviárias/metabolismo , Transição Epitelial-Mesenquimal , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/embriologia , Tubo Neural/embriologia , Via de Sinalização Wnt/fisiologia , Animais , Proteínas Aviárias/genética , Embrião de Galinha , Galinhas , Proteínas do Tecido Nervoso/genética , Crânio/embriologia
10.
Dev Biol ; 444 Suppl 1: S98-S109, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29981692

RESUMO

Arising within the neural tube between the cranial and trunk regions of the body axis, the vagal neural crest shares interesting similarities in its migratory routes and derivatives with other neural crest populations. However, the vagal neural crest is also unique in its ability to contribute to diverse organs including the heart and enteric nervous system. This review highlights the migratory routes of the vagal neural crest and compares them across multiple vertebrates. We also summarize recent advances in understanding vagal neural crest ontogeny and discuss the contribution of this important neural crest population to the cardiovascular system and endoderm-derived organs, including the thymus, lungs and pancreas.


Assuntos
Sistema Nervoso Entérico/embriologia , Sistema Nervoso Entérico/fisiologia , Crista Neural/metabolismo , Animais , Evolução Biológica , Padronização Corporal/fisiologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Gânglios , Coração/embriologia , Humanos , Pulmão/embriologia , Crista Neural/embriologia , Crista Neural/fisiologia , Tubo Neural , Neurogênese , Pâncreas/embriologia , Timo/embriologia , Tronco , Vertebrados/embriologia
11.
Biochem Biophys Res Commun ; 478(2): 772-6, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27501755

RESUMO

hnRNP K is a highly conserved nucleocytoplasmic shuttling protein, which associates with RNAs through synergistic binding via its three KH domains. hnRNP K is required for proper nuclear export and translational control of its mRNA targets, and these processes are controlled by hnRNP K's movement between subcellular compartments. Whereas the nuclear export and localization of hnRNP K that is associated with mRNP complexes has been well studied, the trafficking of hnRNP K that is unbound to mRNA has yet to be elucidated. To that end, we expressed an EGFP-tagged RNA binding-defective form of hnRNP K in intact Xenopus embryos, and found it was rapidly degraded in vivo. Deleting hnRNP K's nuclear localization signal (NLS), which contains two prospective ubiquitination sites, rescued the protein from degradation. These data demonstrate a novel activity for the NLS of hnRNP K in regulating the protein's stability in vivo when it is unbound to nucleic acids.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/genética , Sinais de Localização Nuclear/genética , Proteínas Recombinantes de Fusão/genética , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Sequência Conservada , Embrião não Mamífero , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Mutagênese Sítio-Dirigida , Sinais de Localização Nuclear/metabolismo , Domínios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estabilidade Proteica , Proteólise , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Ubiquitinação , Xenopus laevis/embriologia , Xenopus laevis/metabolismo
12.
Neurosci Lett ; 607: 59-65, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26409787

RESUMO

Post-transcriptional control of cytoskeletal genes fine-tunes the supply of structural materials to growing axons in response to extracellular cues. In Xenopus, heterogeneous nuclear ribonucleoprotein K (hnRNPK) plays a crucial role in the nuclear export and translation of multiple cytoskeletal-related mRNAs required for axon outgrowth, and as a substrate of multiple kinases, is thus a likely molecular target of cell signaling pathways regulating such outgrowth. To study the role of hnRNPK's phosphorylation by extracellular signal-regulated kinase (ERK) in Xenopus axon outgrowth, we identified the only ERK1 phosphorylation site on Xenopus hnRNPK (S257; homologous with S284 of human hnRNPK) using an in vitro phosphorylation assay and tested its function in vivo by expressing phosphomimetic (S257D) and phosphodeficient (S257A) forms of hnRNPK in Xenopus embryos. Although neither form altered hnRNPK nuclear export, only the phosphomimetic form significantly rescued both neurofilament protein expression and axon outgrowth from hnRNPK knockdown. This finding represents a previously unidentified function of phosphorylation at this phylogenetically conserved site and implicates hnRNPK as an intracellular molecular target of ERK-mediated signaling in axon outgrowth.


Assuntos
Axônios/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Xenopus/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Embrião não Mamífero , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Dados de Sequência Molecular , Mutação , Proteínas de Neurofilamentos/metabolismo , Fosforilação , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Ribonucleoproteínas/metabolismo
13.
J Neurosci ; 33(37): 14666-80, 2013 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-24027268

RESUMO

c-Jun N-terminal kinase (JNK) mediates cell signaling essential for axon outgrowth, but the associated substrates and underlying mechanisms are poorly understood. We identified in Xenopus laevis embryos a novel posttranscriptional mechanism whereby JNK regulates axonogenesis by phosphorylating a specific site on heterogeneous nuclear ribonucleoprotein K (hnRNP K). Both JNK inhibition and hnRNP K knockdown inhibited axon outgrowth and translation of hnRNP K-regulated cytoskeletal RNAs (tau and neurofilament medium), effects that were alleviated by expressing phosphomimetic, but not phosphodeficient, forms of hnRNP K. Immunohistochemical and biochemical analyses indicated that JNK phosphorylation of hnRNP K occurred within the cytoplasm and was necessary for the translational initiation of hnRNP K-targeted RNAs but not for hnRNP K intracellular localization or RNA binding. Thus, in addition to its known roles in transcription and cytoskeletal organization, JNK acts posttranscriptionally through hnRNP K to regulate translation of proteins crucial for axonogenesis.


Assuntos
Axônios/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/citologia , Processamento de Proteína Pós-Traducional/genética , Animais , Animais Geneticamente Modificados , Axônios/efeitos dos fármacos , Padronização Corporal/genética , Encéfalo/citologia , Encéfalo/embriologia , Células Cultivadas , Técnicas de Cocultura , Proteínas do Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/citologia , Neurônios/efeitos dos fármacos , Periferinas/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Medula Espinal/citologia , Proteínas de Xenopus/metabolismo , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...