Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters








Publication year range
1.
Cells ; 10(12)2021 12 16.
Article in English | MEDLINE | ID: mdl-34944066

ABSTRACT

During the development of a multicellular organism, the specification of different cell lineages originates in a small group of pluripotent cells, the epiblasts, formed in the preimplantation embryo. The pluripotent epiblast is protected from premature differentiation until exposure to inductive cues in strictly controlled spatially and temporally organized patterns guiding fetus formation. Epiblasts cultured in vitro are embryonic stem cells (ESCs), which recapitulate the self-renewal and lineage specification properties of their endogenous counterparts. The characteristics of totipotency, although less understood than pluripotency, are becoming clearer. Recent studies have shown that a minor ESC subpopulation exhibits expanded developmental potential beyond pluripotency, displaying a characteristic reminiscent of two-cell embryo blastomeres (2CLCs). In addition, reprogramming both mouse and human ESCs in defined media can produce expanded/extended pluripotent stem cells (EPSCs) similar to but different from 2CLCs. Further, the molecular roadmaps driving the transition of various potency states have been clarified. These recent key findings will allow us to understand eutherian mammalian development by comparing the underlying differences between potency network components during development. Using the mouse as a paradigm and recent progress in human PSCs, we review the epiblast's identity acquisition during embryogenesis and their ESC counterparts regarding their pluripotent fates and beyond.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Germ Layers/growth & development , Pluripotent Stem Cells/cytology , Animals , Blastocyst/metabolism , Cell Lineage/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Humans , Mice
2.
Elife ; 102021 08 17.
Article in English | MEDLINE | ID: mdl-34403333

ABSTRACT

Human embryogenesis entails complex signalling interactions between embryonic and extra-embryonic cells. However, how extra-embryonic cells direct morphogenesis within the human embryo remains largely unknown due to a lack of relevant stem cell models. Here, we have established conditions to differentiate human pluripotent stem cells (hPSCs) into yolk sac-like cells (YSLCs) that resemble the post-implantation human hypoblast molecularly and functionally. YSLCs induce the expression of pluripotency and anterior ectoderm markers in human embryonic stem cells (hESCs) at the expense of mesoderm and endoderm markers. This activity is mediated by the release of BMP and WNT signalling pathway inhibitors, and, therefore, resembles the functioning of the anterior visceral endoderm signalling centre of the mouse embryo, which establishes the anterior-posterior axis. Our results implicate the yolk sac in epiblast cell fate specification in the human embryo and propose YSLCs as a tool for studying post-implantation human embryo development in vitro.


Subject(s)
Germ Layers/growth & development , Pluripotent Stem Cells/metabolism , Yolk Sac/growth & development , Animals , Cell Line , Ectoderm/growth & development , Embryonic Development , Humans , Mice
3.
Biochem J ; 478(9): 1809-1825, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33988704

ABSTRACT

Epithelial plasticity involved the terminal and transitional stages that occur during epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET), both are essential at different stages of early embryonic development that have been co-opted by cancer cells to undergo tumor metastasis. These processes are regulated at multiple instances, whereas the post-transcriptional regulation of key genes mediated by microRNAs is gaining major attention as a common and conserved pathway. In this review, we focus on discussing the latest findings of the cellular and molecular basis of the less characterized process of MET during embryonic development, with special attention to the role of microRNAs. Although we take in consideration the necessity of being cautious when extrapolating the obtained evidence, we propose some commonalities between early embryonic development and cancer progression that can shed light into our current understanding of this complex event and might aid in the design of specific therapeutic approaches.


Subject(s)
Embryonic Development/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Disease Progression , Embryo, Mammalian , Gene Expression Regulation, Neoplastic , Germ Layers/cytology , Germ Layers/growth & development , Germ Layers/metabolism , Humans , MicroRNAs/classification , MicroRNAs/metabolism , Neoplasm Metastasis , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Somites/cytology , Somites/growth & development , Somites/metabolism
4.
Cell Rep ; 34(10): 108834, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691117

ABSTRACT

At implantation, the embryo establishes contacts with the maternal endometrium. This stage is associated with a high incidence of preclinical pregnancy losses. While the maternal factors underlying uterine receptivity have been investigated, the signals required by the embryo for successful peri-implantation development remain elusive. To explore these, we studied integrin ß1 signaling, as embryos deficient for this receptor degenerate at implantation. We demonstrate that the coordinated action of pro-survival signals and localized actomyosin suppression via integrin ß1 permits the development of the embryo beyond implantation. Failure of either process leads to developmental arrest and apoptosis. Pharmacological stimulation through fibroblast growth factor 2 (FGF2) and insulin-like growth factor 1 (IGF1), coupled with ROCK-mediated actomyosin inhibition, rescues the deficiency of integrin ß1, promoting progression to post-implantation stages. Mutual exclusion between integrin ß1 and actomyosin seems to be conserved in the human embryo, suggesting the possibility that these mechanisms could also underlie the transition of the human epiblast from pre- to post-implantation.


Subject(s)
Integrin beta1/metabolism , Morphogenesis , Actomyosin/metabolism , Amides/pharmacology , Animals , Embryo Implantation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development , Female , Fibroblast Growth Factor 2/pharmacology , Germ Layers/growth & development , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Insulin-Like Growth Factor I/pharmacology , Integrin beta1/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Morphogenesis/drug effects , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pyridines/pharmacology , Signal Transduction , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
5.
Elife ; 92020 09 24.
Article in English | MEDLINE | ID: mdl-32969790

ABSTRACT

Two distinct mechanisms for primordial germ cell (PGC) specification are observed within Bilatera: early determination by maternal factors or late induction by zygotic cues. Here we investigate the molecular basis for PGC specification in Nematostella, a representative pre-bilaterian animal where PGCs arise as paired endomesodermal cell clusters during early development. We first present evidence that the putative PGCs delaminate from the endomesoderm upon feeding, migrate into the gonad primordia, and mature into germ cells. We then show that the PGC clusters arise at the interface between hedgehog1 and patched domains in the developing mesenteries and use gene knockdown, knockout and inhibitor experiments to demonstrate that Hh signaling is required for both PGC specification and general endomesodermal patterning. These results provide evidence that the Nematostella germline is specified by inductive signals rather than maternal factors, and support the existence of zygotically-induced PGCs in the eumetazoan common ancestor.


Subject(s)
Body Patterning/genetics , Germ Layers , Hedgehog Proteins , Sea Anemones , Signal Transduction/genetics , Animals , Female , Gene Knockdown Techniques , Germ Cells/cytology , Germ Cells/metabolism , Germ Layers/cytology , Germ Layers/growth & development , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Life Cycle Stages/genetics , Male , Sea Anemones/cytology , Sea Anemones/genetics , Sea Anemones/growth & development
6.
Mech Dev ; 163: 103628, 2020 09.
Article in English | MEDLINE | ID: mdl-32603823

ABSTRACT

Among the basally branching metazoans, cnidarians display well-defined gastrulation processes leading to a diploblastic body plan, consisting of an endodermal and an ectodermal cell layer. As the outgroup to all Bilateria, cnidarians are an interesting group to investigate ancestral developmental mechanisms. Interestingly, all known gastrulation mechanisms known in Bilateria are already found in different species of Cnidaria. Here I review the morphogenetic processes found in different Cnidaria and focus on the investigation of the cellular and molecular mechanisms in the sea anemone Nematostella vectensis, which has been a major model organism among cnidarians for evolutionary developmental biology. Many of the genes involved in germ layer specification and morphogenetic processes in Bilateria are also found active during gastrulation of Nematostella and other cnidarians, suggesting an ancestral role of this process. The molecular analyses indicate a tight link between gastrulation and axis patterning processes by Wnt and FGF signaling. Interestingly, the endodermal layer displays many features of the mesodermal layer in Bilateria, while the pharyngeal ectoderm has an endodermal expression profile. Comparative analyses as well as experimental studies using embryonic aggregates suggest that minor differences in the gene regulatory networks allow the embryo to transition relatively easily from one mode of gastrulation to another.


Subject(s)
Body Patterning/genetics , Cnidaria/genetics , Gastrulation/genetics , Germ Layers/growth & development , Animals , Cnidaria/growth & development , Ectoderm/growth & development , Embryo, Nonmammalian/physiology , Endoderm/growth & development , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Germ Layers/metabolism , Mesoderm/growth & development , Sea Anemones/genetics , Sea Anemones/growth & development , Signal Transduction/genetics
7.
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
8.
Mech Dev ; 163: 103617, 2020 09.
Article in English | MEDLINE | ID: mdl-32473204

ABSTRACT

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.


Subject(s)
Body Patterning/genetics , Cell Differentiation/genetics , Embryonic Development/genetics , Gastrulation/genetics , Organ Specificity/genetics , Animals , Cell Lineage/genetics , Ectoderm/growth & development , Endoderm/growth & development , Female , Gastrula/growth & development , Gastrulation/physiology , Germ Layers/growth & development , Mesoderm/growth & development , Mice , Pregnancy
9.
J Cell Physiol ; 235(11): 8640-8652, 2020 11.
Article in English | MEDLINE | ID: mdl-32324269

ABSTRACT

We recently reported that epiblast stem cells (EpiSCs)-like cells could be derived from preimplantation embryos (named as AFSCs). Here, we established AFSCs from pre-implantation embryos of multiple mouse strains and showed that unlike EpiSCs, the derivation efficiency of AFSCs was affected by the genetic background. We then used AFSCs lines to dissect the roles of Activin A (Act A) and basic fibroblast growth factor and reported that Act A alone was capable of maintaining self-renewal but not developmental potential in vivo. Finally, we established a novel experimental system, in which AFSCs were efficiently converted to multipotent progenitor stem cells using Act A and bone morphogenetic protein 4 (named as ABSCs). Importantly, these ABSCs contributed to neural mesodermal progenitors and lateral plate mesoderm in postimplantation chimeras. Taken together, our study established a robust experimental system for the generation of specific multipotent progenitor stem cells that was self-renewable and capable of contributing to embryonic and extra-embryonic tissues.


Subject(s)
Activins/pharmacology , Germ Layers/drug effects , Multipotent Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , Activins/metabolism , Animals , Blastocyst/drug effects , Blastocyst/metabolism , Bone Morphogenetic Protein 4/drug effects , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Embryonic Development/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Germ Layers/growth & development , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects
10.
Exp Cell Res ; 390(2): 111969, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32199920

ABSTRACT

The degree of polymorphism, i.e., DNA sequence divergence, of short AT-rich tandemly arranged simple sequence repeats at or near promoters and 5'- untranslated regions of mRNA may quantitatively regulate transcription of tissue-specific genes. Less polymorphic repeats allow greater gene expression. Preferential binding of hypophosphorylated H1 histone to these repeats may diminish binding of transcription factors. Preferential binding of hypophosphorylated high mobility group chromatin proteins would increase this binding. Shorter simple sequence repeats have undergone fewer point mutations than longer repeats, hence they are less polymorphic and more conserved. The role of transcribed simple sequence repeats in frog embryo germ layer determination is considered.


Subject(s)
High Mobility Group Proteins/genetics , Histones/genetics , Microsatellite Repeats , Polymorphism, Genetic , Transcription Factors/genetics , Transcription, Genetic , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Chromatin/metabolism , Chromatin/ultrastructure , Embryo, Nonmammalian , Germ Layers/cytology , Germ Layers/growth & development , Germ Layers/metabolism , High Mobility Group Proteins/metabolism , Histones/metabolism , Humans , Mice , Phosphorylation , Point Mutation , Promoter Regions, Genetic , Transcription Factors/metabolism , Xenopus/genetics , Xenopus/growth & development , Xenopus/metabolism
11.
Differentiation ; 112: 67-76, 2020.
Article in English | MEDLINE | ID: mdl-32045848

ABSTRACT

To induce and maintain naïve pluripotency in mouse embryonic and induced pluripotent stem cells (ESCs/iPSCs), chemically defined N2B27 medium with PD0325901, CHIR99021, and leukemia inhibitory factor (2i/LIF) is a classic and simple condition. However, this method cannot be simply extrapolated to human ESCs/iPSCs that are principally stabilized in primed pluripotency and become primitive neuroepithelium-like cells in N2B27+2i/LIF culture. Here, we assessed iPSC reprogramming of fibroblasts from chimpanzee, our closest living relative, in N2B27+2i/LIF culture. Under this condition, chimpanzee cells formed alkaline phosphatase-positive dome-shaped colonies. The colony-forming cells could be stably expanded by serial passaging without a ROCK inhibitor. However, their gene expression was distinct from iPSCs and neuroepithelium. They expressed the OCT3/4 transgene and a subset of transcripts associated with pluripotency, mesenchymal-epithelial transition, and neural crest formation. These cells exhibited a differentiation potential into the three germ layers in vivo and in vitro. The current study demonstrated that iPSC reprogramming in N2B27+2i/LIF culture converted chimpanzee fibroblasts into a multipotent cancerous state with unique gene expression, but not fully pluripotent stem cells.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Multipotent Stem Cells/cytology , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Cellular Reprogramming/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Germ Layers/drug effects , Germ Layers/growth & development , Humans , Leukemia Inhibitory Factor/pharmacology , Mice , Multipotent Stem Cells/drug effects , Neural Crest/cytology , Pan troglodytes , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology
12.
Nat Commun ; 10(1): 4269, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31537794

ABSTRACT

Embryonic development yields many different cell types in response to just a few families of inductive signals. The property of signal-receiving cells that determines how they respond to inductive signals is known as competence, and it differs in different cell types. Here, we explore the ways in which maternal factors modify chromatin to specify initial competence in the frog Xenopus tropicalis. We identify early-engaged regulatory DNA sequences, and infer from them critical activators of the zygotic genome. Of these, we show that the pioneering activity of the maternal pluripotency factors Pou5f3 and Sox3 determines competence for germ layer formation by extensively remodelling compacted chromatin before the onset of inductive signalling. This remodelling includes the opening and marking of thousands of regulatory elements, extensive chromatin looping, and the co-recruitment of signal-mediating transcription factors. Our work identifies significant developmental principles that inform our understanding of how pluripotent stem cells interpret inductive signals.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Homeodomain Proteins/genetics , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/genetics , Transcription Factors/genetics , Xenopus Proteins/genetics , Xenopus/embryology , Animals , Cell Differentiation/genetics , Chromatin/metabolism , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Germ Layers/growth & development , Regulatory Sequences, Nucleic Acid/genetics , Xenopus/genetics
13.
Nat Cell Biol ; 21(6): 687-699, 2019 06.
Article in English | MEDLINE | ID: mdl-31160711

ABSTRACT

We recently derived mouse expanded potential stem cells (EPSCs) from individual blastomeres by inhibiting the critical molecular pathways that predispose their differentiation. EPSCs had enriched molecular signatures of blastomeres and possessed developmental potency for all embryonic and extra-embryonic cell lineages. Here, we report the derivation of porcine EPSCs, which express key pluripotency genes, are genetically stable, permit genome editing, differentiate to derivatives of the three germ layers in chimeras and produce primordial germ cell-like cells in vitro. Under similar conditions, human embryonic stem cells and induced pluripotent stem cells can be converted, or somatic cells directly reprogrammed, to EPSCs that display the molecular and functional attributes reminiscent of porcine EPSCs. Importantly, trophoblast stem-cell-like cells can be generated from both human and porcine EPSCs. Our pathway-inhibition paradigm thus opens an avenue for generating mammalian pluripotent stem cells, and EPSCs present a unique cellular platform for translational research in biotechnology and regenerative medicine.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Blastomeres/cytology , Blastomeres/metabolism , Cell Lineage/genetics , Embryonic Stem Cells/cytology , Germ Layers/growth & development , Germ Layers/metabolism , Humans , Mice , Regenerative Medicine , Signal Transduction/genetics , Swine , Trophoblasts/cytology , Trophoblasts/metabolism
14.
Dev Cell ; 50(4): 462-477.e5, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31204172

ABSTRACT

Controlling responsiveness to prevailing signals is critical for robust transitions between cell states during development. For example, fibroblast growth factor (FGF) drives naive pluripotent cells into extraembryonic lineages before implantation but sustains pluripotency in primed cells of the post-implantation epiblast. Nanog supports pluripotency in naive cells, while Nodal supports pluripotency in primed cells, but the handover from Nanog to Nodal does not proceed seamlessly, opening up the risk of aberrant differentiation if FGF is activated before Nodal. Here, we report that Id1 acts as a sensor to detect delays in Nodal activation after the downregulation of Nanog. Id1 then suppresses FGF activity to delay differentiation. Accordingly, Id1 is not required for naive or primed pluripotency but rather stabilizes epiblast identity during the transition between these states. These findings help explain how development proceeds robustly in the face of imprecise signals and highlight the importance of mechanisms that stabilize cell identity during developmental transitions.


Subject(s)
Embryonic Development/genetics , Inhibitor of Differentiation Protein 1/genetics , Nanog Homeobox Protein/genetics , Nodal Protein/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental/genetics , Germ Layers/growth & development , Germ Layers/metabolism , Humans , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/genetics
15.
In Vitro Cell Dev Biol Anim ; 55(5): 355-367, 2019 May.
Article in English | MEDLINE | ID: mdl-30993557

ABSTRACT

N-terminal acetylation (Nt-acetylation) refers to the acetylation of the free α-amino group at the N-terminus of a polypeptide. While the effects of Nt-acetylation are multifaceted, its most known function is in the acetylation-dependent N-end rule protein degradation pathway (Ac/N-end rule pathway), where Nt-acetylation is recognized as a degron by designated E3 ligases, eventually leading to target degradation by the ubiquitin-proteasome system. Naa10 is the catalytic subunit of the major Nt-acetylation enzyme NatA, which Nt-acetylates proteins whose second amino acid has a small side chain. In humans, NAA10 is the responsible mutated gene in Ogden syndrome and is thought to play important roles in development. However, it is unclear how the Ac/N-end rule pathway affects the differentiation ability of mouse embryonic stem cells (mESCs). We hypothesized that the balance of pluripotency factors may be maintained by the Ac/N-end rule pathway. Thus, we established Naa10 knockout mESCs to test this hypothesis. We found that Naa10 deficiency attenuated differentiation towards the epiblast lineage, deviating towards primitive endoderm. However, this was not caused by disturbing the balance of pluripotency factors, rather by augmenting FGF/MAPK signaling.


Subject(s)
Cell Lineage/genetics , Germ Layers/growth & development , Mouse Embryonic Stem Cells/metabolism , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/genetics , Acetylation , Animals , Cell Differentiation/genetics , Endoderm/growth & development , Endoderm/metabolism , Fibroblast Growth Factors/genetics , Gene Knockout Techniques , Germ Layers/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/metabolism , Protein Processing, Post-Translational/genetics , Proteolysis , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
16.
Phys Biol ; 16(4): 041002, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30875695

ABSTRACT

Epiblast is composed of pluripotent cells which will give rise to all cell lineages in a human body. It forms a single-cell layered epithelium conserved among all amniotic vertebrates (birds, reptiles and mammals) and undergoes complex morphogenesis both before and during gastrulation. Our knowledge of the amniote epiblast is based on data acquired through cellular and molecular analyses of early chick and mouse embryos in vivo and mammalian pluripotent stem cells (PSCs) in vitro. Very few studies have been published on biomechanical characteristics of the amniote epiblast, largely due to lack of experimental tools for measuring and perturbing biomechanical properties. Also missing is a conceptual framework that can integrate both biomechanical and molecular parameters of the epiblast. This review is aimed at providing a background based on which epiblast morphogenesis, including its transition between the epithelial and mesenchymal states, can be understood from a biomechanical perspective. This simple developmental biology system is suitable for testing a multitude of theoretical models in biomechanics, leading to a better understanding of biomechanical logics and constraints governing multicellular organization.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Germ Layers/cytology , Germ Layers/growth & development , Morphogenesis/physiology , Animals , Biomechanical Phenomena/physiology , Cell Communication/physiology , Cell Line , Gastrulation/physiology , Humans , Models, Theoretical
17.
Dev Growth Differ ; 60(8): 463-472, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30368783

ABSTRACT

Gastrulation is a key milestone in early mouse development when multipotent epiblast cells are allocated to progenitors of diverse tissue lineages that constitute the ensemble of building blocks of the body plan. The analysis of gene function revealed that the activity of transcription factors is likely to be the fundamental driving force underpinning the lineage specification and tissue patterning in the primary germ layers. The developmental-spatial transcriptome of the gastrulating embryo revealed the concerted and interactive activity of the gene regulatory network anchored by development-related transcription factors. The findings of the network structure offer novel insights into the regionalization of tissue fates and enable tracking of the progression of epiblast patterning, leading to the construction of molecularly annotated fate maps of epiblast during gastrulation.


Subject(s)
Gastrulation/genetics , Gene Regulatory Networks/genetics , Germ Layers/metabolism , Transcription Factors/metabolism , Animals , Germ Layers/cytology , Germ Layers/growth & development , Mice
18.
Annu Rev Genet ; 52: 185-201, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30183407

ABSTRACT

Establishing the different lineages of the early mammalian embryo takes place over several days and several rounds of cell divisions from the fertilized egg. The resulting blastocyst contains the pluripotent cells of the epiblast, from which embryonic stem cells can be derived, as well as the extraembryonic lineages required for a mammalian embryo to survive in the uterine environment. The dynamics of the cellular and genetic interactions controlling the initiation and maintenance of these lineages in the mouse embryo are increasingly well understood through application of the tools of single-cell genomics, gene editing, and in vivo imaging. Exploring the similarities and differences between mouse and human development will be essential for translation of these findings into new insights into human biology, derivation of stem cells, and improvements in fertility treatments.


Subject(s)
Cell Lineage/genetics , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Germ Layers/growth & development , Animals , Embryo, Mammalian , Gene Editing , Germ Layers/cytology , Humans , Mice , Pluripotent Stem Cells/cytology
19.
Article in English | MEDLINE | ID: mdl-29786554

ABSTRACT

We describe the production of a human induced pluripotent stem cell (iPSC) line, SFCi55-ZsGr, that has been engineered to express the fluorescent reporter gene, ZsGreen, in a constitutive manner. The CAG-driven ZsGreen expression cassette was inserted into the AAVS1 locus and a high level of expression was observed in undifferentiated iPSCs and in cell lineages derived from all three germ layers including haematopoietic cells, hepatocytes and neurons. We demonstrate efficient production of terminally differentiated macrophages from the SFCi55-ZsGreen iPSC line and show that they are indistinguishable from those generated from their parental SFCi55 iPSC line in terms of gene expression, cell surface marker expression and phagocytic activity. The high level of ZsGreen expression had no effect on the ability of macrophages to be activated to an M(LPS + IFNγ), M(IL10) or M(IL4) phenotype nor on their plasticity, assessed by their ability to switch from one phenotype to another. Thus, targeting of the AAVS1 locus in iPSCs allows for the production of fully functional, fluorescently tagged human macrophages that can be used for in vivo tracking in disease models. The strategy also provides a platform for the introduction of factors that are predicted to modulate and/or stabilize macrophage function.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.


Subject(s)
Cell Differentiation , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Induced Pluripotent Stem Cells/physiology , Macrophages/metabolism , Cell Lineage/physiology , Germ Layers/growth & development , Humans
20.
Stem Cell Reports ; 10(2): 461-476, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29396181

ABSTRACT

In mouse, although four Argonaute (AGO) proteins with partly overlapping functions in small-RNA pathways exist, only Ago2 deficiency causes embryonic lethality. To investigate the role of AGO2 during mouse early development, we generated Ago2-deficient mouse embryonic stem cells (mESCs) and performed a detailed characterization of their differentiation potential. Ago2 disruption caused a global reduction of microRNAs, which resulted in the misregulation of only a limited number of transcripts. We demonstrated, both in vivo and in vitro, that AGO2 is dispensable for the embryonic germ-layer formation. However, Ago2-deficient mESCs showed a specific defect during conversion into extra-embryonic endoderm cells. We proved that this defect is cell autonomous and can be rescued by both a catalytically active and an inactive Ago2, but not by Ago2 deprived of its RNA binding capacity or by Ago1 overexpression. Overall, our results suggest a role for AGO2 in stem cell differentiation.


Subject(s)
Argonaute Proteins/genetics , Cell Differentiation/genetics , Embryonic Development/genetics , Mouse Embryonic Stem Cells/cytology , Animals , Cell Line , Endoderm/cytology , Endoderm/growth & development , Gastrulation/genetics , Gene Expression Regulation, Developmental/genetics , Germ Layers/cytology , Germ Layers/growth & development , Mice , MicroRNAs/genetics
SELECTION OF CITATIONS
SEARCH DETAIL