Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.606
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(19): e2311685121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38683994

RESUMEN

Neural crest cells exemplify cellular diversification from a multipotent progenitor population. However, the full sequence of early molecular choices orchestrating the emergence of neural crest heterogeneity from the embryonic ectoderm remains elusive. Gene-regulatory-networks (GRN) govern early development and cell specification toward definitive neural crest. Here, we combine ultradense single-cell transcriptomes with machine-learning and large-scale transcriptomic and epigenomic experimental validation of selected trajectories, to provide the general principles and highlight specific features of the GRN underlying neural crest fate diversification from induction to early migration stages using Xenopus frog embryos as a model. During gastrulation, a transient neural border zone state precedes the choice between neural crest and placodes which includes multiple converging gene programs. During neurulation, transcription factor connectome, and bifurcation analyses demonstrate the early emergence of neural crest fates at the neural plate stage, alongside an unbiased multipotent-like lineage persisting until epithelial-mesenchymal transition stage. We also decipher circuits driving cranial and vagal neural crest formation and provide a broadly applicable high-throughput validation strategy for investigating single-cell transcriptomes in vertebrate GRNs in development, evolution, and disease.


Asunto(s)
Cresta Neural , Análisis de la Célula Individual , Xenopus laevis , Animales , Cresta Neural/citología , Cresta Neural/metabolismo , Análisis de la Célula Individual/métodos , Xenopus laevis/embriología , Regulación del Desarrollo de la Expresión Génica , Movimiento Celular , Redes Reguladoras de Genes , Transcriptoma , Gastrulación , Placa Neural/metabolismo , Placa Neural/embriología , Placa Neural/citología , Transición Epitelial-Mesenquimal/genética , Embrión no Mamífero/metabolismo , Embrión no Mamífero/citología , Neurulación/genética , Neurulación/fisiología , Diferenciación Celular
2.
Dev Biol ; 511: 63-75, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38621649

RESUMEN

Loss of function variations in the dual specificity tyrosine-phosphorylation-regulated kinase 1 A (DYRK1A) gene are associated with craniofacial malformations in humans. Here we characterized the effects of deficient DYRK1A in craniofacial development using a developmental model, Xenopus laevis. Dyrk1a mRNA and protein were expressed throughout the developing head and both were enriched in the branchial arches which contribute to the face and jaw. Consistently, reduced Dyrk1a function, using dyrk1a morpholinos and pharmacological inhibitors, resulted in orofacial malformations including hypotelorism, altered mouth shape, slanted eyes, and narrower face accompanied by smaller jaw cartilage and muscle. Inhibition of Dyrk1a function resulted in misexpression of key craniofacial regulators including transcription factors and members of the retinoic acid signaling pathway. Two such regulators, sox9 and pax3 are required for neural crest development and their decreased expression corresponds with smaller neural crest domains within the branchial arches. Finally, we determined that the smaller size of the faces, jaw elements and neural crest domains in embryos deficient in Dyrk1a could be explained by increased cell death and decreased proliferation. This study is the first to provide insight into why craniofacial birth defects might arise in humans with variants of DYRK1A.


Asunto(s)
Quinasas DyrK , Regulación del Desarrollo de la Expresión Génica , Cresta Neural , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , Proteínas de Xenopus , Xenopus laevis , Animales , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Cresta Neural/embriología , Cresta Neural/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Transducción de Señal , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/metabolismo , Región Branquial/embriología , Región Branquial/metabolismo , Embrión no Mamífero/metabolismo , Embrión no Mamífero/embriología
3.
Dis Model Mech ; 17(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38501224

RESUMEN

De novo truncating variants in fibrosin-like 1 (FBRSL1), a member of the AUTS2 gene family, cause a disability syndrome, including organ malformations such as heart defects. Here, we use Xenopus laevis to investigate whether Fbrsl1 plays a role in heart development. Xenopus laevis fbrsl1 is expressed in tissues relevant for heart development, and morpholino-mediated knockdown of Fbrsl1 results in severely hypoplastic hearts. Our data suggest that Fbrsl1 is required for the development of the first heart field, which contributes to the ventricle and the atria, but not for the second heart field, which gives rise to the outflow tract. The morphant heart phenotype could be rescued using a human N-terminal FBRSL1 isoform that contains an alternative exon, but lacks the AUTS2 domain. N-terminal isoforms carrying patient variants failed to rescue. Interestingly, a long human FBRSL1 isoform, harboring the AUTS2 domain, also did not rescue the morphant heart defects. Thus, our data suggest that different FBRSL1 isoforms may have distinct functions and that only the short N-terminal isoform, appears to be critical for heart development.


Asunto(s)
Cardiopatías Congénitas , Corazón , Isoformas de Proteínas , Proteínas de Xenopus , Xenopus laevis , Animales , Xenopus laevis/embriología , Humanos , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Corazón/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Fenotipo , Técnicas de Silenciamiento del Gen , Regulación del Desarrollo de la Expresión Génica
4.
Dev Cell ; 59(8): 1058-1074.e11, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38460509

RESUMEN

During oocyte maturation and early embryogenesis, changes in mRNA poly(A)-tail lengths strongly influence translation, but how these tail-length changes are orchestrated has been unclear. Here, we performed tail-length and translational profiling of mRNA reporter libraries (each with millions of 3' UTR sequence variants) in frog oocytes and embryos and in fish embryos. Contrasting to previously proposed cytoplasmic polyadenylation elements (CPEs), we found that a shorter element, UUUUA, together with the polyadenylation signal (PAS), specify cytoplasmic polyadenylation, and we identified contextual features that modulate the activity of both elements. In maturing oocytes, this tail lengthening occurs against a backdrop of global deadenylation and the action of C-rich elements that specify tail-length-independent translational repression. In embryos, cytoplasmic polyadenylation becomes more permissive, and additional elements specify waves of stage-specific deadenylation. Together, these findings largely explain the complex tapestry of tail-length changes observed in early frog and fish development, with strong evidence of conservation in both mice and humans.


Asunto(s)
Regiones no Traducidas 3' , Oocitos , Poli A , Poliadenilación , Biosíntesis de Proteínas , ARN Mensajero , Animales , Oocitos/metabolismo , Oocitos/citología , Poli A/metabolismo , Poli A/genética , Regiones no Traducidas 3'/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Humanos , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Femenino , Xenopus laevis/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética , Citoplasma/metabolismo
5.
Gene Expr Patterns ; 48: 119318, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37011704

RESUMEN

Development of the vertebrate embryo requires strict coordination of a highly complex series of signaling cascades, that drive cell proliferation, differentiation, migration, and the general morphogenetic program. Members of the Map kinase signaling pathway are repeatedly required throughout development to activate the downstream effectors, ERK, p38, and JNK. Regulation of these pathways occurs at many levels in the signaling cascade, with the Map3Ks playing an essential role in target selection. The thousand and one amino acid kinases (Taoks) are Map3Ks that have been shown to activate both p38 and JNK and are linked to neurodevelopment in both invertebrate and vertebrate organisms. In vertebrates, there are three Taok paralogs (Taok1, Taok2, and Taok3) which have not yet been ascribed a role in early development. Here we describe the spatiotemporal expression of Taok1, Taok2, and Taok3 in the model organism Xenopus laevis. The X. laevis Tao kinases share roughly 80% identity to each other, with the bulk of the conservation in the kinase domain. Taok1 and Taok3 are highly expressed in pre-gastrula and gastrula stage embryos, with initial expression localized to the animal pole and later expression in the ectoderm and mesoderm. All three Taoks are expressed in the neural and tailbud stages, with overlapping expression in the neural tube, notochord, and many anterior structures (including branchial arches, brain, otic vesicles, and eye). The expression patterns described here provide evidence that the Tao kinases may play a central role in early development, in addition to their function during neural development, and establish a framework to better understand the developmental roles of Tao kinase signaling.


Asunto(s)
Desarrollo Embrionario , Quinasas Quinasa Quinasa PAM , Proteínas de Xenopus , Xenopus laevis , Animales , Desarrollo Embrionario/genética , Gástrula , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Xenopus laevis/embriología , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
6.
Cells ; 12(6)2023 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-36980215

RESUMEN

The reciprocal inhibition between two signaling centers, the Spemann organizer (dorsal mesoderm) and ventral region (mesoderm and ectoderm), collectively regulate the overall development of vertebrate embryos. Each center expresses key homeobox transcription factors (TFs) that directly control target gene transcription. Goosecoid (Gsc) is an organizer (dorsal mesoderm)-specific TF known to induce dorsal fate and inhibit ventral/ectodermal specification. Ventx1.1 (downstream of Bmp signaling) induces the epidermal lineage and inhibits dorsal organizer-specific genes from the ventral region. Chordin (Chrd) is an organizer-specific secreted Bmp antagonist whose expression is primarily activated by Gsc. Alternatively, chrd expression is repressed by Bmp/Ventx1.1 in the ventral/epidermal region. However, the regulatory mechanisms underlying the transcription mediated by Gsc and Ventx1.1 remain elusive. Here, we found that the chrd promoter contained two cis-acting response elements that responded negatively to Ventx1.1 and positively to Gsc. In the ventral/ectodermal region, Ventx1.1 was directly bound to the Ventx1.1 response element (VRE) and inhibited chrd transcription. In the organizer region, Gsc was bound to the Gsc response elements (GRE) to activate chrd transcription. The Gsc-mediated positive response on the chrd promoter completely depended on another adjacent Wnt response cis-acting element (WRE), which was the TCF7 (also known as Tcf1) binding element. Site-directed mutagenesis of VRE, GRE, or WRE completely abolished the repressive or activator activity of Ventx1.1 and Gsc, respectively. The ChIP-PCR results confirmed the direct binding of Ventx1.1 and Gsc/Tcf7 to VRE and GRE/WRE, respectively. These results demonstrated that chrd expression is oppositely modulated by homeobox TFs, Ventx1.1, and Gsc/Tcf7 during the embryonic patterning of Xenopus gastrula.


Asunto(s)
Gástrula , Glicoproteínas , Proteína Goosecoide , Factores de Transcripción , Proteínas de Xenopus , Xenopus laevis , Animales , Gástrula/metabolismo , Genes Homeobox , Proteína Goosecoide/genética , Proteína Goosecoide/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Glicoproteínas/metabolismo
7.
Nature ; 613(7945): 712-720, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36653451

RESUMEN

Ribosomes are produced in large quantities during oogenesis and are stored in the egg. However, the egg and early embryo are translationally repressed1-4. Here, using mass spectrometry and cryo-electron microscopy analyses of ribosomes isolated from zebrafish (Danio rerio) and Xenopus laevis eggs and embryos, we provide molecular evidence that ribosomes transition from a dormant state to an active state during the first hours of embryogenesis. Dormant ribosomes are associated with four conserved factors that form two modules, consisting of Habp4-eEF2 and death associated protein 1b (Dap1b) or Dap in complex with eIF5a. Both modules occupy functionally important sites and act together to stabilize ribosomes and repress translation. Dap1b (also known as Dapl1 in mammals) is a newly discovered translational inhibitor that stably inserts into the polypeptide exit tunnel. Addition of recombinant zebrafish Dap1b protein is sufficient to block translation and reconstitute the dormant egg ribosome state in a mammalian translation extract in vitro. Thus, a developmentally programmed, conserved ribosome state has a key role in ribosome storage and translational repression in the egg.


Asunto(s)
Secuencia Conservada , Evolución Molecular , Óvulo , Biosíntesis de Proteínas , Ribosomas , Proteínas de Xenopus , Proteínas de Pez Cebra , Animales , Microscopía por Crioelectrón/métodos , Péptidos/metabolismo , Ribosomas/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Espectrometría de Masas , Xenopus laevis/embriología , Óvulo/metabolismo , Estructuras Embrionarias , Desarrollo Embrionario , Femenino , Factor 5A Eucariótico de Iniciación de Traducción
8.
Kidney Int ; 103(1): 77-86, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36055600

RESUMEN

The kidney is an essential organ that ensures bodily fluid homeostasis and removes soluble waste products from the organism. Nephrons, the functional units of the kidney, comprise a blood filter, the glomerulus or glomus, and an epithelial tubule that processes the filtrate from the blood or coelom and selectively reabsorbs solutes, such as sugars, proteins, ions, and water, leaving waste products to be eliminated in the urine. Genes coding for transporters are segmentally expressed, enabling the nephron to sequentially process the filtrate. The Xenopus embryonic kidney, the pronephros, which consists of a single large nephron, has served as a valuable model to identify genes involved in nephron formation and patterning. Therefore, the developmental patterning program that generates these segments is of great interest. Prior work has defined the gene expression profiles of Xenopus nephron segments via in situ hybridization strategies, but a comprehensive understanding of the cellular makeup of the pronephric kidney remains incomplete. Here, we carried out single-cell mRNA sequencing of the functional Xenopus pronephric nephron and evaluated its cellular composition through comparative analyses with previous Xenopus studies and single-cell mRNA sequencing of the adult mouse kidney. This study reconstructs the cellular makeup of the pronephric kidney and identifies conserved cells, segments, and associated gene expression profiles. Thus, our data highlight significant conservation in podocytes, proximal and distal tubule cells, and divergence in cellular composition underlying the capacity of each nephron to remove wastes in the form of urine, while emphasizing the Xenopus pronephros as a model for physiology and disease.


Asunto(s)
Riñón , Nefronas , Animales , Ratones , Regulación del Desarrollo de la Expresión Génica , Riñón/embriología , Glomérulos Renales/embriología , Nefronas/embriología , ARN Mensajero/genética , Xenopus laevis/embriología
9.
Sci Rep ; 12(1): 1903, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115663

RESUMEN

Xenopus laevis tadpoles possess high regenerative ability and can regenerate functional tails after amputation. An early event in regeneration is the induction of undifferentiated cells that form the regenerated tail. We previously reported that interleukin-11 (il11) is upregulated immediately after tail amputation to induce undifferentiated cells of different cell lineages, indicating a key role of il11 in initiating tail regeneration. As Il11 is a secretory factor, Il11 receptor-expressing cells are thought to mediate its function. X. laevis has a gene annotated as interleukin 11 receptor subunit alpha on chromosome 1L (il11ra.L), a putative subunit of the Il11 receptor complex, but its function has not been investigated. Here, we show that nuclear localization of phosphorylated Stat3 induced by Il11 is abolished in il11ra.L knocked-out culture cells, strongly suggesting that il11ra.L encodes an Il11 receptor component. Moreover, knockdown of il11ra.L impaired tadpole tail regeneration, suggesting its indispensable role in tail regeneration. We also provide a model showing that Il11 functions via il11ra.L-expressing cells in a non-cell autonomous manner. These results highlight the importance of il11ra.L-expressing cells in tail regeneration.


Asunto(s)
Proliferación Celular , Subunidad alfa del Receptor de Interleucina-11/metabolismo , Larva/metabolismo , Regeneración , Cola (estructura animal)/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Interleucina-11/farmacología , Subunidad alfa del Receptor de Interleucina-11/agonistas , Subunidad alfa del Receptor de Interleucina-11/genética , Larva/efectos de los fármacos , Larva/genética , Larva/crecimiento & desarrollo , Fosforilación , Regeneración/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Cola (estructura animal)/efectos de los fármacos , Cola (estructura animal)/embriología , Proteínas de Xenopus/genética , Xenopus laevis/embriología , Xenopus laevis/genética
10.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35193956

RESUMEN

The development of functional topography in the developing brain follows a progression from initially coarse to more precisely organized maps. To examine the emergence of topographically organized maps in the retinotectal system, we performed longitudinal visual receptive field mapping by calcium imaging in the optic tectum of GCaMP6-expressing transgenic Xenopus laevis tadpoles. At stage 42, just 1 d after retinal axons arrived in the optic tectum, a clear retinotopic azimuth map was evident. Animals were imaged over the following week at stages 45 and 48, over which time the tectal neuropil nearly doubled in length and exhibited more precise retinotopic organization. By microinjecting GCaMP6s messenger ribonucleic acid (mRNA) into one blastomere of two-cell stage embryos, we acquired bilateral mosaic tadpoles with GCaMP6s expression in postsynaptic tectal neurons on one side of the animal and in retinal ganglion cell axons crossing to the tectum on the opposite side. Longitudinal observation of retinotopic map emergence revealed the presence of orderly representations of azimuth and elevation as early as stage 42, although presynaptic inputs exhibited relatively less topographic organization than the postsynaptic component for the azimuth axis. Retinotopic gradients in the tectum became smoother between stages 42 and 45. Blocking N-methyl-D-aspartate (NMDA) receptor conductance by rearing tadpoles in MK-801 did not prevent the emergence of retinotopic maps, but it produced more discontinuous topographic gradients and altered receptive field characteristics. These results provide evidence that current through NMDA receptors is dispensable for coarse topographic ordering of retinotectal inputs but does contribute to the fine-scale organization of the retinotectal projection.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Retina/diagnóstico por imagen , Retina/embriología , Animales , Axones/metabolismo , Mapeo Encefálico/métodos , Calcio/metabolismo , Larva/metabolismo , Células Ganglionares de la Retina/fisiología , Colículos Superiores/diagnóstico por imagen , Colículos Superiores/metabolismo , Vías Visuales/crecimiento & desarrollo , Xenopus laevis/embriología
11.
Dev Biol ; 483: 34-38, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34942195

RESUMEN

Proper function of the body is maintained by an intricate interaction and communication among cells. during the animal development how these cells are formed and maintained is an important yet elusive. Understanding of how cells such as muscle and nerve cells maintain their identities would enable us to control the diseases which include malfunctioning in cellular identities such as cancer. In this article, we describe how the concept of formation and maintenance of cell identities has changed over the last 100 years. We will also briefly describe our current experimental work which includes transcriptional dynamics, and protein-protein interaction and how they are bringing new molecular insights. We also describe liquid-liquid phase separation as a potential new mechanism for the stability of gene expression in the non dvididng specialised cells of Xenopus oocytes.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oocitos/citología , Oocitos/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética , Animales , División Celular Asimétrica/genética , Diferenciación Celular/genética , Femenino , Células Musculares/metabolismo , Neuronas/metabolismo , Ovoviviparidad/genética , Mapas de Interacción de Proteínas/genética , Transcripción Genética/genética , Xenopus laevis/metabolismo
12.
Dev Cell ; 57(1): 95-111.e12, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-34919801

RESUMEN

How embryos scale patterning according to size is still not fully understood. Through in silico screening and analysis of reaction-diffusion systems that could be responsible for scaling, we predicted the existence of genes whose expression is sensitive to embryo size and which regulate the scaling of embryonic patterning. To find these scalers, we identified genes with strongly altered expression in half-size Xenopus laevis embryos compared with full-size siblings at the gastrula stage. Among found genes, we investigated the role of matrix metalloproteinase-3 (mmp3), which was most strongly downregulated in half-size embryos. We show that Mmp3 scales dorsal-ventral patterning by degrading the slowly diffusing embryonic inducers Noggin1 and Noggin2 but preventing cleavage of the more rapidly diffusing inducer Chordin via degradation of a Tolloid-type proteinase. In addition to unraveling the mechanism underlying the scaling of dorsal-ventral patterning, this work provides proof of principal for scalers identification in embryos of other species.


Asunto(s)
Tipificación del Cuerpo/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Organizadores Embrionarios/metabolismo , Animales , Tipificación del Cuerpo/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Tamaño de la Célula , Embrión no Mamífero/metabolismo , Gástrula/metabolismo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metaloproteinasa 3 de la Matriz/fisiología , Transducción de Señal/fisiología , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología
13.
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
14.
Dev Biol ; 482: 101-113, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34906546

RESUMEN

During metazoan early embryogenesis, the intracellular properties of proteins and organelles change dynamically through rapid cleavage. In particular, a change in the nucleus size is known to contribute to embryonic development-dependent cell cycle and gene expression regulation. Here, we compared the nuclear sizes of various blastomeres from developing Xenopus embryos and analyzed the mechanisms that control the nuclear expansion dynamics by manipulating the amount of intracellular components in a cell-free system. Nuclear expansion was slower in blastomeres from vegetal hemispheres during a longer interphase than in those from animal hemispheres. Furthermore, upon recapitulating interphase events by manipulating the concentration of yolk platelets, which are originally rich in the vegetal blastomeres, in cell-free cytoplasmic extracts, nuclear expansion and DNA replication became slower than that in normal yolk-free conditions. Under these conditions, the supplemented yolk platelets accumulated around the nucleus in a microtubule-dependent manner and impeded the organization of the endoplasmic reticulum network. Overall, we propose that yolk platelets around the nucleus reduce membrane supply from the endoplasmic reticulum to the nucleus, resulting in slower nuclear expansion and cell cycle progression in the yolk-rich vegetal blastomeres.


Asunto(s)
Blastómeros/fisiología , Membrana Celular/fisiología , Núcleo Celular/fisiología , Retículo Endoplásmico/fisiología , Xenopus laevis/embriología , Animales , Tamaño de la Célula , Embrión no Mamífero/citología , Desarrollo Embrionario/fisiología , Interfase/fisiología
15.
Dev Biol ; 482: 1-6, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34818531

RESUMEN

Wnt signalling plays an eminent role in development, stem cell growth, and tissue homeostasis. Much of what we know about Wnt signalling, we owe to research in developmental biology. Here I review some salient discoveries in the older literature, beginning with the Lithium experiments in sea urchin by Curt Herbst in the 1890ies, when unknown to him he observed the gradual effects of Wnt overactivation upon embryonic axis formation. After revisiting key discoveries into Wingless signalling in Drosophila, I examine the role that the Xenopus embryo has played as model system in this regard. Not only were components of the Wnt cascade dissected and secreted Wnt antagonists discovered in Xenopus, but it also played a key role in unveiling the evolutionary conserved role of Wnt signalling in primary body axis formation. I conclude that Xenopus developmental biology has played a major role in elucidating the mechanisms of embryonic Wnt signalling.


Asunto(s)
Tipificación del Cuerpo/fisiología , Drosophila/embriología , Erizos de Mar/embriología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Xenopus laevis/embriología , Animales , Drosophila/metabolismo , Desarrollo Embrionario/fisiología , Erizos de Mar/metabolismo , Xenopus laevis/metabolismo
16.
Nature ; 600(7890): 690-694, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34880503

RESUMEN

Collective cell migration underlies morphogenesis, wound healing and cancer invasion1,2. Most directed migration in vivo has been attributed to chemotaxis, whereby cells follow a chemical gradient3-5. Cells can also follow a stiffness gradient in vitro, a process called durotaxis3,4,6-8, but evidence for durotaxis in vivo is lacking6. Here we show that in Xenopus laevis the neural crest-an embryonic cell population-self-generates a stiffness gradient in the adjacent placodal tissue, and follows this gradient by durotaxis. The gradient moves with the neural crest, which is continually pursuing a retreating region of high substrate stiffness. Mechanistically, the neural crest induces the gradient due to N-cadherin interactions with the placodes and senses the gradient through cell-matrix adhesions, resulting in polarized Rac activity and actomyosin contractility, which coordinates durotaxis. Durotaxis synergizes with chemotaxis, cooperatively polarizing actomyosin machinery of the cell group to prompt efficient directional collective cell migration in vivo. These results show that durotaxis and dynamic stiffness gradients exist in vivo, and gradients of chemical and mechanical signals cooperate to achieve efficient directional cell migration.


Asunto(s)
Movimiento Celular , Cresta Neural/citología , Docilidad , Actomiosina/metabolismo , Animales , Polaridad Celular , Quimiotaxis , Femenino , Dureza , Xenopus laevis/embriología , Proteínas de Unión al GTP rac/metabolismo
17.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34845026

RESUMEN

All living systems perpetuate themselves via growth in or on the body, followed by splitting, budding, or birth. We find that synthetic multicellular assemblies can also replicate kinematically by moving and compressing dissociated cells in their environment into functional self-copies. This form of perpetuation, previously unseen in any organism, arises spontaneously over days rather than evolving over millennia. We also show how artificial intelligence methods can design assemblies that postpone loss of replicative ability and perform useful work as a side effect of replication. This suggests other unique and useful phenotypes can be rapidly reached from wild-type organisms without selection or genetic engineering, thereby broadening our understanding of the conditions under which replication arises, phenotypic plasticity, and how useful replicative machines may be realized.


Asunto(s)
Fenómenos Biomecánicos/fisiología , Reproducción Asexuada/fisiología , Reproducción/fisiología , Adaptación Fisiológica/fisiología , Animales , Inteligencia Artificial , Ingeniería Genética/métodos , Regeneración Tisular Dirigida/métodos , Fenotipo , Agregado de Proteínas/fisiología , Biología Sintética/métodos , Xenopus laevis/embriología , Xenopus laevis/metabolismo
18.
Mol Cells ; 44(10): 723-735, 2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34711690

RESUMEN

Spemann organizer is a center of dorsal mesoderm and itself retains the mesoderm character, but it has a stimulatory role for neighboring ectoderm cells in becoming neuroectoderm in gastrula embryos. Goosecoid (Gsc) overexpression in ventral region promotes secondary axis formation including neural tissues, but the role of gsc in neural specification could be indirect. We examined the neural inhibitory and stimulatory roles of gsc in the same cell and neighboring cells contexts. In the animal cap explant system, Gsc overexpression inhibited expression of neural specific genes including foxd4l1.1, zic3, ncam, and neurod. Genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) and promoter analysis of early neural genes of foxd4l1.1 and zic3 were performed to show that the neural inhibitory mode of gsc was direct. Site-directed mutagenesis and serially deleted construct studies of foxd4l1.1 promoter revealed that Gsc directly binds within the foxd4l1.1 promoter to repress its expression. Conjugation assay of animal cap explants was also performed to demonstrate an indirect neural stimulatory role for gsc. The genes for secretory molecules, Chordin and Noggin, were up-regulated in gsc injected cells with the neural fate only achieved in gsc uninjected neighboring cells. These experiments suggested that gsc regulates neuroectoderm formation negatively when expressed in the same cell and positively in neighboring cells via soluble factors. One is a direct suppressive circuit of neural genes in gsc expressing mesoderm cells and the other is an indirect stimulatory circuit for neurogenesis in neighboring ectoderm cells via secreted BMP antagonizers.


Asunto(s)
Proteína Goosecoide/metabolismo , Placa Neural/embriología , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales
19.
Cells ; 10(10)2021 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-34685759

RESUMEN

Inhibition of the bone morphogenetic proteins (BMPs) is the primary step toward neuroectoderm formation in vertebrates. In this process, the Spemann organizer of the dorsal mesoderm plays a decisive role by secreting several extracellular BMP inhibitors such as Chordin (Chrd). Chrd physically interacts with BMP proteins and inhibits BMP signaling, which triggers the expression of neural-specific transcription factors (TFs), including Foxd4l1.1. Thus, Chrd induces in a BMP-inhibited manner and promotes neuroectoderm formation. However, the regulatory feedback mechanism of Foxd4l1.1 on mesodermal genes expression during germ-layer specification has not been fully elucidated. In this study, we investigated the regulatory mechanism of Foxd4l1.1 on chrd (a mesodermal gene). We demonstrate that Foxd4l1.1 inhibits chrd expression during neuroectoderm formation in two ways: First, Foxd4l1.1 directly binds to FRE (Foxd4l1.1 response elements) within the chrd promoter region to inhibit transcription. Second, Foxd4l1.1 physically interacts with Smad2 and Smad3, and this interaction blocks Smad2 and Smad3 binding to activin response elements (AREs) within the chrd promoter. Site-directed mutagenesis of FRE within the chrd(-2250) promoter completely abolished repressor activity of the Foxd4l1.1. RT-PCR and reporter gene assay results indicate that Foxd4l1.1 strongly inhibits mesoderm- and ectoderm-specific marker genes to maintain neural fate. Altogether, these results suggest that Foxd4l1.1 negatively regulates chrd transcription by dual mechanism. Thus, our study demonstrates the existence of precise reciprocal regulation of chrd transcription during neuroectoderm and mesoderm germ-layer specification in Xenopus embryos.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Gástrula/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Transcripción Genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética , Activinas/metabolismo , Animales , Proteínas de Unión al ADN/química , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Genes Reporteros , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Dominios Proteicos , Elementos de Respuesta/genética , Proteínas Smad/metabolismo , Proteínas de Xenopus/química
20.
PLoS Biol ; 19(9): e3001377, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34491983

RESUMEN

Forming an embryo from a zygote poses an apparent conflict for epigenetic regulation. On the one hand, the de novo induction of cell fate identities requires the establishment and subsequent maintenance of epigenetic information to harness developmental gene expression. On the other hand, the embryo depends on cell proliferation, and every round of DNA replication dilutes preexisting histone modifications by incorporation of new unmodified histones into chromatin. Here, we investigated the possible relationship between the propagation of epigenetic information and the developmental cell proliferation during Xenopus embryogenesis. We systemically inhibited cell proliferation during the G1/S transition in gastrula embryos and followed their development until the tadpole stage. Comparing wild-type and cell cycle-arrested embryos, we show that the inhibition of cell proliferation is principally compatible with embryo survival and cellular differentiation. In parallel, we quantified by mass spectrometry the abundance of a large set of histone modification states, which reflects the developmental maturation of the embryonic epigenome. The arrested embryos developed abnormal stage-specific histone modification profiles (HMPs), in which transcriptionally repressive histone marks were overrepresented. Embryos released from the cell cycle block during neurulation reverted toward normality on morphological, molecular, and epigenetic levels. These results suggest that the cell cycle block by HUA alters stage-specific HMPs. We propose that this influence is strong enough to control developmental decisions, specifically in cell populations that switch between resting and proliferating states such as stem cells.


Asunto(s)
Epigénesis Genética , Código de Histonas , Xenopus laevis/embriología , Animales , Afidicolina/farmacología , Ciclo Celular , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero/embriología , Inhibidores Enzimáticos/farmacología , Hidroxiurea/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA