Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genesis ; 59(10): e23447, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34478234

RESUMO

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


Assuntos
Desenvolvimento Embrionário/genética , Proteína HMGN1/genética , Crista Neural/crescimento & desenvolvimento , Fatores de Transcrição SOXE/genética , Proteína 1 Relacionada a Twist/genética , Proteínas de Xenopus/genética , Animais , Cromatina/genética , Embrião não Mamífero , Fatores de Transcrição Forkhead/genética , Gástrula/crescimento & desenvolvimento , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas de Silenciamento de Genes , Proteína HMGN1/antagonistas & inibidores , Crista Neural/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição/genética , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento
2.
Genome Biol ; 22(1): 197, 2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34225769

RESUMO

BACKGROUND: Single-cell technologies are transforming biomedical research, including the recent demonstration that unspliced pre-mRNA present in single-cell RNA-Seq permits prediction of future expression states. Here we apply this RNA velocity concept to an extended timecourse dataset covering mouse gastrulation and early organogenesis. RESULTS: Intriguingly, RNA velocity correctly identifies epiblast cells as the starting point, but several trajectory predictions at later stages are inconsistent with both real-time ordering and existing knowledge. The most striking discrepancy concerns red blood cell maturation, with velocity-inferred trajectories opposing the true differentiation path. Investigating the underlying causes reveals a group of genes with a coordinated step-change in transcription, thus violating the assumptions behind current velocity analysis suites, which do not accommodate time-dependent changes in expression dynamics. Using scRNA-Seq analysis of chimeric mouse embryos lacking the major erythroid regulator Gata1, we show that genes with the step-changes in expression dynamics during erythroid differentiation fail to be upregulated in the mutant cells, thus underscoring the coordination of modulating transcription rate along a differentiation trajectory. In addition to the expected block in erythroid maturation, the Gata1-chimera dataset reveals induction of PU.1 and expansion of megakaryocyte progenitors. Finally, we show that erythropoiesis in human fetal liver is similarly characterized by a coordinated step-change in gene expression. CONCLUSIONS: By identifying a limitation of the current velocity framework coupled with in vivo analysis of mutant cells, we reveal a coordinated step-change in gene expression kinetics during erythropoiesis, with likely implications for many other differentiation processes.


Assuntos
Células Eritroides/metabolismo , Eritropoese/genética , Fator de Transcrição GATA1/genética , Regulação da Expressão Gênica no Desenvolvimento , Organogênese/genética , Animais , Diferenciação Celular , Conjuntos de Dados como Assunto , Embrião de Mamíferos , Células Eritroides/citologia , Feto , Fator de Transcrição GATA1/deficiência , Gástrula/crescimento & desenvolvimento , Gástrula/metabolismo , Humanos , Cinética , Fígado/citologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Análise de Célula Única , Transativadores/genética , Transativadores/metabolismo , Ativação Transcricional
3.
Nat Commun ; 12(1): 3277, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-34078907

RESUMO

Generating properly differentiated embryonic structures in vitro from pluripotent stem cells remains a challenge. Here we show that instruction of aggregates of mouse embryonic stem cells with an experimentally engineered morphogen signalling centre, that functions as an organizer, results in the development of embryo-like entities (embryoids). In situ hybridization, immunolabelling, cell tracking and transcriptomic analyses show that these embryoids form the three germ layers through a gastrulation process and that they exhibit a wide range of developmental structures, highly similar to neurula-stage mouse embryos. Embryoids are organized around an axial chordamesoderm, with a dorsal neural plate that displays histological properties similar to the murine embryo neuroepithelium and that folds into a neural tube patterned antero-posteriorly from the posterior midbrain to the tip of the tail. Lateral to the chordamesoderm, embryoids display somitic and intermediate mesoderm, with beating cardiac tissue anteriorly and formation of a vasculature network. Ventrally, embryoids differentiate a primitive gut tube, which is patterned both antero-posteriorly and dorso-ventrally. Altogether, embryoids provide an in vitro model of mammalian embryo that displays extensive development of germ layer derivatives and that promises to be a powerful tool for in vitro studies and disease modelling.


Assuntos
Padronização Corporal/genética , Corpos Embrioides/metabolismo , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias Murinas/metabolismo , Transdução de Sinais/genética , Animais , Ectoderma/citologia , Ectoderma/crescimento & desenvolvimento , Ectoderma/metabolismo , Embrião de Mamíferos , Corpos Embrioides/citologia , Endoderma/citologia , Endoderma/crescimento & desenvolvimento , Endoderma/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Gástrula/citologia , Gástrula/crescimento & desenvolvimento , Gástrula/metabolismo , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Tubo Neural/citologia , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/metabolismo , Notocorda/citologia , Notocorda/crescimento & desenvolvimento , Notocorda/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo
4.
Stem Cell Reports ; 16(5): 1143-1155, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33891872

RESUMO

When stimulated with a pulse from an exogenous WNT pathway activator, small aggregates of mouse embryonic stem cells (ESCs) can undergo embryo-like axial morphogenesis and patterning along the three major body axes. However, these structures, called gastruloids, currently lack the anterior embryonic regions, such as those belonging to the brain. Here, we describe an approach to generate gastruloids that have a more complete antero-posterior development. We used hydrogel microwell arrays to promote the robust derivation of mouse ESCs into post-implantation epiblast-like (EPI) aggregates in a reproducible and scalable manner. These EPI aggregates break symmetry and axially elongate without external chemical stimulation. Inhibition of WNT signaling in early stages of development leads to the formation of gastruloids with anterior neural tissues. Thus, we provide a new tool to study the development of the mouse after implantation in vitro, especially the formation of anterior neural regions.


Assuntos
Padronização Corporal , Gástrula/crescimento & desenvolvimento , Tecido Nervoso/crescimento & desenvolvimento , Organogênese , Proteínas Wnt/metabolismo , Animais , Padronização Corporal/efeitos dos fármacos , Agregação Celular/efeitos dos fármacos , Linhagem Celular , Gástrula/efeitos dos fármacos , Camadas Germinativas/citologia , Camadas Germinativas/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/farmacologia , Hidrogéis/farmacologia , Camundongos , Tecido Nervoso/efeitos dos fármacos , Organogênese/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
5.
Mech Dev ; 163: 103629, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32615151

RESUMO

Gastrulation is generally understood as the morphogenetic processes that result in the spatial organization of the blastomere into the three germ layers, ectoderm, mesoderm and endoderm. This review summarizes our current knowledge of the morphogenetic mechanisms in Drosophila gastrulation. In addition to the events that drive mesoderm invagination and germband elongation, we pay particular attention to other, less well-known mechanisms including midgut invagination, cephalic furrow formation, dorsal fold formation, and mesoderm layer formation. This review covers topics ranging from the identification and functional characterization of developmental and morphogenetic control genes to the analysis of the physical properties of cells and tissues and the control of cell and tissue mechanics of the morphogenetic movements in the gastrula.


Assuntos
Drosophila melanogaster/genética , Gástrula/crescimento & desenvolvimento , Gastrulação/genética , Morfogênese/genética , Animais , Fenômenos Biomecânicos/genética , Drosophila melanogaster/crescimento & desenvolvimento , Ectoderma/crescimento & desenvolvimento , Embrião não Mamífero , Endoderma/crescimento & desenvolvimento , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Mesoderma/crescimento & desenvolvimento
6.
Mech Dev ; 163: 103625, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32526279

RESUMO

The Brachyury gene encodes a transcription factor that is conserved across all animals. In non-chordate metazoans, brachyury is primarily expressed in ectoderm regions that are added to the endodermal gut during development, and often form a ring around the site of endoderm internalization in the gastrula, the blastopore. In chordates, this brachyury ring is conserved, but the gene has taken on a new role in the formation of the mesoderm. In this phylum, a novel type of mesoderm that develops into notochord and somites has been added to the ancestral lateral plate mesoderm. Brachyury contributes to a shift in cell fate from neural ectoderm to posterior notochord and somites during a major lineage segregation event that in Xenopus and in the zebrafish takes place in the early gastrula. In the absence of this brachyury function, impaired formation of posterior mesoderm indirectly affects the gastrulation movements of peak involution and convergent extension. These movements are confined to specific regions and stages, leaving open the question why brachyury expression in an extensive, coherent ring, before, during and after gastrulation, is conserved in the two species whose gastrulation modes differ considerably, and also in many other metazoan gastrulae of diverse structure.


Assuntos
Ectoderma/crescimento & desenvolvimento , Proteínas Fetais/genética , Gástrula/crescimento & desenvolvimento , Morfogênese/genética , Proteínas com Domínio T/genética , Animais , Endoderma/crescimento & desenvolvimento , Proteínas Fetais/ultraestrutura , Mesoderma/crescimento & desenvolvimento , Notocorda/crescimento & desenvolvimento , Proteínas com Domínio T/ultraestrutura , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
7.
Proc Natl Acad Sci U S A ; 117(27): 15712-15723, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32561646

RESUMO

The mechanisms used by embryos to pattern tissues across their axes has fascinated developmental biologists since the founding of embryology. Here, using single-cell technology, we interrogate complex patterning defects and define a Hedgehog (Hh)-fibroblast growth factor (FGF) signaling axis required for anterior mesoderm lineage development during gastrulation. Single-cell transcriptome analysis of Hh-deficient mesoderm revealed selective deficits in anterior mesoderm populations, culminating in defects to anterior embryonic structures, including the pharyngeal arches, heart, and anterior somites. Transcriptional profiling of Hh-deficient mesoderm during gastrulation revealed disruptions to both transcriptional patterning of the mesoderm and FGF signaling for mesoderm migration. Mesoderm-specific Fgf4/Fgf8 double-mutants recapitulated anterior mesoderm defects and Hh-dependent GLI transcription factors modulated enhancers at FGF gene loci. Cellular migration defects during gastrulation induced by Hh pathway antagonism were mitigated by the addition of FGF4 protein. These findings implicate a multicomponent signaling hierarchy activated by Hh ligands from the embryonic node and executed by FGF signals in nascent mesoderm to control anterior mesoderm patterning.


Assuntos
Fator 4 de Crescimento de Fibroblastos/genética , Fator 8 de Crescimento de Fibroblasto/genética , Gastrulação/genética , Proteína GLI1 em Dedos de Zinco/genética , Animais , Padronização Corporal/genética , Linhagem da Célula/genética , Embrião de Galinha , Fatores de Crescimento de Fibroblastos/genética , Gástrula/crescimento & desenvolvimento , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Hedgehog/genética , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Transdução de Sinais/genética , Análise de Célula Única , Transcriptoma/genética
8.
Mech Dev ; 163: 103624, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32562871

RESUMO

Gastrulation consists in the dramatic reorganisation of the epiblast, a one-cell thick epithelial sheet, into a multilayered embryo. In chick, the formation of the internal layers requires the generation of a macroscopic convection-like flow, which involves up to 50,000 epithelial cells in the epiblast. These cell movements locate the mesendoderm precursors into the midline of the epiblast to form the primitive streak. There they acquire a mesenchymal phenotype, ingress into the embryo and migrate outward to populate the inner embryonic layers. This review covers what is currently understood about how cell behaviours ultimately cause these morphogenetic events and how they are regulated. We discuss 1) how the biochemical patterning of the embryo before gastrulation creates compartments of differential cell behaviours, 2) how the global epithelial flows arise from the coordinated actions of individual cells, 3) how the cells delaminate individually from the epiblast during the ingression, and 4) how cells move after the ingression following stereotypical migration routes. We conclude by exploring new technical advances that will facilitate future research in the chick model system.


Assuntos
Gástrula/embriologia , Gastrulação/genética , Camadas Germinativas/embriologia , Morfogênese/genética , Animais , Embrião de Galinha , Galinhas/crescimento & desenvolvimento , Gástrula/crescimento & desenvolvimento , Camadas Germinativas/crescimento & desenvolvimento , Mesoderma/embriologia
9.
Mech Dev ; 163: 103617, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32473204

RESUMO

During mouse embryonic development a mass of pluripotent epiblast tissue is transformed during gastrulation to generate the three definitive germ layers: endoderm, mesoderm, and ectoderm. During gastrulation, a spatiotemporally controlled sequence of events results in the generation of organ progenitors and positions them in a stereotypical fashion throughout the embryo. Key to the correct specification and differentiation of these cell fates is the establishment of an axial coordinate system along with the integration of multiple signals by individual epiblast cells to produce distinct outcomes. These signaling domains evolve as the anterior-posterior axis is established and the embryo grows in size. Gastrulation is initiated at the posteriorly positioned primitive streak, from which nascent mesoderm and endoderm progenitors ingress and begin to diversify. Advances in technology have facilitated the elaboration of landmark findings that originally described the epiblast fate map and signaling pathways required to execute those fates. Here we will discuss the current state of the field and reflect on how our understanding has shifted in recent years.


Assuntos
Padronização Corporal/genética , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética , Gastrulação/genética , Especificidade de Órgãos/genética , Animais , Linhagem da Célula/genética , Ectoderma/crescimento & desenvolvimento , Endoderma/crescimento & desenvolvimento , Feminino , Gástrula/crescimento & desenvolvimento , Gastrulação/fisiologia , Camadas Germinativas/crescimento & desenvolvimento , Mesoderma/crescimento & desenvolvimento , Camundongos , Gravidez
10.
Proc Natl Acad Sci U S A ; 117(21): 11444-11449, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32381735

RESUMO

Morphogenetic flows in developmental biology are characterized by the coordinated motion of thousands of cells that organize into tissues, naturally raising the question of how this collective organization arises. Using only the kinematics of tissue deformation, which naturally integrates local and global mechanisms along cell paths, we identify the dynamic morphoskeletons behind morphogenesis, i.e., the evolving centerpieces of multicellular trajectory patterns. These features are model- and parameter-free, frame-invariant, and robust to measurement errors and can be computed from unfiltered cell-velocity data. We reveal the spatial attractors and repellers of the embryo by quantifying its Lagrangian deformation, information that is inaccessible to simple trajectory inspection or Eulerian methods that are local and typically frame-dependent. Computing these dynamic morphoskeletons in wild-type and mutant chick and fly embryos, we find that they capture the early footprint of known morphogenetic features, reveal new ones, and quantitatively distinguish between different phenotypes.


Assuntos
Embrião de Galinha/citologia , Embrião de Galinha/crescimento & desenvolvimento , Drosophila melanogaster/embriologia , Modelos Biológicos , Animais , Animais Geneticamente Modificados , Fenômenos Biomecânicos , Embrião de Galinha/efeitos dos fármacos , Simulação por Computador , Proteínas de Drosophila/genética , Embrião não Mamífero/citologia , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/metabolismo , Gástrula/crescimento & desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Indazóis/farmacologia , Microscopia/métodos , Morfogênese , Mutação , Proteína 1 Relacionada a Twist/genética
11.
Sci Rep ; 9(1): 12597, 2019 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-31467377

RESUMO

Deciphering the key mechanisms of morphogenesis during embryonic development is crucial to understanding the guiding principles of the body plan and promote applications in biomedical research fields. Although several computational tissue reconstruction methods using cellular gene expression data have been proposed, those methods are insufficient with regard to arranging cells in their correct positions in tissues or organs unless spatial information is explicitly provided. Here, we report SPRESSO, a new in silico three-dimensional (3D) tissue reconstruction method using stochastic self-organizing map (stochastic-SOM) clustering, to estimate the spatial domains of cells in tissues or organs from only their gene expression profiles. With only five gene sets defined by Gene Ontology (GO), we successfully demonstrated the reconstruction of a four-domain structure of mid-gastrula mouse embryo (E7.0) with high reproducibility (success rate = 99%). Interestingly, the five GOs contain 20 genes, most of which are related to differentiation and morphogenesis, such as activin A receptor and Wnt family member genes. Further analysis indicated that Id2 is the most influential gene contributing to the reconstruction. SPRESSO may provide novel and better insights on the mechanisms of 3D structure formation of living tissues via informative genes playing a role as spatial discriminators.


Assuntos
Simulação por Computador , Gástrula/crescimento & desenvolvimento , Morfogênese , Animais , Sequência de Bases , Gástrula/metabolismo , Perfilação da Expressão Gênica , Ontologia Genética , Camundongos , Modelos Biológicos , Processos Estocásticos
12.
Methods Cell Biol ; 151: 433-442, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30948024

RESUMO

During sea urchin embryogenesis, primary mesenchyme cells (PMCs) follow a stereotypical migratory program, arrange into a primary pattern, then begin to secrete a bilaterally symmetric calcium carbonate skeleton. Recently identified genes are expressed in spatially-restricted domains within the PMC population (Sun & Ettensohn, 2014). To better understand the molecular mechanisms orchestrating PMC positioning, we are characterizing the expression profiles of PMC subset-specific genes. To deconvolve the spatiotemporal expression patterns within PMCs, we detect cell-specific mRNA expression with combined RNA fluorescence in situ hybridization and immunolabeling of PMCs. Subsequent confocal microscopy provides 3D position and expression information for individual PMCs. We extract PMC positions and relative gene expression levels, then model these results using open-source 3D modeling software. This versatile protocol can be extended to other models and systems.


Assuntos
Hibridização in Situ Fluorescente/métodos , Mesoderma/crescimento & desenvolvimento , Microscopia de Fluorescência/métodos , Ouriços-do-Mar/genética , Animais , Desenvolvimento Embrionário/genética , Gástrula/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Mesenquimais/citologia , Ouriços-do-Mar/crescimento & desenvolvimento
13.
Cold Spring Harb Protoc ; 2019(1)2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29769394

RESUMO

The fates of individual cleavage-stage blastomeres and of groups of cells at the blastula through gastrula stages of Xenopus embryos have been mapped in great detail. These studies identified the major contributors of the three germ layers as well as a variety of tissues and organs and several specific cell types. One can use these fate maps to test the commitment of single cells or groups of cells to produce their normal repertoire of descendants, to identify the genes that regulate fate commitment, and to modulate the levels of gene expression in specific lineages to determine gene function in a variety of developmental processes. Here we introduce methods that include how to identify specific blastomeres, inject them with lineage tracers, and alter gene expression levels. We also discuss methods for assaying protein and mRNA expression in situ and for providing novel embryonic environments to test fate commitment. These techniques draw upon classical approaches that are quite easy to perform in the versatile Xenopus embryo.


Assuntos
Blástula/citologia , Diferenciação Celular , Gástrula/citologia , Xenopus/embriologia , Animais , Blástula/crescimento & desenvolvimento , Gástrula/crescimento & desenvolvimento , Coloração e Rotulagem/métodos
14.
Cold Spring Harb Protoc ; 2019(2)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29321278

RESUMO

In 1924, Hans Spemann and Hilde Mangold (née Pröscholdt) published their famous work describing the transplantation of dorsal blastopore lip of one newt gastrula embryo onto the ventral side of a host embryo at the same stage. They performed these grafts using two newt species with different pigmentation (Triturus taeniatus and Triturus cristatus) to follow the fate of the grafted tissue. These experiments resulted in the development of conjoined twins attached through their belly. Because of the difference in embryo pigmentation between the two Triturus species, they determined that the bulk of the secondary embryo arose from the host embryo while the grafted tissue per se gave increase to the notochord and a few somitic cells. This meant that the dorsal blastopore lip was able to organize an almost complete embryo out of ventral tissue. The dorsal blastopore lip is now called the Spemann-Mangold organizer. Here, we describe a simple yet efficient protocol to perform these grafts using the anuran Xenopus laevis.


Assuntos
Transplante de Tecido Fetal/métodos , Gástrula/crescimento & desenvolvimento , Organizadores Embrionários , Transplante Homólogo/métodos , Xenopus/embriologia , Animais
15.
Cold Spring Harb Protoc ; 2019(2)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29321288

RESUMO

Einsteck procedure refers to a method whereby the experimenter inserts material into the blastocoel cavity of an early amphibian embryo. This procedure is simpler to perform than other types of grafts, such as Spemann-Mangold, and with practice yields a sizable amount of data suitable for statistical analysis. This protocol for Einsteck transplantation in Xenopus describes the insertion of the gastrula-stage blastopore lip into the blastocoel cavity of a host embryo.


Assuntos
Transplante de Tecido Fetal/métodos , Gástrula/crescimento & desenvolvimento , Transplante Homólogo/métodos , Xenopus/embriologia , Animais
16.
Wiley Interdiscip Rev Dev Biol ; 7(6): e325, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29944210

RESUMO

Xenopus gastrulation movements are in large part based on the rearrangement of cells by differential cell-on-cell migration within multilayered tissues. Different patterns of migration-based cell intercalation drive endoderm and mesoderm internalization and their positioning along their prospective body axes. C-cadherin, fibronectin, integrins, and focal contact components are expressed in all gastrula cells and play putative roles in cell-on-cell migration, but their actual functions in this respect are not yet understood. The gastrula can be subdivided into two motility domains, and in the vegetal, migratory domain, two modes of cell migration are discerned. Vegetal endoderm cells show ingression-type migration, a variant of amoeboid migration characterized by the lack of locomotory protrusions and by macropinocytosis as a mechanism of trailing edge resorption. Mesendoderm and prechordal mesoderm cells use lamellipodia in a mesenchymal mode of migration. Gastrula cell motility can be dissected into traits, such as cell polarity, adhesion, mobility, or protrusive activity, which are controlled separately yet in complex, combinatorial ways. Cells can instantaneously switch between different combinations of traits, showing plasticity as they respond to substratum properties. This article is categorized under: Early Embryonic Development > Gastrulation and Neurulation.


Assuntos
Padronização Corporal/genética , Ectoderma/citologia , Endoderma/citologia , Gástrula/citologia , Mesoderma/citologia , Xenopus laevis/embriologia , Animais , Caderinas/genética , Caderinas/metabolismo , Movimento Celular , Polaridade Celular , Ectoderma/crescimento & desenvolvimento , Ectoderma/metabolismo , Embrião não Mamífero , Endoderma/crescimento & desenvolvimento , Endoderma/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Gástrula/crescimento & desenvolvimento , Gástrula/metabolismo , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Integrinas/genética , Integrinas/metabolismo , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Pseudópodes/genética , Pseudópodes/metabolismo , Pseudópodes/ultraestrutura , Transdução de Sinais , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
17.
Elife ; 72018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29533180

RESUMO

Indirect evidence suggests that blastopore closure during gastrulation of anamniotes, including amphibians such as Xenopus laevis, depends on circumblastoporal convergence forces generated by the marginal zone (MZ), but direct evidence is lacking. We show that explanted MZs generate tensile convergence forces up to 1.5 µN during gastrulation and over 4 µN thereafter. These forces are generated by convergent thickening (CT) until the midgastrula and increasingly by convergent extension (CE) thereafter. Explants from ventralized embryos, which lack tissues expressing CE but close their blastopores, produce up to 2 µN of tensile force, showing that CT alone generates forces sufficient to close the blastopore. Uniaxial tensile stress relaxation assays show stiffening of mesodermal and ectodermal tissues around the onset of neurulation, potentially enhancing long-range transmission of convergence forces. These results illuminate the mechanobiology of early vertebrate morphogenic mechanisms, aid interpretation of phenotypes, and give insight into the evolution of blastopore closure mechanisms.


Assuntos
Evolução Biológica , Gástrula/crescimento & desenvolvimento , Morfogênese/fisiologia , Xenopus/crescimento & desenvolvimento , Animais , Ectoderma/crescimento & desenvolvimento , Gastrulação/fisiologia , Mesoderma/crescimento & desenvolvimento , Resistência à Tração , Xenopus laevis/crescimento & desenvolvimento
18.
Nat Cell Biol ; 20(1): 36-45, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29203884

RESUMO

Epithelial folding is typically driven by localized actomyosin contractility. However, it remains unclear how epithelia deform when myosin levels are low and uniform. In the Drosophila gastrula, dorsal fold formation occurs despite a lack of localized myosin changes, while the fold-initiating cells reduce cell height following basal shifts of polarity via an unknown mechanism. We show that cell shortening depends on an apical microtubule network organized by the CAMSAP protein Patronin. Prior to gastrulation, microtubule forces generated by the minus-end motor dynein scaffold the apical cell cortex into a dome-like shape, while the severing enzyme Katanin facilitates network remodelling to ensure tissue-wide cell size homeostasis. During fold initiation, Patronin redistributes following basal polarity shifts in the initiating cells, apparently weakening the scaffolding forces to allow dome descent. The homeostatic network that ensures size/shape homogeneity is thus repurposed for cell shortening, linking epithelial polarity to folding via a microtubule-based mechanical mechanism.


Assuntos
Proteínas de Drosophila/genética , Gástrula/metabolismo , Homeostase/genética , Katanina/genética , Mecanotransdução Celular , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Animais Geneticamente Modificados , Polaridade Celular , Tamanho Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Dineínas/genética , Dineínas/metabolismo , Embrião não Mamífero , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Gástrula/citologia , Gástrula/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Katanina/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Miosinas/genética , Miosinas/metabolismo
19.
Dev Cell ; 43(2): 198-211.e12, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-29033362

RESUMO

Cell-cell contact formation constitutes an essential step in evolution, leading to the differentiation of specialized cell types. However, remarkably little is known about whether and how the interplay between contact formation and fate specification affects development. Here, we identify a positive feedback loop between cell-cell contact duration, morphogen signaling, and mesendoderm cell-fate specification during zebrafish gastrulation. We show that long-lasting cell-cell contacts enhance the competence of prechordal plate (ppl) progenitor cells to respond to Nodal signaling, required for ppl cell-fate specification. We further show that Nodal signaling promotes ppl cell-cell contact duration, generating a positive feedback loop between ppl cell-cell contact duration and cell-fate specification. Finally, by combining mathematical modeling and experimentation, we show that this feedback determines whether anterior axial mesendoderm cells become ppl or, instead, turn into endoderm. Thus, the interdependent activities of cell-cell signaling and contact formation control fate diversification within the developing embryo.


Assuntos
Comunicação Celular , Linhagem da Célula , Retroalimentação Fisiológica , Gástrula/metabolismo , Morfogênese/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Padronização Corporal , Diferenciação Celular , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Gástrula/crescimento & desenvolvimento , Gastrulação/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Modelos Teóricos , Proteína Nodal/genética , Proteína Nodal/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
20.
Mech Dev ; 148: 3-10, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28684256

RESUMO

BACKGROUND: Gastrulation is a complex orchestration of movements by cells that are specified early in development. Until now, classical convergent extension was considered to be the main contributor to sea urchin archenteron extension, and the relative contributions of cell divisions were unknown. Active migration of cells along the axis of extension was also not considered as a major factor in invagination. RESULTS: Cell transplantations plus live imaging were used to examine endoderm cell morphogenesis during gastrulation at high-resolution in the optically clear sea urchin embryo. The invagination sequence was imaged throughout gastrulation. One of the eight macromeres was replaced by a fluorescently labeled macromere at the 32 cell stage. At gastrulation those patches of fluorescent endoderm cell progeny initially about 4 cells wide, released a column of cells about 2 cells wide early in gastrulation and then often this column narrowed to one cell wide by the end of archenteron lengthening. The primary movement of the column of cells was in the direction of elongation of the archenteron with the narrowing (convergence) occurring as one of the two cells moved ahead of its neighbor. As the column narrowed, the labeled endoderm cells generally remained as a contiguous population of cells, rarely separated by intrusion of a lateral unlabeled cell. This longitudinal cell migration mechanism was assessed quantitatively and accounted for almost 90% of the elongation process. Much of the extension was the contribution of Veg2 endoderm with a minor contribution late in gastrulation by Veg1 endoderm cells. We also analyzed the contribution of cell divisions to elongation. Endoderm cells in Lytechinus variagatus were determined to go through approximately one cell doubling during gastrulation. That doubling occurs without a net increase in cell mass, but the question remained as to whether oriented divisions might contribute to archenteron elongation. We learned that indeed there was a biased orientation of cell divisions along the plane of archenteron elongation, but when the impact of that bias was analyzed quantitatively, it contributed a maximum 15% to the total elongation of the gut. CONCLUSIONS: The major driver of archenteron elongation in the sea urchin, Lytechinus variagatus, is directed movement of Veg2 endoderm cells as a narrowing column along the plane of elongation. The narrowing occurs as cells in the column converge as they migrate, so that the combination of migration and the angular convergence provide the major component of the lengthening. A minor contributor to elongation is oriented cell divisions that contribute to the lengthening but no more than about 15%.


Assuntos
Gástrula/crescimento & desenvolvimento , Gastrulação/fisiologia , Morfogênese/fisiologia , Ouriços-do-Mar/embriologia , Animais , Movimento Celular/genética , Endoderma/crescimento & desenvolvimento , Endoderma/ultraestrutura , Gástrula/ultraestrutura , Ouriços-do-Mar/genética , Ouriços-do-Mar/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...