Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
Cell Syst ; 15(5): 445-461.e4, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38692274

ABSTRACT

BMP signaling is essential for mammalian gastrulation, as it initiates a cascade of signals that control self-organized patterning. As development is highly dynamic, it is crucial to understand how time-dependent combinatorial signaling affects cellular differentiation. Here, we show that BMP signaling duration is a crucial control parameter that determines cell fates upon the exit from pluripotency through its interplay with the induced secondary signal WNT. BMP signaling directly converts cells from pluripotent to extraembryonic fates while simultaneously upregulating Wnt signaling, which promotes primitive streak and mesodermal specification. Using live-cell imaging of signaling and cell fate reporters together with a simple mathematical model, we show that this circuit produces a temporal morphogen effect where, once BMP signal duration is above a threshold for differentiation, intermediate and long pulses of BMP signaling produce specification of mesoderm and extraembryonic fates, respectively. Our results provide a systems-level picture of how these signaling pathways control the landscape of early human development.


Subject(s)
Bone Morphogenetic Proteins , Cell Differentiation , Primitive Streak , Signal Transduction , Primitive Streak/metabolism , Primitive Streak/embryology , Bone Morphogenetic Proteins/metabolism , Humans , Signal Transduction/physiology , Animals , Mesoderm/metabolism , Mesoderm/embryology , Wnt Signaling Pathway/physiology , Wnt Proteins/metabolism , Gene Expression Regulation, Developmental , Gastrulation/physiology
2.
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
3.
Nature ; 622(7983): 574-583, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37369348

ABSTRACT

Investigating human development is a substantial scientific challenge due to the technical and ethical limitations of working with embryonic samples. In the face of these difficulties, stem cells have provided an alternative to experimentally model inaccessible stages of human development in vitro1-13. Here we show that human pluripotent stem cells can be triggered to self-organize into three-dimensional structures that recapitulate some key spatiotemporal events of early human post-implantation embryonic development. Our system reproducibly captures spontaneous differentiation and co-development of embryonic epiblast-like and extra-embryonic hypoblast-like lineages, establishes key signalling hubs with secreted modulators and undergoes symmetry breaking-like events. Single-cell transcriptomics confirms differentiation into diverse cell states of the perigastrulating human embryo14,15 without establishing placental cell types, including signatures of post-implantation epiblast, amniotic ectoderm, primitive streak, mesoderm, early extra-embryonic endoderm, as well as initial yolk sac induction. Collectively, our system captures key features of human embryonic development spanning from Carnegie stage16 4-7, offering a reproducible, tractable and scalable experimental platform to understand the basic cellular and molecular mechanisms that underlie human development, including new opportunities to dissect congenital pathologies with high throughput.


Subject(s)
Cell Lineage , Embryo Implantation , Embryonic Development , Pluripotent Stem Cells , Female , Humans , Pregnancy , Cell Differentiation , Germ Layers/cytology , Germ Layers/enzymology , Human Embryonic Stem Cells/cytology , Placenta/cytology , Pluripotent Stem Cells/cytology , Primitive Streak/cytology , Primitive Streak/embryology , Yolk Sac/cytology , Yolk Sac/embryology
4.
Nature ; 612(7941): 732-738, 2022 12.
Article in English | MEDLINE | ID: mdl-36517595

ABSTRACT

Our understanding of human early development is severely hampered by limited access to embryonic tissues. Due to their close evolutionary relationship with humans, nonhuman primates are often used as surrogates to understand human development but currently suffer from a lack of in vivo datasets, especially from gastrulation to early organogenesis during which the major embryonic cell types are dynamically specified. To fill this gap, we collected six Carnegie stage 8-11 cynomolgus monkey (Macaca fascicularis) embryos and performed in-depth transcriptomic analyses of 56,636 single cells. Our analyses show transcriptomic features of major perigastrulation cell types, which help shed light on morphogenetic events including primitive streak development, somitogenesis, gut tube formation, neural tube patterning and neural crest differentiation in primates. In addition, comparative analyses with mouse embryos and human embryoids uncovered conserved and divergent features of perigastrulation development across species-for example, species-specific dependency on Hippo signalling during presomitic mesoderm differentiation-and provide an initial assessment of relevant stem cell models of human early organogenesis. This comprehensive single-cell transcriptome atlas not only fills the knowledge gap in the nonhuman primate research field but also serves as an invaluable resource for understanding human embryogenesis and developmental disorders.


Subject(s)
Gastrulation , Macaca fascicularis , Organogenesis , Single-Cell Analysis , Animals , Humans , Mice , Gastrulation/genetics , Macaca fascicularis/embryology , Macaca fascicularis/genetics , Organogenesis/genetics , Embryoid Bodies , Gene Expression Profiling , Primitive Streak/cytology , Primitive Streak/embryology , Neural Tube/cytology , Neural Tube/embryology , Neural Crest/cytology , Neural Crest/embryology , Hippo Signaling Pathway , Mesoderm/cytology , Mesoderm/embryology , Stem Cells
6.
Elife ; 102021 07 06.
Article in English | MEDLINE | ID: mdl-34227938

ABSTRACT

In classical descriptions of vertebrate development, the segregation of the three embryonic germ layers completes by the end of gastrulation. Body formation then proceeds in a head to tail fashion by progressive deposition of lineage-committed progenitors during regression of the primitive streak (PS) and tail bud (TB). The identification by retrospective clonal analysis of a population of neuromesodermal progenitors (NMPs) contributing to both musculoskeletal precursors (paraxial mesoderm) and spinal cord during axis formation challenged these notions. However, classical fate mapping studies of the PS region in amniotes have so far failed to provide direct evidence for such bipotential cells at the single-cell level. Here, using lineage tracing and single-cell RNA sequencing in the chicken embryo, we identify a resident cell population of the anterior PS epiblast, which contributes to neural and mesodermal lineages in trunk and tail. These cells initially behave as monopotent progenitors as classically described and only acquire a bipotential fate later, in more posterior regions. We show that NMPs exhibit a conserved transcriptomic signature during axis elongation but lose their epithelial characteristicsin the TB. Posterior to anterior gradients of convergence speed and ingression along the PS lead to asymmetric exhaustion of PS mesodermal precursor territories. Through limited ingression and increased proliferation, NMPs are maintained and amplified as a cell population which constitute the main progenitors in the TB. Together, our studies provide a novel understanding of the PS and TB contribution through the NMPs to the formation of the body of amniote embryos.


Subject(s)
Gene Expression Regulation, Developmental , Mesoderm/embryology , Neural Stem Cells/cytology , Primitive Streak/embryology , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Chick Embryo/embryology , Mesoderm/metabolism , Neural Stem Cells/physiology , Primitive Streak/metabolism
7.
PLoS Biol ; 19(5): e3001200, 2021 05.
Article in English | MEDLINE | ID: mdl-33999917

ABSTRACT

The heart develops from 2 sources of mesoderm progenitors, the first and second heart field (FHF and SHF). Using a single-cell transcriptomic assay combined with genetic lineage tracing and live imaging, we find the FHF and SHF are subdivided into distinct pools of progenitors in gastrulating mouse embryos at earlier stages than previously thought. Each subpopulation has a distinct origin in the primitive streak. The first progenitors to leave the primitive streak contribute to the left ventricle, shortly after right ventricle progenitor emigrate, followed by the outflow tract and atrial progenitors. Moreover, a subset of atrial progenitors are gradually incorporated in posterior locations of the FHF. Although cells allocated to the outflow tract and atrium leave the primitive streak at a similar stage, they arise from different regions. Outflow tract cells originate from distal locations in the primitive streak while atrial progenitors are positioned more proximally. Moreover, single-cell RNA sequencing demonstrates that the primitive streak cells contributing to the ventricles have a distinct molecular signature from those forming the outflow tract and atrium. We conclude that cardiac progenitors are prepatterned within the primitive streak and this prefigures their allocation to distinct anatomical structures of the heart. Together, our data provide a new molecular and spatial map of mammalian cardiac progenitors that will support future studies of heart development, function, and disease.


Subject(s)
Cell Lineage/genetics , Heart/embryology , Primitive Streak/embryology , Animals , Cell Lineage/physiology , Female , Gastrula , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Heart/physiology , Heart Atria/embryology , Heart Ventricles/embryology , Male , Mesoderm , Mice , Mice, Inbred C57BL , Morphogenesis , Primitive Streak/physiology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
8.
Indian J Pathol Microbiol ; 64(2): 390-393, 2021.
Article in English | MEDLINE | ID: mdl-33851643

ABSTRACT

Gonadal dysgenesis is a distinct variety of Disorders of Sexual Differentiation (DSD) characterised by incomplete or defective formation of the gonads due to either structural or numerical anomalies of the sex chromosomes or mutations in the genes involved in the development of the gland. Here we present two such rare cases that presented during childhood. Both patients presented with ambiguous genitalia with a 45XO/46XY mosaic chromosome pattern. First case, an infant underwent laparoscopic excision of streak gonad, and a single stage hypospadias repair later. Second case, an adolescent who underwent gonadectomy as a child, presented with a mass which was excised and found to contain uterine and ovarian tissue; second stage hypospadias repair is being planned. Mixed gonadal dysgenesis usually presents with a unilateral testis, a streak gonad on the contralateral side and persistent mullerian structures. The most common karyotype noted is 45XO/46XY. These cases are known to have ambiguous external genitalia. The streak gonads have an increased malignant potential and thus, these patients should be carefully screened and followed up for gonadoblastoma.


Subject(s)
Disorders of Sex Development/physiopathology , Gonadal Dysgenesis, Mixed/physiopathology , Sex Chromosome Aberrations , Sex Differentiation/physiology , Abnormal Karyotype , Adolescent , Chromosome Deletion , Cryptorchidism/genetics , Female , Humans , Hypospadias/physiopathology , Infant , Male , Mosaicism , Primitive Streak/embryology , Testis/abnormalities , Uterus/abnormalities
9.
Stem Cell Reports ; 16(5): 1210-1227, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33891870

ABSTRACT

Human embryonic stem cells cultured in 2D micropatterns with BMP4 differentiate into a radial arrangement of germ layers and extraembryonic cells. Single-cell transcriptomes demonstrate generation of cell types transcriptionally similar to their in vivo counterparts in Carnegie stage 7 human gastrula. Time-course analyses indicate sequential differentiation, where the epiblast arises by 12 h between the prospective ectoderm in the center and the cells initiating differentiation toward extraembryonic fates at the edge. Extraembryonic and mesendoderm precursors arise from the epiblast by 24 h, while nascent mesoderm, endoderm, and primordial germ cell-like cells form by 44 h. Dynamic changes in transcripts encoding signaling components support a BMP, WNT, and Nodal hierarchy underlying germ-layer specification conserved across mammals, and FGF and HIPPO pathways being active throughout differentiation. This work also provides a resource for mining genes and pathways expressed in a stereotyped 2D gastruloid model, common with other species or unique to human gastrulation.


Subject(s)
Cell Culture Techniques/methods , Cell Lineage/genetics , Gastrula/cytology , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Amnion/cytology , Cell Differentiation/genetics , Gastrulation , Germ Cells/cytology , Germ Layers/cytology , Humans , Mesoderm/cytology , Primitive Streak/embryology , Signal Transduction , Time Factors , Transcription, Genetic
11.
Development ; 148(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33199445

ABSTRACT

Anterior mesoderm (AM) and definitive endoderm (DE) progenitors represent the earliest embryonic cell types that are specified during germ layer formation at the primitive streak (PS) of the mouse embryo. Genetic experiments indicate that both lineages segregate from Eomes-expressing progenitors in response to different Nodal signaling levels. However, the precise spatiotemporal pattern of the emergence of these cell types and molecular details of lineage segregation remain unexplored. We combined genetic fate labeling and imaging approaches with single-cell RNA sequencing (scRNA-seq) to follow the transcriptional identities and define lineage trajectories of Eomes-dependent cell types. Accordingly, all cells moving through the PS during the first day of gastrulation express Eomes AM and DE specification occurs before cells leave the PS from Eomes-positive progenitors in a distinct spatiotemporal pattern. ScRNA-seq analysis further suggested the immediate and complete separation of AM and DE lineages from Eomes-expressing cells as last common bipotential progenitor.


Subject(s)
Cell Lineage , Endoderm/cytology , Endoderm/metabolism , Gastrulation , Mesoderm/cytology , Mesoderm/metabolism , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Germ Layers/cytology , Mice , Models, Biological , Primitive Streak/embryology , Primitive Streak/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Time Factors , Transcription, Genetic
12.
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
13.
Nature ; 577(7791): 537-542, 2020 01.
Article in English | MEDLINE | ID: mdl-31830756

ABSTRACT

Our understanding of how human embryos develop before gastrulation, including spatial self-organization and cell type ontogeny, remains limited by available two-dimensional technological platforms1,2 that do not recapitulate the in vivo conditions3-5. Here we report a three-dimensional (3D) blastocyst-culture system that enables human blastocyst development up to the primitive streak anlage stage. These 3D embryos mimic developmental landmarks and 3D architectures in vivo, including the embryonic disc, amnion, basement membrane, primary and primate unique secondary yolk sac, formation of anterior-posterior polarity and primitive streak anlage. Using single-cell transcriptome profiling, we delineate ontology and regulatory networks that underlie the segregation of epiblast, primitive endoderm and trophoblast. Compared with epiblasts, the amniotic epithelium shows unique and characteristic phenotypes. After implantation, specific pathways and transcription factors trigger the differentiation of cytotrophoblasts, extravillous cytotrophoblasts and syncytiotrophoblasts. Epiblasts undergo a transition to pluripotency upon implantation, and the transcriptome of these cells is maintained until the generation of the primitive streak anlage. These developmental processes are driven by different pluripotency factors. Together, findings from our 3D-culture approach help to determine the molecular and morphogenetic developmental landscape that occurs during human embryogenesis.


Subject(s)
Cell Culture Techniques/methods , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development , Primitive Streak/cytology , Primitive Streak/embryology , Amnion/cytology , Amnion/embryology , Blastocyst/cytology , Cell Differentiation , Cell Lineage , Cell Polarity , Collagen , Drug Combinations , Epithelium/embryology , Gastrulation , Germ Layers/cytology , Germ Layers/embryology , Humans , Laminin , Proteoglycans , RNA-Seq , Single-Cell Analysis , Transcription Factors/metabolism , Transcriptome , Trophoblasts/cytology , Yolk Sac/cytology , Yolk Sac/embryology
14.
Wiley Interdiscip Rev Dev Biol ; 9(2): e362, 2020 03.
Article in English | MEDLINE | ID: mdl-31622045

ABSTRACT

In Placentalia, the fetus depends upon an organized vascular connection with its mother for survival and development. Yet, this connection was, until recently, obscure. Here, we summarize how two unrelated tissues, the primitive streak, or body axis, and extraembryonic visceral endoderm collaborate to create and organize the fetal-placental arterial connection in the mouse gastrula. The primitive streak reaches into the extraembryonic space, where it marks the site of arterial union and creates a progenitor cell pool. Through contact with the streak, associated visceral endoderm undergoes an epithelial-to-mesenchymal transition, contributing extraembryonic mesoderm to the placental arterial vasculature, and to the allantois, or pre-umbilical tissue. In addition, visceral endoderm bifurcates into the allantois where, with the primitive streak, it organizes the nascent umbilical artery and promotes allantoic elongation to the chorion, the site of fetal-maternal exchange. Brachyury mediates streak extension and vascular patterning, while Hedgehog is involved in visceral endoderm's conversion to mesoderm. A unique CASPASE-3-positive cell separates streak- and non-streak-associated domains in visceral endoderm. Based on these new insights at the posterior embryonic-extraembryonic interface, we conclude by asking whether so-called primordial germ cells are truly antecedents to the germ line that segregate within the allantois, or whether they are placental progenitor cells. Incorporating these new working hypotheses into mutational analyses in which the placentae are affected will aid understanding a spectrum of disorders, including orphan diseases, which often include abnormalities of the umbilical cord, yolk sac, and hindgut, whose developmental relationship to each other has, until now, been poorly understood. This article is categorized under: Birth Defects > Associated with Preimplantation and Gastrulation Early Embryonic Development > Gastrulation and Neurulation.


Subject(s)
Arteries/embryology , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Endoderm/embryology , Fetus/embryology , Placenta/cytology , Primitive Streak/embryology , Animals , Female , Gastrula/cytology , Gastrula/physiology , Humans , Mice , Pregnancy
15.
PLoS One ; 14(7): e0219221, 2019.
Article in English | MEDLINE | ID: mdl-31260508

ABSTRACT

Cul4b-null (Cul4bΔ/Y) mice undergo growth arrest and degeneration during the early embryonic stages and die at E9.5. The pathogenic causes of this lethality remain incompletely characterized. However, it has been hypothesized that the loss of Cul4b function in extraembryonic tissues plays a key role. In this study, we investigated possible causes of death for Cul4b-null embryos, particularly in regard to the role of embryonic Cul4b. First, we show that the loss of embryonic Cul4b affects the growth of the inner cell mass in vitro and delays epiblast development during the gastrulation period at E6.5~E7.5 in vivo, as highlighted by the absence of the epiblastic transcription factor Brachyury from E6.5~E7.5. Additionally, at E7.5, strong and laterally expanded expression of Eomes and Fgf8 signaling was detected. Sectioning of these embryos showed disorganized primitive streak layer cells. Second, we observed that Mash2-expressing cells were present in the extraembryonic tissues of Cul4b-deficient embryos at E6.5 but were absent at E7.5. In addition, the loss of Cul4b resulted in decreased expression of cyclin proteins, which are required for the cell cycle transition from G1 to S. Taken together, these observations suggest that the embryonic expression of Cul4b is important for epiblast growth during E6.5~E7.5, and the loss of Cul4b results in either delayed growth of the epiblast or defective localization of primitive streak layer cells. As a result, the signaling activity mediated by the epiblast for subsequent ectoplacental cone development is affected, with the potential to induce growth retardation and lethality in Cul4bΔ/Y embryos.


Subject(s)
Cullin Proteins/physiology , Gastrulation/physiology , Germ Layers/embryology , Primitive Streak/embryology , Animals , Blastocyst Inner Cell Mass/metabolism , Embryo, Mammalian , Female , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Heterozygote , Male , Mice , Mice, Knockout , Models, Animal , T-Box Domain Proteins/metabolism
16.
Nat Cell Biol ; 21(7): 900-910, 2019 07.
Article in English | MEDLINE | ID: mdl-31263269

ABSTRACT

Breaking the anterior-posterior symmetry in mammals occurs at gastrulation. Much of the signalling network underlying this process has been elucidated in the mouse; however, there is no direct molecular evidence of events driving axis formation in humans. Here, we use human embryonic stem cells to generate an in vitro three-dimensional model of a human epiblast whose size, cell polarity and gene expression are similar to a day 10 human epiblast. A defined dose of BMP4 spontaneously breaks axial symmetry, and induces markers of the primitive streak and epithelial-to-mesenchymal transition. We show that WNT signalling and its inhibitor DKK1 play key roles in this process downstream of BMP4. Our work demonstrates that a model human epiblast can break axial symmetry despite the absence of asymmetry in the initial signal and of extra-embryonic tissues or maternal cues. Our three-dimensional model is an assay for the molecular events underlying human axial symmetry breaking.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Gene Expression Regulation, Developmental/physiology , Germ Layers/metabolism , Primitive Streak/metabolism , Tissue Culture Techniques , Cell Polarity/physiology , Epithelial-Mesenchymal Transition , Gastrulation/physiology , Humans , Primitive Streak/embryology , Signal Transduction/physiology
17.
Development ; 146(6)2019 03 25.
Article in English | MEDLINE | ID: mdl-30814117

ABSTRACT

Long-range signaling by morphogens and their inhibitors define embryonic patterning yet quantitative data and models are rare, especially in humans. Here, we use a human embryonic stem cell micropattern system to model formation of the primitive streak (PS) by WNT. In the pluripotent state, E-cadherin (E-CAD) transduces boundary forces to focus WNT signaling to the colony border. Following application of WNT ligand, E-CAD mediates a front or wave of epithelial-to-mesenchymal (EMT) conversion analogous to PS extension in an embryo. By knocking out the secreted WNT inhibitors active in our system, we show that DKK1 alone controls the extent and duration of patterning. The NODAL inhibitor cerberus 1 acts downstream of WNT to refine the endoderm versus mesoderm fate choice. Our EMT wave is a generic property of a bistable system with diffusion and we present a single quantitative model that describes both the wave and our knockout data.


Subject(s)
Body Patterning , Human Embryonic Stem Cells/cytology , Primitive Streak/embryology , Wnt Proteins/metabolism , Wnt Signaling Pathway , Actin Cytoskeleton/metabolism , Animals , Cell Line , Cell Lineage , Cytokines/metabolism , Endoderm/metabolism , Epithelial-Mesenchymal Transition , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Mesoderm/metabolism , Mice , Phenotype , Pluripotent Stem Cells/cytology , Protein Domains , Transforming Growth Factor beta/metabolism , Wnt3A Protein/metabolism
18.
Nature ; 566(7745): 490-495, 2019 02.
Article in English | MEDLINE | ID: mdl-30787436

ABSTRACT

Across the animal kingdom, gastrulation represents a key developmental event during which embryonic pluripotent cells diversify into lineage-specific precursors that will generate the adult organism. Here we report the transcriptional profiles of 116,312 single cells from mouse embryos collected at nine sequential time points ranging from 6.5 to 8.5 days post-fertilization. We construct a molecular map of cellular differentiation from pluripotency towards all major embryonic lineages, and explore the complex events involved in the convergence of visceral and primitive streak-derived endoderm. Furthermore, we use single-cell profiling to show that Tal1-/- chimeric embryos display defects in early mesoderm diversification, and we thus demonstrate how combining temporal and transcriptional information can illuminate gene function. Together, this comprehensive delineation of mammalian cell differentiation trajectories in vivo represents a baseline for understanding the effects of gene mutations during development, as well as a roadmap for the optimization of in vitro differentiation protocols for regenerative medicine.


Subject(s)
Cell Differentiation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gastrulation , Organogenesis , Single-Cell Analysis , Animals , Cell Lineage/genetics , Chimera/embryology , Chimera/genetics , Chimera/metabolism , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Endothelium/cytology , Endothelium/embryology , Endothelium/metabolism , Female , Gastrulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Hematopoiesis/genetics , Male , Mesoderm/cytology , Mesoderm/embryology , Mice , Mutation/genetics , Myeloid Cells/cytology , Organogenesis/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Primitive Streak/cytology , Primitive Streak/embryology , T-Cell Acute Lymphocytic Leukemia Protein 1/deficiency , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics
19.
Development ; 146(1)2019 01 02.
Article in English | MEDLINE | ID: mdl-30559277

ABSTRACT

The elongating mouse anteroposterior axis is supplied by progenitors with distinct tissue fates. It is not known whether these progenitors confer anteroposterior pattern to the embryo. We have analysed the progenitor population transcriptomes in the mouse primitive streak and tail bud throughout axial elongation. Transcriptomic signatures distinguish three known progenitor types (neuromesodermal, lateral/paraxial mesoderm and notochord progenitors; NMPs, LPMPs and NotoPs). Both NMP and LPMP transcriptomes change extensively over time. In particular, NMPs upregulate Wnt, Fgf and Notch signalling components, and many Hox genes as progenitors transit from production of the trunk to the tail and expand in number. In contrast, the transcriptome of NotoPs is stable throughout axial elongation and they are required for normal axis elongation. These results suggest that NotoPs act as a progenitor niche whereas anteroposterior patterning originates within NMPs and LPMPs.


Subject(s)
Body Patterning/physiology , Embryo, Mammalian/embryology , Mesoderm/embryology , Notochord/embryology , Signal Transduction/physiology , Animals , Embryo, Mammalian/cytology , Mesoderm/cytology , Mice , Mice, Transgenic , Notochord/cytology , Primitive Streak/cytology , Primitive Streak/embryology , Receptors, Notch/genetics , Receptors, Notch/metabolism
20.
Curr Opin Cell Biol ; 55: 81-86, 2018 12.
Article in English | MEDLINE | ID: mdl-30015151

ABSTRACT

The large display of body shapes and sizes observed among vertebrates ultimately represent variations of a common basic body plan. This likely results from the use of homologous developmental schemes, just differentially tinkered both in amplitude and timing by natural selection. In this review, we will revisit, discuss and combine old ideas with new concepts to update our view on how the vertebrate body is built. Recent advances, particularly at the molecular level, will guide our deconstruction of the individual developmental modules that sequentially produce head, neck, trunk and tail structures, and the transitions between them.


Subject(s)
Body Patterning/genetics , Vertebrates/genetics , Animals , Biological Evolution , Mesoderm/embryology , Models, Biological , Primitive Streak/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...