Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
Add more filters










Publication year range
1.
Cell ; 187(13): 3224-3228, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906097

ABSTRACT

The next 50 years of developmental biology will illuminate exciting new discoveries but are also poised to provide solutions to important problems society faces. Ten scientists whose work intersects with developmental biology in various capacities tell us about their vision for the future.


Subject(s)
Developmental Biology , Developmental Biology/trends , Humans , Stem Cells/cytology , Animals , Stem Cell Research
3.
Nat Struct Mol Biol ; 31(6): 964-976, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38789684

ABSTRACT

The mouse and human embryo gradually loses totipotency before diversifying into the inner cell mass (ICM, future organism) and trophectoderm (TE, future placenta). The transcription factors TFAP2C and TEAD4 with activated RHOA accelerate embryo polarization. Here we show that these factors also accelerate the loss of totipotency. TFAP2C and TEAD4 paradoxically promote and inhibit Hippo signaling before lineage diversification: they drive expression of multiple Hippo regulators while also promoting apical domain formation, which inactivates Hippo. Each factor activates TE specifiers in bipotent cells, while TFAP2C also activates specifiers of the ICM fate. Asymmetric segregation of the apical domain reconciles the opposing regulation of Hippo signaling into Hippo OFF and the TE fate, or Hippo ON and the ICM fate. We propose that the bistable switch established by TFAP2C and TEAD4 is exploited to trigger robust lineage diversification in the developing embryo.


Subject(s)
DNA-Binding Proteins , TEA Domain Transcription Factors , Transcription Factor AP-2 , Transcription Factors , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice , Humans , Signal Transduction , Cell Lineage , Gene Expression Regulation, Developmental , Muscle Proteins/metabolism , Muscle Proteins/genetics , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Hippo Signaling Pathway , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Embryonic Development/genetics
4.
Cell ; 187(11): 2838-2854.e17, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38744282

ABSTRACT

Retrospective lineage reconstruction of humans predicts that dramatic clonal imbalances in the body can be traced to the 2-cell stage embryo. However, whether and how such clonal asymmetries arise in the embryo is unclear. Here, we performed prospective lineage tracing of human embryos using live imaging, non-invasive cell labeling, and computational predictions to determine the contribution of each 2-cell stage blastomere to the epiblast (body), hypoblast (yolk sac), and trophectoderm (placenta). We show that the majority of epiblast cells originate from only one blastomere of the 2-cell stage embryo. We observe that only one to three cells become internalized at the 8-to-16-cell stage transition. Moreover, these internalized cells are more frequently derived from the first cell to divide at the 2-cell stage. We propose that cell division dynamics and a cell internalization bottleneck in the early embryo establish asymmetry in the clonal composition of the future human body.


Subject(s)
Blastomeres , Cell Lineage , Embryo, Mammalian , Female , Humans , Blastomeres/cytology , Blastomeres/metabolism , Cell Division , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development , Germ Layers/cytology , Germ Layers/metabolism , Male , Animals , Mice
5.
Dev Cell ; 59(10): 1252-1268.e13, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38579720

ABSTRACT

The blueprint of the mammalian body plan is laid out during gastrulation, when a trilaminar embryo is formed. This process entails a burst of proliferation, the ingression of embryonic epiblast cells at the primitive streak, and their priming toward primitive streak fates. How these different events are coordinated remains unknown. Here, we developed and characterized a 3D culture of self-renewing mouse embryonic cells that captures the main transcriptional and architectural features of the early gastrulating mouse epiblast. Using this system in combination with microfabrication and in vivo experiments, we found that proliferation-induced crowding triggers delamination of cells that express high levels of the apical polarity protein aPKC. Upon delamination, cells become more sensitive to Wnt signaling and upregulate the expression of primitive streak markers such as Brachyury. This mechanistic coupling between ingression and differentiation ensures that the right cell types become specified at the right place during embryonic development.


Subject(s)
Cell Differentiation , Gastrulation , Germ Layers , Animals , Mice , Germ Layers/cytology , Germ Layers/metabolism , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Primitive Streak/cytology , Primitive Streak/metabolism , Fetal Proteins/metabolism , Fetal Proteins/genetics , Wnt Signaling Pathway , Cell Proliferation , Gene Expression Regulation, Developmental , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism
6.
Nat Cell Biol ; 26(3): 353-365, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38443567

ABSTRACT

Development requires coordinated interactions between the epiblast, which generates the embryo proper; the trophectoderm, which generates the placenta; and the hypoblast, which forms both the anterior signalling centre and the yolk sac. These interactions remain poorly understood in human embryogenesis because mechanistic studies have only recently become possible. Here we examine signalling interactions post-implantation using human embryos and stem cell models of the epiblast and hypoblast. We find anterior hypoblast specification is NODAL dependent, as in the mouse. However, while BMP inhibits anterior signalling centre specification in the mouse, it is essential for its maintenance in human. We also find contrasting requirements for BMP in the naive pre-implantation epiblast of mouse and human embryos. Finally, we show that NOTCH signalling is important for human epiblast survival. Our findings of conserved and species-specific factors that drive these early stages of embryonic development highlight the strengths of comparative species studies.


Subject(s)
Embryo, Mammalian , Germ Layers , Pregnancy , Female , Humans , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Signal Transduction , Embryo Implantation
7.
Methods Mol Biol ; 2767: 123-134, 2024.
Article in English | MEDLINE | ID: mdl-37284940

ABSTRACT

In this chapter, we detail the experimental protocol leading to the generation of stem cell-based mouse embryo-like structures termed "ETiX-embryoids." ETiX-embryoids are formed from combined embryonic stem cells, trophoblast stem cells, and embryonic stem cells transiently induced to express Gata4. Cells are seeded into AggreWell dishes where they form aggregates that develop to resemble post-implantation mouse embryos following 4 days of culture. ETiX-embryoids establish an anterior signaling center and undergo gastrulation over the following 2 days. By day 7, ETiX-embryoids undergo neurulation and form an anterior-posterior axis with head folds at one end and a tail bud on the other. On day 8, they develop a brain and form a heart-like structure and a gut tube.


Subject(s)
Embryo, Mammalian , Embryonic Development , Mice , Animals , Gastrulation , Embryonic Stem Cells , Trophoblasts
8.
Curr Opin Genet Dev ; 84: 102134, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38052116

ABSTRACT

Stem cell-based mammalian embryo models facilitate the discovery of developmental mechanisms because they are more amenable to genetic and epigenetic perturbations than natural embryos. Here, we highlight exciting recent advances that have yielded a plethora of models of embryonic development. Imperfections in these models highlight gaps in our current understanding and outline future research directions, ushering in an exciting new era for embryology.


Subject(s)
Embryo, Mammalian , Embryonic Development , Animals , Female , Pregnancy , Embryonic Development/genetics , Organogenesis , Stem Cells , Mammals
9.
Nat Methods ; 20(12): 1844-1848, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38057512
10.
Nat Protoc ; 18(12): 3662-3689, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37821625

ABSTRACT

The interaction between embryonic and extraembryonic tissues is critical in natural mouse embryogenesis. Here, to enable such interaction in vitro, we describe a protocol to assemble a complete mouse embryo model using mouse embryonic stem cells and induced embryonic stem cells to express Cdx2 (or trophoblast stem cells) and Gata4 to reconstitute the epiblast, extraembryonic ectoderm and visceral endoderm lineages, respectively. The resulting complete embryo models recapitulate development from embryonic day 5.0 to 8.5, generating advanced embryonic and extraembryonic tissues that develop through gastrulation to initiate organogenesis to form a head and a beating heart structure as well as a yolk sac and chorion. Once the required stem cell lines are stably maintained in culture, the protocol requires 1 day to assemble complete embryo models and a further 8 days to culture them until headfold stages, although structures can be collected at earlier developmental stages as required. This protocol can be easily performed by researchers with experience in mouse stem cell culture, although they will benefit from knowledge of natural mouse embryos at early postimplantation stages.


Subject(s)
Embryo, Mammalian , Germ Layers , Mice , Animals , Endoderm , Embryonic Development , Embryonic Stem Cells
12.
Nat Commun ; 14(1): 4788, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37553326

ABSTRACT

Droplet microfluidic methods have massively increased the throughput of single-cell sequencing campaigns. The benefit of scale-up is, however, accompanied by increased background noise when processing challenging samples and the overall RNA capture efficiency is lower. These drawbacks stem from the lack of strategies to enrich for high-quality material or specific cell types at the moment of cell encapsulation and the absence of implementable multi-step enzymatic processes that increase capture. Here we alleviate both bottlenecks using fluorescence-activated droplet sorting to enrich for droplets that contain single viable cells, intact nuclei, fixed cells or target cell types and use reagent addition to droplets by picoinjection to perform multi-step lysis and reverse transcription. Our methodology increases gene detection rates fivefold, while reducing background noise by up to half. We harness these properties to deliver a high-quality molecular atlas of mouse brain development, despite starting with highly damaged input material, and provide an atlas of nascent RNA transcription during mouse organogenesis. Our method is broadly applicable to other droplet-based workflows to deliver sensitive and accurate single-cell profiling at a reduced cost.


Subject(s)
Microfluidic Analytical Techniques , Microfluidics , Animals , Mice , Microfluidic Analytical Techniques/methods , RNA , Single-Cell Analysis/methods
13.
Nature ; 622(7983): 584-593, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37369347

ABSTRACT

The human embryo undergoes morphogenetic transformations following implantation into the uterus, but our knowledge of this crucial stage is limited by the inability to observe the embryo in vivo. Models of the embryo derived from stem cells are important tools for interrogating developmental events and tissue-tissue crosstalk during these stages1. Here we establish a model of the human post-implantation embryo, a human embryoid, comprising embryonic and extraembryonic tissues. We combine two types of extraembryonic-like cell generated by overexpression of transcription factors with wild-type embryonic stem cells and promote their self-organization into structures that mimic several aspects of the post-implantation human embryo. These self-organized aggregates contain a pluripotent epiblast-like domain surrounded by extraembryonic-like tissues. Our functional studies demonstrate that the epiblast-like domain robustly differentiates into amnion, extraembryonic mesenchyme and primordial germ cell-like cells in response to bone morphogenetic protein cues. In addition, we identify an inhibitory role for SOX17 in the specification of anterior hypoblast-like cells2. Modulation of the subpopulations in the hypoblast-like compartment demonstrates that extraembryonic-like cells influence epiblast-like domain differentiation, highlighting functional tissue-tissue crosstalk. In conclusion, we present a modular, tractable, integrated3 model of the human embryo that will enable us to probe key questions of human post-implantation development, a critical window during which substantial numbers of pregnancies fail.


Subject(s)
Embryo Implantation , Embryo, Mammalian , Embryonic Development , Models, Biological , Pluripotent Stem Cells , Female , Humans , Pregnancy , Bone Morphogenetic Proteins , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryoid Bodies/cytology , Germ Layers/cytology , Germ Layers/embryology , Human Embryonic Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Pluripotent Stem Cells/cytology
15.
Curr Top Dev Biol ; 154: 169-196, 2023.
Article in English | MEDLINE | ID: mdl-37100517

ABSTRACT

The first cell fate decision is the process by which cells of an embryo take on distinct lineage identities for the first time, representing the beginning of patterning during development. In mammals, this process separates an embryonic inner cell mass lineage (future new organism) from an extra-embryonic trophectoderm lineage (future placenta), and in the mouse, this is classically attributed to the consequences of apical-basal polarity. The mouse embryo acquires this polarity at the 8-cell stage, indicated by cap-like protein domains on the apical surface of each cell; those cells which retain polarity over subsequent divisions are specified as trophectoderm, and the rest as inner cell mass. Recent research has advanced our knowledge of this process - this review will discuss mechanisms behind the establishment of polarity and distribution of the apical domain, different factors affecting the first cell fate decision including heterogeneities between cells of the very early embryo, and the conservation of developmental mechanisms across species, including human.


Subject(s)
Blastocyst , Embryo, Mammalian , Animals , Mice , Humans , Cell Lineage , Cell Differentiation , Cell Polarity , Mammals
17.
Nat Cell Biol ; 24(9): 1341-1349, 2022 09.
Article in English | MEDLINE | ID: mdl-36100738

ABSTRACT

Mammalian embryos sequentially differentiate into trophectoderm and an inner cell mass, the latter of which differentiates into primitive endoderm and epiblast. Trophoblast stem (TS), extraembryonic endoderm (XEN) and embryonic stem (ES) cells derived from these three lineages can self-assemble into synthetic embryos, but the mechanisms remain unknown. Here, we show that a stem cell-specific cadherin code drives synthetic embryogenesis. The XEN cell cadherin code enables XEN cell sorting into a layer below ES cells, recapitulating the sorting of epiblast and primitive endoderm before implantation. The TS cell cadherin code enables TS cell sorting above ES cells, resembling extraembryonic ectoderm clustering above epiblast following implantation. Whereas differential cadherin expression drives initial cell sorting, cortical tension consolidates tissue organization. By optimizing cadherin code expression in different stem cell lines, we tripled the frequency of correctly formed synthetic embryos. Thus, by exploiting cadherin codes from different stages of development, lineage-specific stem cells bypass the preimplantation structure to directly assemble a postimplantation embryo.


Subject(s)
Cadherins , Endoderm , Mammals/embryology , Animals , Blastocyst , Cadherins/genetics , Cadherins/metabolism , Embryonic Stem Cells/metabolism , Germ Layers
18.
Cell Stem Cell ; 29(10): 1445-1458.e8, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36084657

ABSTRACT

Several in vitro models have been developed to recapitulate mouse embryogenesis solely from embryonic stem cells (ESCs). Despite mimicking many aspects of early development, they fail to capture the interactions between embryonic and extraembryonic tissues. To overcome this difficulty, we have developed a mouse ESC-based in vitro model that reconstitutes the pluripotent ESC lineage and the two extraembryonic lineages of the post-implantation embryo by transcription-factor-mediated induction. This unified model recapitulates developmental events from embryonic day 5.5 to 8.5, including gastrulation; formation of the anterior-posterior axis, brain, and a beating heart structure; and the development of extraembryonic tissues, including yolk sac and chorion. Comparing single-cell RNA sequencing from individual structures with time-matched natural embryos identified remarkably similar transcriptional programs across lineages but also showed when and where the model diverges from the natural program. Our findings demonstrate an extraordinary plasticity of ESCs to self-organize and generate a whole-embryo-like structure.


Subject(s)
Embryo, Mammalian , Neurulation , Animals , Embryonic Development , Embryonic Stem Cells , Mice , Mouse Embryonic Stem Cells
19.
Nature ; 610(7930): 143-153, 2022 10.
Article in English | MEDLINE | ID: mdl-36007540

ABSTRACT

Embryonic stem (ES) cells can undergo many aspects of mammalian embryogenesis in vitro1-5, but their developmental potential is substantially extended by interactions with extraembryonic stem cells, including trophoblast stem (TS) cells, extraembryonic endoderm stem (XEN) cells and inducible XEN (iXEN) cells6-11. Here we assembled stem cell-derived embryos in vitro from mouse ES cells, TS cells and iXEN cells and showed that they recapitulate the development of whole natural mouse embryo in utero up to day 8.5 post-fertilization. Our embryo model displays headfolds with defined forebrain and midbrain regions and develops a beating heart-like structure, a trunk comprising a neural tube and somites, a tail bud containing neuromesodermal progenitors, a gut tube, and primordial germ cells. This complete embryo model develops within an extraembryonic yolk sac that initiates blood island development. Notably, we demonstrate that the neurulating embryo model assembled from Pax6-knockout ES cells aggregated with wild-type TS cells and iXEN cells recapitulates the ventral domain expansion of the neural tube that occurs in natural, ubiquitous Pax6-knockout embryos. Thus, these complete embryoids are a powerful in vitro model for dissecting the roles of diverse cell lineages and genes in development. Our results demonstrate the self-organization ability of ES cells and two types of extraembryonic stem cells to reconstitute mammalian development through and beyond gastrulation to neurulation and early organogenesis.


Subject(s)
Embryo, Mammalian , Gastrulation , Models, Biological , Neurulation , Organogenesis , Animals , Cell Lineage , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Stem Cells/cytology , Endoderm/cytology , Endoderm/embryology , Heart/embryology , Mesencephalon/embryology , Mice , Neural Tube/embryology , PAX6 Transcription Factor/deficiency , PAX6 Transcription Factor/genetics , Prosencephalon/embryology , Somites/embryology
20.
Curr Opin Genet Dev ; 76: 101970, 2022 10.
Article in English | MEDLINE | ID: mdl-35988317

ABSTRACT

Synthetic embryology aims to develop embryo-like structures from stem cells to provide new insight into early stages of mammalian development. Recent advances in synthetic embryology have highlighted the remarkable capacity of stem cells to self-organize under certain biochemical or biophysical stimulations, generating structures that recapitulate the fate and form of early mouse/human embryos, in which symmetry breaking, pattern formation, or proper morphogenesis can be observed spontaneously. Here we review recent progress on the design principles for different types of embryoids and discuss the impact of different biochemical and biophysical factors on the process of stem-cell self-organization. We also offer our thoughts about the principal future challenges.


Subject(s)
Embryo, Mammalian , Stem Cells , Animals , Embryonic Development/genetics , Humans , Mammals , Mice , Morphogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...