Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 270
Filtrar
1.
Dev Growth Differ ; 66(5): 320-328, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38925637

RESUMEN

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


Asunto(s)
Placa Neural , Tubo Neural , Xenopus laevis , Animales , Tubo Neural/embriología , Tubo Neural/citología , Tubo Neural/metabolismo , Placa Neural/embriología , Placa Neural/metabolismo , Placa Neural/citología , Xenopus laevis/embriología , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ectodermo/citología , Ectodermo/metabolismo , Microscopía de Fuerza Atómica , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Embrión no Mamífero/embriología
2.
Mol Cells ; 47(6): 100076, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38825188

RESUMEN

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


Asunto(s)
Movimiento Celular , Destrina , Desarrollo Embrionario , Xenopus laevis , Animales , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Destrina/metabolismo , Destrina/genética , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Cresta Neural/metabolismo , Cresta Neural/embriología , Cresta Neural/citología , Neurogénesis , Placa Neural/metabolismo , Placa Neural/embriología , Actinas/metabolismo , Regulación del Desarrollo de la Expresión Génica
3.
Development ; 151(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38828854

RESUMEN

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


Asunto(s)
Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , MicroARNs , Placa Neural , Factor de Transcripción AP-2 , Animales , MicroARNs/genética , MicroARNs/metabolismo , Embrión de Pollo , Metilación de ADN/genética , Placa Neural/metabolismo , Placa Neural/embriología , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A/metabolismo , Regiones Promotoras Genéticas/genética , Sitios de Unión
4.
Mol Genet Metab ; 142(2): 108492, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38759397

RESUMEN

Pathogenic variants in the O-GlcNAc transferase gene (OGT) have been associated with a congenital disorder of glycosylation (OGT-CDG), presenting with intellectual disability which may be of neuroectodermal origin. To test the hypothesis that pathology is linked to defects in differentiation during early embryogenesis, we developed an OGT-CDG induced pluripotent stem cell line together with isogenic control generated by CRISPR/Cas9 gene-editing. Although the OGT-CDG variant leads to a significant decrease in OGT and O-GlcNAcase protein levels, there were no changes in differentiation potential or stemness. However, differentiation into ectoderm resulted in significant differences in O-GlcNAc homeostasis. Further differentiation to neuronal stem cells revealed differences in morphology between patient and control lines, accompanied by disruption of the O-GlcNAc pathway. This suggests a critical role for O-GlcNAcylation in early neuroectoderm architecture, with robust compensatory mechanisms in the earliest stages of stem cell differentiation.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , N-Acetilglucosaminiltransferasas , Placa Neural , Fenotipo , Humanos , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Placa Neural/metabolismo , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/patología , Trastornos Congénitos de Glicosilación/metabolismo , Sistemas CRISPR-Cas , Glicosilación , Edición Génica , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología
5.
Commun Biol ; 7(1): 415, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38580843

RESUMEN

The ten-eleven-translocation family of proteins (TET1/2/3) are epigenetic regulators of gene expression. They regulate genes by promoting DNA demethylation (i.e., catalytic activity) and by partnering with regulatory proteins (i.e., non-catalytic functions). Unlike Tet1 and Tet2, Tet3 is not expressed in mouse embryonic stem cells (ESCs) but is induced upon ESC differentiation. However, the significance of its dual roles in lineage specification is less defined. By generating TET3 catalytic-mutant (Tet3m/m) and knockout (Tet3-/-) mouse ESCs and differentiating them to neuroectoderm (NE), we identify distinct catalytic-dependent and independent roles of TET3 in NE specification. We find that the catalytic activity of TET3 is important for activation of neural genes while its non-catalytic functions are involved in suppressing mesodermal programs. Interestingly, the vast majority of differentially methylated regions (DMRs) in Tet3m/m and Tet3-/- NE cells are hypomethylated. The hypo-DMRs are associated to aberrantly upregulated genes while the hyper-DMRs are linked to downregulated neural genes. We find the maintenance methyltransferase Dnmt1 as a direct target of TET3, which is downregulated in TET3-deficient NE cells and may contribute to the increased DNA hypomethylation. Our findings establish that the catalytic-dependent and -independent roles of TET3 have distinct contributions to NE specification with potential implications in development.


Asunto(s)
Dioxigenasas , Animales , Ratones , Diferenciación Celular/genética , Dioxigenasas/genética , Dioxigenasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Placa Neural/metabolismo
6.
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
7.
Sci Adv ; 10(9): eadj5107, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38427725

RESUMEN

Cell fate decisions are achieved with gene expression changes driven by lineage-specific transcription factors (TFs). These TFs depend on chromatin remodelers including the Brahma-related gene 1 (BRG1)-associated factor (BAF) complex to activate target genes. BAF complex subunits are essential for development and frequently mutated in cancer. Thus, interrogating how BAF complexes contribute to cell fate decisions is critical for human health. We examined the requirement for the catalytic BAF subunit BRG1 in neural progenitor cell (NPC) specification from human embryonic stem cells. During the earliest stages of differentiation, BRG1 was required to establish chromatin accessibility at neuroectoderm-specific enhancers. Depletion of BRG1 dorsalized NPCs and promoted precocious neural crest specification and enhanced neuronal differentiation. These findings demonstrate that BRG1 mediates NPC specification by ensuring proper expression of lineage-specific TFs and appropriate activation of their transcriptional programs.


Asunto(s)
Cromatina , Placa Neural , Humanos , Cromatina/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Placa Neural/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
8.
Nat Commun ; 15(1): 1642, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38388461

RESUMEN

Folate supplementation reduces the occurrence of neural tube defects (NTDs), birth defects consisting in the failure of the neural tube to form and close. The mechanisms underlying NTDs and their prevention by folate remain unclear. Here we show that folate receptor 1 (FOLR1) is necessary for the formation of neural tube-like structures in human-cell derived neural organoids. FOLR1 knockdown in neural organoids and in Xenopus laevis embryos leads to NTDs that are rescued by pteroate, a folate precursor that is unable to participate in metabolism. We demonstrate that FOLR1 interacts with and opposes the function of CD2-associated protein, molecule essential for apical endocytosis and turnover of C-cadherin in neural plate cells. In addition, folates increase Ca2+ transient frequency, suggesting that folate and FOLR1 signal intracellularly to regulate neural plate folding. This study identifies a mechanism of action of folate distinct from its vitamin function during neural tube formation.


Asunto(s)
Ácido Fólico , Defectos del Tubo Neural , Humanos , Ácido Fólico/metabolismo , Tubo Neural/metabolismo , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Placa Neural/metabolismo
9.
Development ; 150(19)2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37756587

RESUMEN

The Foxi3 transcription factor, expressed in the neural plate border at the end of gastrulation, is necessary for the formation of posterior placodes and is thus important for ectodermal patterning. We have created two knock-in mouse lines expressing GFP or a tamoxifen-inducible Cre recombinase to show that Foxi3 is one of the earliest genes to label the border between the neural tube and epidermis, and that Foxi3-expressing neural plate border progenitors contribute primarily to cranial placodes and epidermis from the onset of expression, but not to the neural crest or neural tube lineages. By simultaneously knocking out Foxi3 in neural plate border cells and following their fates, we show that neural plate border cells lacking Foxi3 contribute to all four lineages of the ectoderm - placodes, epidermis, crest and neural tube. We contrast Foxi3 with another neural plate border transcription factor, Zic5, the progenitors of which initially contribute broadly to all germ layers until gastrulation and gradually become restricted to the neural crest lineage and dorsal neural tube cells. Our study demonstrates that Foxi3 uniquely acts early at the neural plate border to restrict progenitors to a placodal and epidermal fate.


Asunto(s)
Placa Neural , Factores de Transcripción , Animales , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Placa Neural/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ectodermo/metabolismo , Cresta Neural/metabolismo , Factores de Transcripción Forkhead/metabolismo
10.
Biol Rev Camb Philos Soc ; 98(6): 2271-2283, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37534608

RESUMEN

Neurulation transforms the neuroectoderm into the neural tube. This transformation relies on reorganising the configurational relationships between the orientations of intrinsic polarities of neighbouring cells. These orientational intercellular relationships are established, maintained, and modulated by orientational cell adhesions (OCAs). Here, using zebrafish (Danio rerio) neurulation as a major model, we propose a new perspective on how OCAs contribute to the parallel, antiparallel, and opposing intercellular relationships that underlie the neural plate-keel-rod-tube transformation, a stepwise process of cell aggregation followed by cord hollowing. We also discuss how OCAs in neurulation may be regulated by various adhesion molecules, including cadherins, Eph/Ephrins, Claudins, Occludins, Crumbs, Na+ /K+ -ATPase, and integrins. By comparing neurulation among species, we reveal that antiparallel OCAs represent a conserved mechanism for the fusion of the neural tube. Throughout, we highlight some outstanding questions regarding OCAs in neurulation. Answers to these questions will help us understand better the mechanisms of tubulogenesis of many tissues.


Asunto(s)
Neurulación , Pez Cebra , Animales , Adhesión Celular , Tubo Neural/metabolismo , Placa Neural/metabolismo
11.
Development ; 150(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37213081

RESUMEN

The most anterior structure of the ascidian larva is made of three palps with sensory and adhesive functions essential for metamorphosis. They derive from the anterior neural border and their formation is regulated by FGF and Wnt. Given that they also share gene expression profiles with vertebrate anterior neural tissue and cranial placodes, their study should shed light on the emergence of the unique vertebrate telencephalon. We show that BMP signaling regulates two phases of palp formation in Ciona intestinalis. During gastrulation, the anterior neural border is specified in a domain of inactive BMP signaling, and activating BMP prevented its formation. During neurulation, BMP defines ventral palp identity and indirectly specifies the inter-papilla territory separating the ventral and dorsal palps. Finally, we show that BMP has similar functions in the ascidian Phallusia mammillata, for which we identified novel palp markers. Collectively, we provide a better molecular description of palp formation in ascidians that will be instrumental for comparative studies.


Asunto(s)
Urocordados , Animales , Urocordados/genética , Sistema Nervioso/metabolismo , Transducción de Señal , Gastrulación/genética , Placa Neural/metabolismo , Regulación del Desarrollo de la Expresión Génica
12.
Dev Genes Evol ; 233(1): 13-23, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37079132

RESUMEN

Cranial neurogenic placodes have been considered vertebrate innovations. However, anterior neural plate border (ANB) cells of ascidian embryos share many properties with vertebrate neurogenic placodes; therefore, it is now believed that the last common ancestor of vertebrates and ascidians had embryonic structures similar to neurogenic placodes of vertebrate embryos. Because BMP signaling is important for specifying the placode region in vertebrate embryos, we examined whether BMP signaling is also involved in gene expression in the ANB region of ascidian embryos. Our data indicated that Admp, a divergent BMP family member, is mainly responsible for BMP signaling in the ANB region, and that two BMP-antagonists, Noggin and Chordin, restrict the domain, in which BMP signaling is activated, to the ANB region, and prevent it from expanding to the neural plate. BMP signaling is required for expression of Foxg and Six1/2 at the late gastrula stage, and also for expression of Zf220, which encodes a zinc finger transcription factor in late neurula embryos. Because Zf220 negatively regulates Foxg, when we downregulated Zf220 by inhibiting BMP signaling, Foxg was upregulated, resulting in one large palp instead of three palps (adhesive organs derived from ANB cells). Functions of BMP signaling in specification of the ANB region give further support to the hypothesis that ascidian ANB cells share an evolutionary origin with vertebrate cranial placodes.


Asunto(s)
Urocordados , Animales , Urocordados/genética , Placa Neural/metabolismo , Vertebrados/genética , Evolución Biológica , Proteínas Morfogenéticas Óseas/metabolismo , Regulación del Desarrollo de la Expresión Génica
13.
Dev Biol ; 496: 87-94, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36739958

RESUMEN

The nervous system has various types of cells derived from three neuroectodermal regions: neural plate (NP), neural crest (NC), and preplacodal ectoderm (PPE). Differentiation of these regions is regulated by various morphogens. However, regulatory mechanisms of morphogen distribution in neural patterning are still debated. In general, an extracellular component, heparan sulfate (HS), is essential to regulate morphogen gradients by modulating morphogen binding. The present study focused on an HS modification enzyme, heparan sulfate 6-O-sulfotransferase 1 (Hs6st1), which is highly expressed during the neurula stage in Xenopus. Our present in situ hybridization analysis revealed that Hs6st1 is expressed in the lateral sensorial layer of neuroectoderm. Overexpression of Hs6st1 expands Sox3 (NP marker gene) expression, and slightly dampens FoxD3 (NC marker) expression. Hs6st1 knockout using the CRISPR/Cas9 system also expands the neural plate region, followed by retinal malformation. These results imply that 6-O sulfation, mediated by Hs6st1, selectively regulates morphogen distribution required for neuroectodermal patterning. Among morphogens required for patterning, Fgf8a accumulates on Hs6st1-expressing cells, whereas a secreted BMP antagonist, Noggin, diffuses away from those cells. Thus, cell-autonomous 6-O sulfation of HS at the sensorial layer of neuroectoderm also affects neuroectodermal patterning in neighboring regions, including neural plate and neural crest, not only through accumulation, but also through dispersal of specific morphogens.


Asunto(s)
Heparitina Sulfato , Placa Neural , Animales , Xenopus laevis/metabolismo , Placa Neural/metabolismo , Heparitina Sulfato/metabolismo , Ectodermo/metabolismo , Cresta Neural/metabolismo , Proteínas de Xenopus/metabolismo , Factores de Transcripción SOXB1
14.
Dev Biol ; 492: 14-24, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36162552

RESUMEN

The vertebrate nervous system develops from embryonic neural plate and neural crest. Although genetic mechanisms governing vertebrate neural development have been investigated in depth, epigenetic regulation of this process remains less understood. Redundancy of epigenetic factors and early lethality of animals deficient in critical epigenetic components pose major challenges in characterization of epigenetic factors in vertebrate neural development. In this study, we use the amphibian model Xenopus laevis to investigate the roles of non-redundant, obligatory components of all histone H3K4 activating methylation complexes (COMPASS, also known as SET1/MLL complexes) in early neural development. The two genes that we focus on, Ash2l and Dpy30, regulate mesendodermal differentiation in mouse embryonic stem cells and cause early embryonic lethality when removed from mouse embryos. Using targeted knockdown of the genes in dorsal ectoderm of Xenopus that gives rise to future nervous system, we show here that ash2l and dpy30 are required for neural and neural crest marker expression in Xenopus late neurula embryos but are dispensable for early neural and neural plate border gene expression. Co-immunoprecipitation assays reveal that Dpy30 and Ash2L associate with the neural plate border transcription factors, such as Msx1 and Tfap2a. Chromatin immunoprecipitation (ChIP) assay further demonstrates that Ash2L and the H3K4me3 active histone mark accumulate at the promoter regions of the neural crest gene sox10 in a Tfap2a-dependent manner. Collectively, our data suggest that Ash2l and Dpy30 interact with specific transcription factors to recruit COMPASS complexes to the regulatory regions of neural crest specification genes to control their expression and influence development of the nervous system during vertebrate embryogenesis.


Asunto(s)
Epigénesis Genética , Cresta Neural , Animales , Ratones , Regulación del Desarrollo de la Expresión Génica/genética , Metilación , Placa Neural/metabolismo , Factores de Transcripción/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
15.
Dev Biol ; 489: 62-75, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35697116

RESUMEN

Mcrs1 is a multifunctional protein that is critical for many cellular processes in a wide range of cell types. Previously, we showed that Mcrs1 binds to the Six1 transcription factor and reduces the ability of the Six1-Eya1 complex to upregulate transcription, and that Mcrs1 loss-of-function leads to the expansion of several neural plate genes, reduction of neural border and pre-placodal ectoderm (PPR) genes, and pleiotropic effects on various neural crest (NC) genes. Because the affected embryonic structures give rise to several of the cranial tissues affected in Branchio-otic/Branchio-oto-renal (BOR) syndrome, herein we tested whether these gene expression changes subsequently alter the development of the proximate precursors of BOR affected structures - the otic vesicles (OV) and branchial arches (BA). We found that Mcrs1 is required for the expression of several OV genes involved in inner ear formation, patterning and otic capsule cartilage formation. Mcrs1 knockdown also reduced the expression domains of many genes expressed in the larval BA, derived from either NC or PPR, except for emx2, which was expanded. Reduced Mcrs1 also diminished the length of the expression domain of tbx1 in BA1 and BA2 and interfered with cranial NC migration from the dorsal neural tube; this subsequently resulted in defects in the morphology of lower jaw cartilages derived from BA1 and BA2, including the infrarostral, Meckel's, and ceratohyal as well as the otic capsule. These results demonstrate that Mcrs1 plays an important role in processes that lead to the formation of craniofacial cartilages and its loss results in phenotypes consistent with reduced Six1 activity associated with BOR.


Asunto(s)
Región Branquial , Síndrome Branquio Oto Renal , Región Branquial/metabolismo , Síndrome Branquio Oto Renal/genética , Síndrome Branquio Oto Renal/metabolismo , Cartílago/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Cresta Neural , Placa Neural/metabolismo , Proteínas de Unión al ARN/metabolismo
16.
Dev Biol ; 488: 81-90, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35598626

RESUMEN

Pre-placodal ectoderm (PPE), a horseshoe-shaped narrow region formed during early vertebrate development, gives rise to multiple types of sensory organs and ganglia. For PPE induction, a certain level of FGF signal activation is required. However, it is difficult to reproducibly induce the narrow region with variations in gene expression, including FGF, among individuals. An intracellular regulatory factor of FGF signaling, Dusp6, is expressed by FGF signal activation and inactivates a downstream regulator, ERK1/2, in adult tissues; however, its role in early development is not well known. Here, we reveal that Dusp6 is expressed in an FGF-dependent manner in Xenopus PPE. Gain- and loss-of-function experiments showed that Dusp6 is required for expression of a PPE gene, Six1, and patterning of adjacent regions, neural plate, and neural crest. To reveal the importance of Dusp6 in variable FGF production, we performed Dusp6 knockdown with FGF-bead implantation, which resulted in varying Six1 expression patterns. Taken together, these results suggest that Dusp6 is required for PPE formation and that it contributes to the robust patterning of PPE by mediating FGF signaling.


Asunto(s)
Ectodermo , Placa Neural , Animales , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Ectodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Humanos , Cresta Neural/metabolismo , Placa Neural/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
17.
Methods Mol Biol ; 2438: 147-161, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35147941

RESUMEN

Planar cell polarity (PCP) refers to coordinated cell polarization in the plane of the tissue. Genetic studies in Drosophila identified several core PCP genes, whose products function together in a signaling pathway that regulates cell shape, epithelial tissue organization and remodeling during morphogenesis. PCP is detected by the asymmetric distribution of core PCP proteins at different borders of epithelial cells. Believed to be critical for signaling, this segregation is studied by a variety of techniques, such as direct immunostaining and imaging of fluorescent PCP protein fusions or fluorescence recovery after photobleaching (FRAP). All of the above techniques can be applied to the analysis of the Xenopus neural plate to study the dynamics of tissue polarization, making this system one of the best vertebrate PCP models. This chapter describes how to image PCP proteins in Xenopus neuroectoderm for both fixed and live samples. These robust cellular techniques will contribute to mechanistic studies of PCP in vertebrate embryos.


Asunto(s)
Polaridad Celular , Placa Neural , Animales , Polaridad Celular/genética , Proteínas de la Membrana/metabolismo , Placa Neural/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
18.
PLoS Genet ; 18(1): e1010012, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35041640

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Precursores del ARN/metabolismo , ARN Ribosómico 18S/metabolismo , ARN Ribosómico 28S/metabolismo , Xenopus laevis/crecimiento & desarrollo , Animales , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Técnicas de Silenciamiento del Gen , Metilación , Cresta Neural/crecimiento & desarrollo , Cresta Neural/metabolismo , Placa Neural/crecimiento & desarrollo , Placa Neural/metabolismo , Procesamiento Postranscripcional del ARN , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética
19.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35042818

RESUMEN

The protovertebrate Ciona intestinalis type A (sometimes called Ciona robusta) contains a series of sensory cell types distributed across the head-tail axis of swimming tadpoles. They arise from lateral regions of the neural plate that exhibit properties of vertebrate placodes and neural crest. The sensory determinant POU IV/Brn3 is known to work in concert with regional determinants, such as Foxg and Neurogenin, to produce palp sensory cells (PSCs) and bipolar tail neurons (BTNs), in head and tail regions, respectively. A combination of single-cell RNA-sequencing (scRNA-seq) assays, computational analysis, and experimental manipulations suggests that misexpression of POU IV results in variable transformations of epidermal cells into hybrid sensory cell types, including those exhibiting properties of both PSCs and BTNs. Hybrid properties are due to coexpression of Foxg and Neurogenin that is triggered by an unexpected POU IV feedback loop. Hybrid cells were also found to express a synthetic gene battery that is not coexpressed in any known cell type. We discuss these results with respect to the opportunities and challenges of reprogramming cell types through the targeted misexpression of cellular determinants.


Asunto(s)
Ciona intestinalis/genética , Neuronas/metabolismo , Factores del Dominio POU/metabolismo , Animales , Evolución Biológica , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Ciona intestinalis/metabolismo , Epidermis/inervación , Epidermis/metabolismo , Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Cresta Neural/metabolismo , Placa Neural/metabolismo , Factores del Dominio POU/genética , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Vertebrados/genética
20.
Dev Biol ; 483: 66-75, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34968443

RESUMEN

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


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Inactivación de Genes/métodos , MicroARNs/genética , Xenopus laevis/embriología , Xenopus laevis/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Hibridación in Situ/métodos , Morfolinos/genética , Cresta Neural/embriología , Cresta Neural/metabolismo , Placa Neural/embriología , Placa Neural/metabolismo , Neurulación/genética , Fenotipo , ARN Guía de Kinetoplastida/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA