Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
Add more filters











Publication year range
1.
Elife ; 122024 May 10.
Article in English | MEDLINE | ID: mdl-38727576

ABSTRACT

Large-scale cell flow characterizes gastrulation in animal development. In amniote gastrulation, particularly in avian gastrula, a bilateral vortex-like counter-rotating cell flow, called 'polonaise movements', appears along the midline. Here, through experimental manipulations, we addressed relationships between the polonaise movements and morphogenesis of the primitive streak, the earliest midline structure in amniotes. Suppression of the Wnt/planar cell polarity (PCP) signaling pathway maintains the polonaise movements along a deformed primitive streak. Mitotic arrest leads to diminished extension and development of the primitive streak and maintains the early phase of the polonaise movements. Ectopically induced Vg1, an axis-inducing morphogen, generates the polonaise movements, aligned to the induced midline, but disturbs the stereotypical cell flow pattern at the authentic midline. Despite the altered cell flow, induction and extension of the primitive streak are preserved along both authentic and induced midlines. Finally, we show that ectopic axis-inducing morphogen, Vg1, is capable of initiating the polonaise movements without concomitant PS extension under mitotic arrest conditions. These results are consistent with a model wherein primitive streak morphogenesis is required for the maintenance of the polonaise movements, but the polonaise movements are not necessarily responsible for primitive streak morphogenesis. Our data describe a previously undefined relationship between the large-scale cell flow and midline morphogenesis in gastrulation.


Subject(s)
Gastrulation , Morphogenesis , Animals , Cell Movement , Primitive Streak/embryology , Cell Polarity , Gastrula/embryology , Chick Embryo
2.
Cell ; 187(11): 2855-2874.e19, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38657603

ABSTRACT

Progress in understanding early human development has been impeded by the scarcity of reference datasets from natural embryos, particularly those with spatial information during crucial stages like gastrulation. We conducted high-resolution spatial transcriptomics profiling on 38,562 spots from 62 transverse sections of an intact Carnegie stage (CS) 8 human embryo. From this spatial transcriptomic dataset, we constructed a 3D model of the CS8 embryo, in which a range of cell subtypes are identified, based on gene expression patterns and positional register, along the anterior-posterior, medial-lateral, and dorsal-ventral axis in the embryo. We further characterized the lineage trajectories of embryonic and extra-embryonic tissues and associated regulons and the regionalization of signaling centers and signaling activities that underpin lineage progression and tissue patterning during gastrulation. Collectively, the findings of this study provide insights into gastrulation and post-gastrulation development of the human embryo.


Subject(s)
Embryo, Mammalian , Gastrulation , Gene Expression Regulation, Developmental , Imaging, Three-Dimensional , Humans , Embryo, Mammalian/metabolism , Transcriptome/genetics , Gastrula/metabolism , Gastrula/embryology , Signal Transduction , Cell Lineage , Gene Expression Profiling , Body Patterning/genetics
3.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355799

ABSTRACT

The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.


Subject(s)
Animals, Newborn , Embryo, Mammalian , Embryonic Development , Gastrula , Single-Cell Analysis , Time-Lapse Imaging , Animals , Female , Mice , Pregnancy , Animals, Newborn/embryology , Animals, Newborn/genetics , Cell Differentiation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development/genetics , Gastrula/cytology , Gastrula/embryology , Gastrulation/genetics , Kidney/cytology , Kidney/embryology , Mesoderm/cytology , Mesoderm/enzymology , Neurons/cytology , Neurons/metabolism , Retina/cytology , Retina/embryology , Somites/cytology , Somites/embryology , Time Factors , Transcription Factors/genetics , Transcription, Genetic , Organ Specificity/genetics
4.
Nature ; 626(7998): 357-366, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38052228

ABSTRACT

Recently, several studies using cultures of human embryos together with single-cell RNA-seq analyses have revealed differences between humans and mice, necessitating the study of human embryos1-8. Despite the importance of human embryology, ethical and legal restrictions have limited post-implantation-stage studies. Thus, recent efforts have focused on developing in vitro self-organizing models using human stem cells9-17. Here, we report genetic and non-genetic approaches to generate authentic hypoblast cells (naive hPSC-derived hypoblast-like cells (nHyCs))-known to give rise to one of the two extraembryonic tissues essential for embryonic development-from naive human pluripotent stem cells (hPSCs). Our nHyCs spontaneously assemble with naive hPSCs to form a three-dimensional bilaminar structure (bilaminoids) with a pro-amniotic-like cavity. In the presence of additional naive hPSC-derived analogues of the second extraembryonic tissue, the trophectoderm, the efficiency of bilaminoid formation increases from 20% to 40%, and the epiblast within the bilaminoids continues to develop in response to trophectoderm-secreted IL-6. Furthermore, we show that bilaminoids robustly recapitulate the patterning of the anterior-posterior axis and the formation of cells reflecting the pregastrula stage, the emergence of which can be shaped by genetically manipulating the DKK1/OTX2 hypoblast-like domain. We have therefore successfully modelled and identified the mechanisms by which the two extraembryonic tissues efficiently guide the stage-specific growth and progression of the epiblast as it establishes the post-implantation landmarks of human embryogenesis.


Subject(s)
Embryonic Development , Germ Layers , Pluripotent Stem Cells , Humans , Cell Differentiation , Embryo Implantation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Embryonic Development/physiology , Germ Layers/cytology , Germ Layers/embryology , Germ Layers/metabolism , Pluripotent Stem Cells/cytology , Interleukin-6/metabolism , Gastrula/cytology , Gastrula/embryology , Amnion/cytology , Amnion/embryology , Amnion/metabolism , Ectoderm/cytology , Ectoderm/embryology , Ectoderm/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism
5.
Nature ; 625(7993): 126-133, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38123680

ABSTRACT

Chemical signalling is the primary means by which cells communicate in the embryo. The underlying principle refers to a group of ligand-producing cells and a group of cells that respond to this signal because they express the appropriate receptors1,2. In the zebrafish embryo, Wnt5b binds to the receptor Ror2 to trigger the Wnt-planar cell polarity (PCP) signalling pathway to regulate tissue polarity and cell migration3,4. However, it remains unclear how this lipophilic ligand is transported from the source cells through the aqueous extracellular space to the target tissue. In this study, we provide evidence that Wnt5b, together with Ror2, is loaded on long protrusions called cytonemes. Our data further suggest that the active Wnt5b-Ror2 complexes form in the producing cell and are handed over from these cytonemes to the receiving cell. Then, the receiving cell has the capacity to initiate Wnt-PCP signalling, irrespective of its functional Ror2 receptor status. On the tissue level, we further show that cytoneme-dependent spreading of active Wnt5b-Ror2 affects convergence and extension in the zebrafish gastrula. We suggest that cytoneme-mediated transfer of ligand-receptor complexes is a vital mechanism for paracrine signalling. This may prompt a reevaluation of the conventional concept of characterizing responsive and non-responsive tissues solely on the basis of the expression of receptors.


Subject(s)
Pseudopodia , Receptor Tyrosine Kinase-like Orphan Receptors , Wnt Proteins , Zebrafish , Animals , Gastrula/cytology , Gastrula/embryology , Gastrula/metabolism , Ligands , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Zebrafish/embryology , Zebrafish/metabolism , Cell Polarity , Cell Movement , Pseudopodia/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Paracrine Communication
6.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Article in English | MEDLINE | ID: mdl-35101917

ABSTRACT

In warm-blooded vertebrate embryos (mammals and birds), the axial tissues of the body form from a growth zone at the tail end, Hensen's node, which generates neural, mesodermal, and endodermal structures along the midline. While most cells only pass through this region, the node has been suggested to contain a small population of resident stem cells. However, it is unknown whether the rest of the node constitutes an instructive niche that specifies this self-renewal behavior. Here, we use heterotopic transplantation of groups and single cells and show that cells not destined to enter the node can become resident and self-renew. Long-term resident cells are restricted to the posterior part of the node and single-cell RNA-sequencing reveals that the majority of these resident cells preferentially express G2/M phase cell-cycle-related genes. These results provide strong evidence that the node functions as a niche to maintain self-renewal of axial progenitors.


Subject(s)
Body Patterning/physiology , Organizers, Embryonic/physiology , Stem Cell Niche/physiology , Animals , Chick Embryo , Endoderm/embryology , Gastrula/embryology , Mesoderm/embryology , Nervous System , Notochord/embryology , Organizers, Embryonic/metabolism , Stem Cell Niche/genetics , Stem Cells/metabolism , Stem Cells/physiology
7.
Biol Open ; 10(7)2021 07 15.
Article in English | MEDLINE | ID: mdl-34259326

ABSTRACT

Coordinated polarization of cells in the tissue plane, known as planar cell polarity (PCP), is associated with a signaling pathway critical for the control of morphogenetic processes. Although the segregation of PCP components to opposite cell borders is believed to play a critical role in this pathway, whether PCP derives from egg polarity or preexistent long-range gradient, or forms in response to a localized cue, remains a challenging question. Here we investigate the Xenopus neural plate, a tissue that has been previously shown to exhibit PCP. By imaging Vangl2 and Prickle3, we show that PCP is progressively acquired in the neural plate and requires a signal from the posterior region of the embryo. Tissue transplantations indicated that PCP is triggered in the neural plate by a planar cue from the dorsal blastopore lip. The PCP cue did not depend on the orientation of the graft and was distinct from neural inducers. These observations suggest that neuroectodermal PCP is not instructed by a preexisting molecular gradient but induced by a signal from the dorsal blastopore lip.


Subject(s)
Cell Polarity/physiology , Gastrula/embryology , Morphogenesis/physiology , Neural Plate/embryology , Xenopus/embryology , Animals , Signal Transduction
8.
Biochem Biophys Res Commun ; 569: 29-34, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34225077

ABSTRACT

Xenopus laevis is highly suitable as a toxicology animal model owing to its advantages in embryogenesis research. For toxicological studies, a large number of embryos must be handled simultaneously because they very rapidly develop into the target stages within a short period of time. To efficiently handle the embryos, a convenient embryo housing device is essential for fast and reliable assessment and statistical evaluation of malformation caused by toxicants. Here, we suggest 3D fabrication of single-egg trapping devices in which Xenopus eggs are fertilized in vitro, and the embryos are cultured. We used manual pipetting to insert the Xenopus eggs inside the trapping sites of the chip. By introducing a liquid circulating system, we connected a sperm-mixed solution with the chip to induce in vitro fertilization of the eggs. After the eggs were fertilized, we observed embryo development involving the formation of egg cleavage, blastula, gastrula, and tadpole. After the tadpoles grew inside the chip, we saved their lives by enabling their escape from the chip through reverse flow of the culture medium. The Xenopus chip can serve as an incubator to induce fertilization and monitor normal and abnormal development of the Xenopus from egg to tadpole.


Subject(s)
Embryo, Nonmammalian/embryology , Fertilization in Vitro/methods , Oocytes/cytology , Xenopus laevis/embryology , Animals , Blastula/cytology , Blastula/embryology , Blastula/physiology , Cell Division/physiology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Female , Fertilization in Vitro/instrumentation , Gastrula/cytology , Gastrula/embryology , Gastrula/physiology , Larva/cytology , Larva/growth & development , Larva/physiology , Locomotion/physiology , Male , Oocytes/physiology , Xenopus laevis/physiology
9.
Biochem Biophys Res Commun ; 559: 168-175, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33945994

ABSTRACT

Transforming growth factor (TGF)ß/activin superfamily regulates diverse biological processes including germ layer specification and axis patterning in vertebrates. TGFß/activin leads to phosphorylation of Smad2 and Smad3, followed by regulation of their target genes. Activin treatment also induces the essential organizer gene chordin (chrd). The involvement of Smad2/3 in chrd expression has been unclear as to whether Smad2/3 involvement is direct or indirect and whether any cis-acting response elements for Smad2/3 are present in the proximal or distal regions of its promoter. In the present study, we isolated the -2250 bps portion of the chrd promoter, showing that it contained Smad2/3 direct binding sites at its distal portion, separate from the proximal locations of other organizer genes, goosecoid and cerberus. The pattern of transcription activation for the promoter (-2250 bps) was indistinguishable from that of the endogenous chrd in gastrula Xenopus embryos. Reporter gene assays and site-directed mutagenesis analysis of the chrd promoter mapped two active activin/Smad response elements (ARE1 and ARE2) for Smad2 and Smad3. For a differential chrd induction, Smad2 acted on both ARE1 and ARE2, but Smad3 was only active for ARE2. Collectively, the results demonstrate that the distal region of chrd promoter contains the direct binding cis-acting elements for Smad2 and Smad3, which differentially modulate chrd transcription in gastrula Xenopus embryos.


Subject(s)
Gene Expression Regulation, Developmental , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Gastrula/embryology , Gastrula/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Smad2 Protein/genetics , Smad3 Protein/genetics , Transcriptional Activation , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
10.
Biol Open ; 10(2)2021 02 25.
Article in English | MEDLINE | ID: mdl-33563608

ABSTRACT

The blastula Chordin- and Noggin-expressing (BCNE) center comprises animal-dorsal and marginal-dorsal cells of the amphibian blastula and contains the precursors of the brain and the gastrula organizer. Previous findings suggested that the BCNE behaves as a homogeneous cell population that only depends on nuclear ß-catenin activity but does not require Nodal and later segregates into its descendants during gastrulation. In contrast to previous findings, in this work, we show that the BCNE does not behave as a homogeneous cell population in response to Nodal antagonists. In fact, we found that chordin.1 expression in a marginal subpopulation of notochordal precursors indeed requires Nodal input. We also establish that an animal BCNE subpopulation of cells that express both, chordin.1 and sox2 (a marker of pluripotent neuroectodermal cells), and gives rise to most of the brain, persisted at blastula stage after blocking Nodal. Therefore, Nodal signaling is required to define a population of chordin.1+ cells and to restrict the recruitment of brain precursors within the BCNE as early as at blastula stage. We discuss our findings in Xenopus in comparison to other vertebrate models, uncovering similitudes in early brain induction and delimitation through Nodal signaling.


Subject(s)
Blastula/metabolism , Brain/embryology , Brain/metabolism , Organizers, Embryonic/embryology , Organizers, Embryonic/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Biomarkers , Blastula/cytology , Embryonic Development/genetics , Gastrula/embryology , Gastrula/metabolism , Gene Expression Regulation, Developmental , Models, Biological , Organogenesis , Xenopus laevis
11.
Nature ; 584(7819): 102-108, 2020 08.
Article in English | MEDLINE | ID: mdl-32728215

ABSTRACT

During ontogeny, proliferating cells become restricted in their fate through the combined action of cell-type-specific transcription factors and ubiquitous epigenetic machinery, which recognizes universally available histone residues or nucleotides in a context-dependent manner1,2. The molecular functions of these regulators are generally well understood, but assigning direct developmental roles to them is hampered by complex mutant phenotypes that often emerge after gastrulation3,4. Single-cell RNA sequencing and analytical approaches have explored this highly conserved, dynamic period across numerous model organisms5-8, including mouse9-18. Here we advance these strategies using a combined zygotic perturbation and single-cell RNA-sequencing platform in which many mutant mouse embryos can be assayed simultaneously, recovering robust  morphological and transcriptional information across a panel of ten essential regulators. Deeper analysis of central Polycomb repressive complex (PRC) 1 and 2 components indicates substantial cooperativity, but distinguishes a dominant role for PRC2 in restricting the germline. Moreover, PRC mutant phenotypes emerge after gross epigenetic and transcriptional changes within the initial conceptus prior to gastrulation. Our experimental framework may eventually lead to a fully quantitative view of how cellular diversity emerges using an identical genetic template and from a single totipotent cell.


Subject(s)
Epigenesis, Genetic , Gastrula/embryology , Gastrula/metabolism , Gastrulation/genetics , Animals , Cell Lineage , Female , Gastrula/cytology , Gene Expression Regulation, Developmental , Male , Mice , Mutation , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Single-Cell Analysis , Transcription, Genetic
12.
Mech Dev ; 163: 103624, 2020 09.
Article in English | MEDLINE | ID: mdl-32562871

ABSTRACT

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.


Subject(s)
Gastrula/embryology , Gastrulation/genetics , Germ Layers/embryology , Morphogenesis/genetics , Animals , Chick Embryo , Chickens/growth & development , Gastrula/growth & development , Germ Layers/growth & development , Mesoderm/embryology
13.
Nature ; 582(7811): 253-258, 2020 06.
Article in English | MEDLINE | ID: mdl-32523119

ABSTRACT

Tissue sculpting during development has been attributed mainly to cellular events through processes such as convergent extension or apical constriction1,2. However, recent work has revealed roles for basement membrane remodelling in global tissue morphogenesis3-5. Upon implantation, the epiblast and extraembryonic ectoderm of the mouse embryo become enveloped by a basement membrane. Signalling between the basement membrane and these tissues is critical for cell polarization and the ensuing morphogenesis6,7. However, the mechanical role of the basement membrane in post-implantation embryogenesis remains unknown. Here we demonstrate the importance of spatiotemporally regulated basement membrane remodelling during early embryonic development. Specifically, we show that Nodal signalling directs the generation and dynamic distribution of perforations in the basement membrane by regulating the expression of matrix metalloproteinases. This basement membrane remodelling facilitates embryo growth before gastrulation. The establishment of the anterior-posterior axis8,9 further regulates basement membrane remodelling by localizing Nodal signalling-and therefore the activity of matrix metalloproteinases and basement membrane perforations-to the posterior side of the embryo. Perforations on the posterior side are essential for primitive-streak extension during gastrulation by rendering the basement membrane of the prospective primitive streak more prone to breaching. Thus spatiotemporally regulated basement membrane remodelling contributes to the coordination of embryo growth, morphogenesis and gastrulation.


Subject(s)
Basement Membrane/embryology , Basement Membrane/metabolism , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development , Animals , Basement Membrane/cytology , Blastocyst/cytology , Blastocyst/metabolism , Embryo, Mammalian/cytology , Extracellular Matrix/metabolism , Female , Gastrula/embryology , Male , Matrix Metalloproteinases/metabolism , Mice , Nodal Signaling Ligands/metabolism , Primitive Streak/cytology , Primitive Streak/embryology , Primitive Streak/metabolism
14.
Nature ; 582(7812): 410-415, 2020 06.
Article in English | MEDLINE | ID: mdl-32528178

ABSTRACT

The body plan of the mammalian embryo is shaped through the process of gastrulation, an early developmental event that transforms an isotropic group of cells into an ensemble of tissues that is ordered with reference to three orthogonal axes1. Although model organisms have provided much insight into this process, we know very little about gastrulation in humans, owing to the difficulty of obtaining embryos at such early stages of development and the ethical and technical restrictions that limit the feasibility of observing gastrulation ex vivo2. Here we show that human embryonic stem cells can be used to generate gastruloids-three-dimensional multicellular aggregates that differentiate to form derivatives of the three germ layers organized spatiotemporally, without additional extra-embryonic tissues. Human gastruloids undergo elongation along an anteroposterior axis, and we use spatial transcriptomics to show that they exhibit patterned gene expression. This includes a signature of somitogenesis that suggests that 72-h human gastruloids show some features of Carnegie-stage-9 embryos3. Our study represents an experimentally tractable model system to reveal and examine human-specific regulatory processes that occur during axial organization in early development.


Subject(s)
Body Patterning , Gastrula/cytology , Human Embryonic Stem Cells/cytology , Organoids/cytology , Organoids/embryology , Somites/cytology , Somites/embryology , Body Patterning/genetics , Gastrula/embryology , Gastrula/metabolism , Gene Expression Regulation, Developmental , Humans , In Vitro Techniques , Organoids/metabolism , Signal Transduction , Somites/metabolism , Transcriptome
15.
Zygote ; 28(3): 196-202, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32083523

ABSTRACT

Marine angelfish (family: Pomacanthidae) are among the most sought-after fish species in the saltwater aquarium trade. However, there is a lack of information in the literature on their early ontogeny. The objective of this study was to describe the embryonic and early larval development of two dwarf angelfish, the bicolour angelfish, Centropyge bicolor and the coral beauty angelfish, Centropyge bispinosa. The eggs of these two species were obtained from spontaneous spawning of the broodstock fish in captivity and incubated at 26.0 ± 0.2°C throughout the study. Fertilized eggs (n = 15) of both species are transparent, pelagic and spherical; the mean diameters of the eggs were measured at 703.6 ± 7.8 µm for C. bicolor and 627.6 ± 7.8 µm for C. bispinosa. The eggs of both species possessed a narrow perivitelline space, smooth and thin chorion, a homogenous and non-segmented yolk as well as a single oil globule. Overall, the observed embryonic development pattern of C. bicolor and C. bispinosa was very similar, and the main difference was the embryonic pigmentation pattern, which only became evident close to hatching. Larvae of both species started hatching at 13 h 30 min after fertilization, and the larval characteristics of both species also showed high levels of similarities. However, the mouth opening time for C. bicolor was 72 h after hatching (AH) and 96 AH for C. bispinosa. In general, the observed early ontogeny of C. bicolor and C. bispinosa also resembled that of other Centropyge species documented in the literature.


Subject(s)
Embryo, Nonmammalian/embryology , Embryonic Development/physiology , Ovum/growth & development , Perciformes/growth & development , Zygote/growth & development , Animals , Blastula/cytology , Blastula/embryology , Embryo, Nonmammalian/cytology , Female , Gastrula/cytology , Gastrula/embryology , Larva/growth & development , Ovum/cytology , Perciformes/classification , Perciformes/embryology , Pigmentation/physiology , Somites/cytology , Somites/embryology , Species Specificity , Time Factors , Zygote/cytology
16.
Nature ; 582(7812): 405-409, 2020 06.
Article in English | MEDLINE | ID: mdl-32076263

ABSTRACT

Gastruloids are three-dimensional aggregates of embryonic stem cells that display key features of mammalian development after implantation, including germ-layer specification and axial organization1-3. To date, the expression pattern of only a small number of genes in gastruloids has been explored with microscopy, and the extent to which genome-wide expression patterns in gastruloids mimic those in embryos is unclear. Here we compare mouse gastruloids with mouse embryos using single-cell RNA sequencing and spatial transcriptomics. We identify various embryonic cell types that were not previously known to be present in gastruloids, and show that key regulators of somitogenesis are expressed similarly between embryos and gastruloids. Using live imaging, we show that the somitogenesis clock is active in gastruloids and has dynamics that resemble those in vivo. Because gastruloids can be grown in large quantities, we performed a small screen that revealed how reduced FGF signalling induces a short-tail phenotype in embryos. Finally, we demonstrate that embedding in Matrigel induces gastruloids to generate somites with the correct rostral-caudal patterning, which appear sequentially in an anterior-to-posterior direction over time. This study thus shows the power of gastruloids as a model system for exploring development and somitogenesis in vitro in a high-throughput manner.


Subject(s)
Gastrula , Mouse Embryonic Stem Cells/cytology , Organoids/cytology , Organoids/embryology , Single-Cell Analysis , Somites/cytology , Somites/embryology , Transcriptome , Animals , Collagen , Drug Combinations , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development , Female , Gastrula/cytology , Gastrula/embryology , Gastrula/metabolism , Gene Expression Regulation, Developmental , Laminin , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Organoids/metabolism , Proteoglycans , RNA-Seq , Somites/metabolism , Time Factors
17.
Dev Biol ; 460(2): 176-186, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31904373

ABSTRACT

In Cnidaria, modes of gastrulation to produce the two body layers vary greatly between species. In the hydrozoan species Clytia hemisphaerica gastrulation involves unipolar ingression of presumptive endoderm cells from an oral domain of the blastula, followed by migration of these cells to fill the blastocoel with concomitant narrowing of the gastrula and elongation along the oral-aboral axis. We developed a 2D computational boundary model capable of simulating the morphogenetic changes during embryonic development from early blastula stage to the end of gastrulation. Cells are modeled as polygons with elastic membranes and cytoplasm, colliding and adhering to other cells, and capable of forming filopodia. With this model we could simulate compaction of the embryo preceding gastrulation, bottle cell formation, ingression, and intercalation between cells of the ingressing presumptive endoderm. We show that embryo elongation is dependent on the number of endodermal cells, low endodermal cell-cell adhesion, and planar cell polarity (PCP). When the strength of PCP is reduced in our model, resultant embryo morphologies closely resemble those reported previously following morpholino-mediated knockdown of the core PCP proteins Strabismus and Frizzled. Based on our results, we postulate that cellular processes of apical constriction, compaction, ingression, and then reduced cell-cell adhesion and mediolateral intercalation in the presumptive endoderm, are required and when combined, sufficient for Clytia gastrulation.


Subject(s)
Cnidaria/embryology , Gastrula/embryology , Gastrulation/physiology , Models, Biological , Animals , Cnidaria/cytology , Gastrula/cytology
18.
Cold Spring Harb Protoc ; 2020(3): 105551, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31857437

ABSTRACT

Over many years, the Xenopus laevis embryo has provided a powerful model system to investigate how mechanical forces regulate cellular function. Here, we describe a system to apply reproducible tensile and compressive force to X. laevis animal cap tissue explants and to simultaneously assess cellular behavior using live confocal imaging.


Subject(s)
Embryo, Nonmammalian/embryology , Gastrula/embryology , Stress, Mechanical , Xenopus laevis/embryology , Animals , Body Patterning , Cell Division , Elastic Modulus , Embryo, Nonmammalian/cytology , Embryonic Development , Gastrula/cytology , Microscopy, Confocal
19.
Methods Mol Biol ; 2047: 347-359, 2020.
Article in English | MEDLINE | ID: mdl-31552664

ABSTRACT

In the last decades, the cephalochordate amphioxus has reached a peculiar place in research laboratories as an excellent animal model to answer Evo/Devo questions. Nevertheless, mainly due to its restricted spawning season and to the small size of its embryos, only a few basic techniques in developmental biology could be used until recently. Fortunately, these last years, and thanks to the development of high-throughput techniques, new technical approaches have been possible, such as comparative transcriptomics and/or genomics. However, classic micromanipulation techniques are still difficult to apply. Here we present simple protocols for the manipulation of amphioxus embryos. First, we present the spawning induction method used with the European amphioxus species Branchiostoma lanceolatum. Second, we explain simple methods to manipulate the developing amphioxus embryo during the first steps of its development (before the hatching stage). These methods open many technical possibilities for future functional studies. Thus, we present here a simple technique to efficiently dechorionate a large number of embryos, we detail a protocol for the dissociation of cells during the first steps of the embryonic development and, finally, we describe micromanipulation approaches for tissue isolation during the gastrula stage.


Subject(s)
Embryo, Mammalian/physiology , Lancelets/embryology , Lancelets/physiology , Reproduction/physiology , Animals , Ectoderm/embryology , Ectoderm/physiology , Gastrula/embryology , Gastrula/physiology
20.
Nature ; 576(7787): 487-491, 2019 12.
Article in English | MEDLINE | ID: mdl-31827285

ABSTRACT

Formation of the three primary germ layers during gastrulation is an essential step in the establishment of the vertebrate body plan and is associated with major transcriptional changes1-5. Global epigenetic reprogramming accompanies these changes6-8, but the role of the epigenome in regulating early cell-fate choice remains unresolved, and the coordination between different molecular layers is unclear. Here we describe a single-cell multi-omics map of chromatin accessibility, DNA methylation and RNA expression during the onset of gastrulation in mouse embryos. The initial exit from pluripotency coincides with the establishment of a global repressive epigenetic landscape, followed by the emergence of lineage-specific epigenetic patterns during gastrulation. Notably, cells committed to mesoderm and endoderm undergo widespread coordinated epigenetic rearrangements at enhancer marks, driven by ten-eleven translocation (TET)-mediated demethylation and a concomitant increase of accessibility. By contrast, the methylation and accessibility landscape of ectodermal cells is already established in the early epiblast. Hence, regulatory elements associated with each germ layer are either epigenetically primed or remodelled before cell-fate decisions, providing the molecular framework for a hierarchical emergence of the primary germ layers.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gastrula/cytology , Gastrula/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Demethylation , Embryoid Bodies/cytology , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Enhancer Elements, Genetic/genetics , Epigenome/genetics , Erythropoiesis , Factor Analysis, Statistical , Gastrula/embryology , Gastrulation/physiology , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA/analysis , Time Factors , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL