Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
Add more filters

Publication year range
1.
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
2.
Cell ; 187(13): 3194-3219, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906095

ABSTRACT

Developing functional organs from stem cells remains a challenging goal in regenerative medicine. Existing methodologies, such as tissue engineering, bioprinting, and organoids, only offer partial solutions. This perspective focuses on two promising approaches emerging for engineering human organs from stem cells: stem cell-based embryo models and interspecies organogenesis. Both approaches exploit the premise of guiding stem cells to mimic natural development. We begin by summarizing what is known about early human development as a blueprint for recapitulating organogenesis in both embryo models and interspecies chimeras. The latest advances in both fields are discussed before highlighting the technological and knowledge gaps to be addressed before the goal of developing human organs could be achieved using the two approaches. We conclude by discussing challenges facing embryo modeling and interspecies organogenesis and outlining future prospects for advancing both fields toward the generation of human tissues and organs for basic research and translational applications.


Subject(s)
Chimera , Organogenesis , Animals , Humans , Chimera/embryology , Embryo Implantation , Embryo, Mammalian/cytology , Embryonic Development , Embryonic Stem Cells , Models, Biological , Organoids , Regenerative Medicine , Tissue Engineering/methods
3.
Cell ; 186(26): 5859-5875.e24, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38052213

ABSTRACT

Embryogenesis necessitates harmonious coordination between embryonic and extraembryonic tissues. Although stem cells of both embryonic and extraembryonic origins have been generated, they are grown in different culture conditions. In this study, utilizing a unified culture condition that activates the FGF, TGF-ß, and WNT pathways, we have successfully derived embryonic stem cells (FTW-ESCs), extraembryonic endoderm stem cells (FTW-XENs), and trophoblast stem cells (FTW-TSCs) from the three foundational tissues of mouse and cynomolgus monkey (Macaca fascicularis) blastocysts. This approach facilitates the co-culture of embryonic and extraembryonic stem cells, revealing a growth inhibition effect exerted by extraembryonic endoderm cells on pluripotent cells, partially through extracellular matrix signaling. Additionally, our cross-species analysis identified both shared and unique transcription factors and pathways regulating FTW-XENs. The embryonic and extraembryonic stem cell co-culture strategy offers promising avenues for developing more faithful embryo models and devising more developmentally pertinent differentiation protocols.


Subject(s)
Embryo, Mammalian , Embryonic Stem Cells , Animals , Coculture Techniques , Macaca fascicularis , Embryonic Stem Cells/metabolism , Cell Differentiation , Endoderm/metabolism , Cell Lineage
4.
Cell ; 185(18): 3290-3306.e25, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35988542

ABSTRACT

In vitro cultured stem cells with distinct developmental capacities can contribute to embryonic or extraembryonic tissues after microinjection into pre-implantation mammalian embryos. However, whether cultured stem cells can independently give rise to entire gastrulating embryo-like structures with embryonic and extraembryonic compartments remains unknown. Here, we adapt a recently established platform for prolonged ex utero growth of natural embryos to generate mouse post-gastrulation synthetic whole embryo models (sEmbryos), with both embryonic and extraembryonic compartments, starting solely from naive ESCs. This was achieved by co-aggregating non-transduced ESCs, with naive ESCs transiently expressing Cdx2 or Gata4 to promote their priming toward trophectoderm and primitive endoderm lineages, respectively. sEmbryos adequately accomplish gastrulation, advance through key developmental milestones, and develop organ progenitors within complex extraembryonic compartments similar to E8.5 stage mouse embryos. Our findings highlight the plastic potential of naive pluripotent cells to self-organize and functionally reconstitute and model the entire mammalian embryo beyond gastrulation.


Subject(s)
Embryonic Stem Cells , Gastrulation , Animals , Cell Differentiation/physiology , Embryo, Mammalian/physiology , Embryonic Development , Endoderm , Mammals , Mice
5.
Cell ; 184(11): 2843-2859.e20, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33991488

ABSTRACT

Since establishment of the first embryonic stem cells (ESCs), in vitro culture of totipotent cells functionally and molecularly comparable with in vivo blastomeres with embryonic and extraembryonic developmental potential has been a challenge. Here we report that spliceosomal repression in mouse ESCs drives a pluripotent-to-totipotent state transition. Using the splicing inhibitor pladienolide B, we achieve stable in vitro culture of totipotent ESCs comparable at molecular levels with 2- and 4-cell blastomeres, which we call totipotent blastomere-like cells (TBLCs). Mouse chimeric assays combined with single-cell RNA sequencing (scRNA-seq) demonstrate that TBLCs have a robust bidirectional developmental capability to generate multiple embryonic and extraembryonic cell lineages. Mechanically, spliceosomal repression causes widespread splicing inhibition of pluripotent genes, whereas totipotent genes, which contain few short introns, are efficiently spliced and transcriptionally activated. Our study provides a means for capturing and maintaining totipotent stem cells.


Subject(s)
Totipotent Stem Cells/cytology , Totipotent Stem Cells/metabolism , Animals , Blastomeres/cytology , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mouse Embryonic Stem Cells/cytology , Totipotent Stem Cells/physiology
6.
Cell ; 169(2): 243-257.e25, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388409

ABSTRACT

Of all known cultured stem cell types, pluripotent stem cells (PSCs) sit atop the landscape of developmental potency and are characterized by their ability to generate all cell types of an adult organism. However, PSCs show limited contribution to the extraembryonic placental tissues in vivo. Here, we show that a chemical cocktail enables the derivation of stem cells with unique functional and molecular features from mice and humans, designated as extended pluripotent stem (EPS) cells, which are capable of chimerizing both embryonic and extraembryonic tissues. Notably, a single mouse EPS cell shows widespread chimeric contribution to both embryonic and extraembryonic lineages in vivo and permits generating single-EPS-cell-derived mice by tetraploid complementation. Furthermore, human EPS cells exhibit interspecies chimeric competency in mouse conceptuses. Our findings constitute a first step toward capturing pluripotent stem cells with extraembryonic developmental potentials in culture and open new avenues for basic and translational research. VIDEO ABSTRACT.


Subject(s)
Cell Culture Techniques/methods , Pluripotent Stem Cells/cytology , Animals , Blastocyst/cytology , Cell Line , Chimera/metabolism , Dimethindene/pharmacology , Humans , Indicators and Reagents/chemistry , Mice , Minocycline/chemistry , Minocycline/pharmacology , Pluripotent Stem Cells/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism
7.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38752427

ABSTRACT

Bone morphogenic protein (BMP) signaling plays an essential and highly conserved role in embryo axial patterning in animal species. However, in mammalian embryos, which develop inside the mother, early development includes a preimplantation stage, which does not occur in externally developing embryos. During preimplantation, the epiblast is segregated from extra-embryonic lineages that enable implantation and development in utero. Yet, the requirement for BMP signaling is imprecisely defined in mouse early embryos. Here, we show that, in contrast to previous reports, BMP signaling (SMAD1/5/9 phosphorylation) is not detectable until implantation when it is detected in the primitive endoderm - an extra-embryonic lineage. Moreover, preimplantation development appears to be normal following deletion of maternal and zygotic Smad4, an essential effector of canonical BMP signaling. In fact, mice lacking maternal Smad4 are viable. Finally, we uncover a new requirement for zygotic Smad4 in epiblast scaling and cavitation immediately after implantation, via a mechanism involving FGFR/ERK attenuation. Altogether, our results demonstrate no role for BMP4/SMAD4 in the first lineage decisions during mouse development. Rather, multi-pathway signaling among embryonic and extra-embryonic cell types drives epiblast morphogenesis postimplantation.


Subject(s)
Embryo Implantation , Germ Layers , Morphogenesis , Signal Transduction , Smad4 Protein , Animals , Smad4 Protein/metabolism , Smad4 Protein/genetics , Germ Layers/metabolism , Embryo Implantation/genetics , Mice , Morphogenesis/genetics , Female , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Gene Expression Regulation, Developmental , Embryonic Development/genetics , Mice, Knockout , Embryo, Mammalian/metabolism , Endoderm/metabolism , Endoderm/embryology , Blastocyst/metabolism , Blastocyst/cytology
8.
EMBO J ; 41(7): e108747, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35266581

ABSTRACT

Mesoderm arises at gastrulation and contributes to both the mouse embryo proper and its extra-embryonic membranes. Two-photon live imaging of embryos bearing a keratin reporter allowed recording filament nucleation and elongation in the extra-embryonic region. Upon separation of amniotic and exocoelomic cavities, keratin 8 formed apical cables co-aligned across multiple cells in the amnion, allantois, and blood islands. An influence of substrate rigidity and composition on cell behavior and keratin content was observed in mesoderm explants. Embryos lacking all keratin filaments displayed a deflated extra-embryonic cavity, a narrow thick amnion, and a short allantois. Single-cell RNA sequencing of sorted mesoderm cells and micro-dissected amnion, chorion, and allantois, provided an atlas of transcriptomes with germ layer and regional information. It defined the cytoskeleton and adhesion expression profile of mesoderm-derived keratin 8-enriched cells lining the exocoelomic cavity. Those findings indicate a novel role for keratin filaments in the expansion of extra-embryonic structures and suggest mechanisms of mesoderm adaptation to the environment.


Subject(s)
Gastrulation , Mesoderm , Animals , Embryo, Mammalian , Extraembryonic Membranes , Keratins/genetics , Keratins/metabolism , Mesoderm/metabolism , Mice
9.
Proc Natl Acad Sci U S A ; 120(52): e2313200120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38113263

ABSTRACT

In female mice, the gene dosage from X chromosomes is adjusted by a process called X chromosome inactivation (XCI) that occurs in two steps. An imprinted form of XCI (iXCI) that silences the paternally inherited X chromosome (Xp) is initiated at the 2- to 4-cell stages. As extraembryonic cells including trophoblasts keep the Xp silenced, epiblast cells that give rise to the embryo proper reactivate the Xp and undergo a random form of XCI (rXCI) around implantation. Both iXCI and rXCI require the lncRNA Xist, which is expressed from the X to be inactivated. The X-linked E3 ubiquitin ligase Rlim (Rnf12) in conjunction with its target protein Rex1 (Zfp42), a critical repressor of Xist, have emerged as major regulators of iXCI. However, their roles in rXCI remain controversial. Investigating early mouse development, we show that the Rlim-Rex1 axis is active in pre-implantation embryos. Upon implantation Rex1 levels are downregulated independently of Rlim specifically in epiblast cells. These results provide a conceptual framework of how the functional dynamics between Rlim and Rex1 ensures regulation of iXCI but not rXCI in female mice.


Subject(s)
RNA, Long Noncoding , X Chromosome Inactivation , Animals , Female , Mice , Embryo, Mammalian/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , X Chromosome/genetics , X Chromosome/metabolism , X Chromosome Inactivation/genetics
10.
EMBO J ; 40(12): e106818, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33909924

ABSTRACT

Mouse embryonic stem cells (mESCs) are biased toward producing embryonic rather than extraembryonic endoderm fates. Here, we identify the mechanism of this barrier and report that the histone deacetylase Hdac3 and the transcriptional corepressor Dax1 cooperatively limit the lineage repertoire of mESCs by silencing an enhancer of the extraembryonic endoderm-specifying transcription factor Gata6. This restriction is opposed by the pluripotency transcription factors Nr5a2 and Esrrb, which promote cell type conversion. Perturbation of the barrier extends mESC potency and allows formation of 3D spheroids that mimic the spatial segregation of embryonic epiblast and extraembryonic endoderm in early embryos. Overall, this study shows that transcriptional repressors stabilize pluripotency by biasing the equilibrium between embryonic and extraembryonic lineages that is hardwired into the mESC transcriptional network.


Subject(s)
DAX-1 Orphan Nuclear Receptor , Histone Deacetylases , Mouse Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism , Female , GATA6 Transcription Factor/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Male , Mice , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
11.
Development ; 149(9)2022 05 01.
Article in English | MEDLINE | ID: mdl-35552395

ABSTRACT

In multicellular systems, cells communicate with adjacent cells to determine their positions and fates, an arrangement important for cellular development. Orientation of cell division, cell-cell interactions (i.e. attraction and repulsion) and geometric constraints are three major factors that define cell arrangement. In particular, geometric constraints are difficult to reveal in experiments, and the contribution of the local contour of the boundary has remained elusive. In this study, we developed a multicellular morphology model based on the phase-field method so that precise geometric constraints can be incorporated. Our application of the model to nematode embryos predicted that the amount of extra-embryonic space, the empty space within the eggshell that is not occupied by embryonic cells, affects cell arrangement in a manner dependent on the local contour and other factors. The prediction was validated experimentally by increasing the extra-embryonic space in the Caenorhabditis elegans embryo. Overall, our analyses characterized the roles of geometrical contributors, specifically the amount of extra-embryonic space and the local contour, on cell arrangements. These factors should be considered for multicellular systems.


Subject(s)
Caenorhabditis elegans Proteins , Nematoda , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cell Division , Embryo, Nonmammalian , Models, Biological
12.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-35993866

ABSTRACT

Embryogenesis is supported by dynamic loops of cellular interactions. Here, we create a partial mouse embryo model to elucidate the principles of epiblast (Epi) and extra-embryonic endoderm co-development (XEn). We trigger naive mouse embryonic stem cells to form a blastocyst-stage niche of Epi-like cells and XEn-like cells (3D, hydrogel free and serum free). Once established, these two lineages autonomously progress in minimal medium to form an inner pro-amniotic-like cavity surrounded by polarized Epi-like cells covered with visceral endoderm (VE)-like cells. The progression occurs through reciprocal inductions by which the Epi supports the primitive endoderm (PrE) to produce a basal lamina that subsequently regulates Epi polarization and/or cavitation, which, in return, channels the transcriptomic progression to VE. This VE then contributes to Epi bifurcation into anterior- and posterior-like states. Similarly, boosting the formation of PrE-like cells within blastoids supports developmental progression. We argue that self-organization can arise from lineage bifurcation followed by a pendulum of induction that propagates over time.


Subject(s)
Endoderm , Germ Layers , Animals , Blastocyst , Cell Differentiation , Cell Lineage/physiology , Embryo Implantation , Embryo, Mammalian , Mice
13.
Stem Cells ; 41(6): 578-591, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36648303

ABSTRACT

The interplay among mitogenic signaling pathways is crucial for proper embryogenesis. These pathways collaboratively act through intracellular master regulators to determine specific cell fates. Identifying the master regulators is critical to understanding embryogenesis and to developing new applications of pluripotent stem cells. In this report, we demonstrate protein kinase C (PKC) as an intrinsic master switch between embryonic and extraembryonic cell fates in the differentiation of human pluripotent stem cells (hPSCs). PKCs are essential to induce the extraembryonic lineage downstream of BMP4 and other mitogenic modulators. PKC-alpha (PKCα) suppresses BMP4-induced mesoderm differentiation, and PKC-delta (PKCδ) is required for trophoblast cell fate. PKC activation overrides mesoderm induction conditions and leads to extraembryonic fate. In contrast, PKC inhibition leads to ß-catenin (CTNNB1) activation, switching cell fate from trophoblast to mesoderm lineages. This study establishes PKC as a signaling boundary directing the segregation of extraembryonic and embryonic lineages. The manipulation of intrinsic PKC activity could greatly enhance cell differentiation under mitogenic regulation in stem cell applications.


Subject(s)
Pluripotent Stem Cells , Protein Kinase C , Humans , Protein Kinase C/metabolism , Embryonic Stem Cells/metabolism , Cell Differentiation , Pluripotent Stem Cells/metabolism , Mesoderm/metabolism , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 4/metabolism
14.
Gen Comp Endocrinol ; 347: 114439, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38158163

ABSTRACT

When females experience stress during reproduction, developing embryos can be exposed to elevated levels of glucocorticoids, which can permanently affect offspring development, physiology, and behavior. However, the embryo can regulate exposure to glucocorticoids. In placental species, the placenta regulates embryonic exposure to maternal steroids via metabolism. In a comparable way, recent evidence has shown the extraembryonic membranes of avian species also regulate embryonic exposure to a number of maternal steroids deposited in the yolk via metabolism early in development. However, despite the known effects of embryonic exposure to glucocorticoids, it is not yet understood how glucocorticoids are metabolized early in development. To address this knowledge gap, we injected corticosterone into freshly laid chicken (Gallus gallus) eggs and identified corticosterone metabolites, located metabolomic enzyme transcript expression, tracked metabolomic enzyme transcript expression during the first six days of development, and determined the effect of corticosterone and metabolites on embryonic survival. We found that yolk corticosterone was metabolized before day four of development into two metabolites: 5ß-corticosterone and 20ß-corticosterone. The enzymes, AKR1D1 and CBR1 respectively, were expressed in the extraembryonic membranes. Expression was dynamic during early development, peaking on day two of development. Finally, we found that corticosterone exposure is lethal to the embryos, yet exposure to the metabolites is not, suggesting that metabolism protects the embryo. Ultimately, we show that the extraembryonic membranes of avian species actively regulate their endocrine environment very early in development.


Subject(s)
Corticosterone , Placenta , Animals , Female , Pregnancy , Corticosterone/pharmacology , Corticosterone/metabolism , Placenta/metabolism , Egg Yolk/metabolism , Glucocorticoids/metabolism , Chickens/metabolism , Steroids/metabolism
15.
Stem Cells ; 40(3): 239-259, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35323987

ABSTRACT

Metabolism plays a crucial role for cell survival and function; however, recent evidence has implicated it in regulating embryonic development. In the embryo, the inner cell mass undergoes orchestrated cellular divisions resulting in the formation of pluripotent epiblast stem cells and primitive endoderm cells. However, both lineages can be captured in vitro as embryonic stem (ES) cells and extraembryonic endoderm (XEN) cells. Concomitantly, changes in the metabolic profile occurs during development, and are well documented in the embryonic lineages. However, a comprehensive multi-omic analysis of these features in XEN cells remains lacking. We observed that mouse XEN cells exhibited high sensitivity to glycolytic inhibition in addition to maintaining elevated intra- and extracellular lactate levels in vitro. Extraembryonic endoderm cells maintain high lactate levels by increased LDHA activity, and re-routing pyruvate away from the mitochondria resulting in reduced mitochondrial activity due to disruptions in electron transport chain stoichiometry. Importantly, exogenous lactate supplementation or promoting intracellular lactate accumulation enhances XEN differentiation in vitro. These results highlight how lactate contributes to XEN differentiation in vitro and may serve to enhance reprogramming efficiency of cells used for regenerative medicine.


Subject(s)
Endoderm , Lactic Acid , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Lactic Acid/metabolism , Mice , Mouse Embryonic Stem Cells
16.
J Perinat Med ; 51(7): 891-895, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37067543

ABSTRACT

OBJECTIVES: In-vivo studies of the bioavailability of major components of the tumor necrosis factor alpha (TNFα) biosystem inside the gestational sac during embryogenesis have not been reported. We sought to determine the concentration of TNFα, soluble (s) TNFα receptors (sTNFR1, sTNFR2), and RANTES in the primate extraembryonic celomic fluid (ECF). METHODS: A validated timed-pregnant baboon animal model (N: 10) for experimental research in pregnancy was used to collect paired maternal blood and ECF samples in ongoing pregnancies. The concentrations (pg/dL) of TNFα, sTNFR1, sTNFR2, and RANTES were then determined by ELISA immunoassays. RESULTS: All animals delivered at term healthy newborns. The differential concentration of TNFα, sTNFR1, sTNFR2, and RANTES between the maternal plasma and the ECF could be determined with ratios for TNFα (5.4), sTNFR2 (1.85) and RANTES (3.59) that contrasted with that of sTNFR1 (0.07), which favored the gestational sac compartment. No significant correlations were noted between maternal plasma and ECF TNFR1, sTNFR2 and RANTES. There was a trend for a correlation between TNFα in maternal plasma and ECF (R=0.74; p=0.07). CONCLUSIONS: We report the physiological concentrations of TNFα, sTNFR1, sTNFR2, and RANTES in extraembryonic celomic fluid during embryogenesis in primates.


Subject(s)
Receptors, Tumor Necrosis Factor, Type II , Tumor Necrosis Factor-alpha , Female , Pregnancy , Humans , Chemokine CCL5 , Biological Availability , Gestational Sac/metabolism , T-Lymphocytes/metabolism
17.
Genes Dev ; 29(12): 1239-55, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26109048

ABSTRACT

Transcription factor-mediated reprograming is a powerful method to study cell fate changes. In this study, we demonstrate that the transcription factor Gata6 can initiate reprograming of multiple cell types to induced extraembryonic endoderm stem (iXEN) cells. Intriguingly, Gata6 is sufficient to drive iXEN cells from mouse pluripotent cells and differentiated neural cells. Furthermore, GATA6 induction in human embryonic stem (hES) cells also down-regulates pluripotency gene expression and up-regulates extraembryonic endoderm (ExEn) genes, revealing a conserved function in mediating this cell fate switch. Profiling transcriptional changes following Gata6 induction in mES cells reveals step-wise pluripotency factor disengagement, with initial repression of Nanog and Esrrb, then Sox2, and finally Oct4, alongside step-wise activation of ExEn genes. Chromatin immunoprecipitation and subsequent high-throughput sequencing analysis shows Gata6 enrichment near pluripotency and endoderm genes, suggesting that Gata6 functions as both a direct repressor and activator. Together, this demonstrates that Gata6 is a versatile and potent reprograming factor that can act alone to drive a cell fate switch from diverse cell types.


Subject(s)
Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Endoderm/cytology , GATA6 Transcription Factor/metabolism , Pluripotent Stem Cells/cytology , Animals , Binding Sites , Cell Differentiation , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Binding , Signal Transduction
18.
Int J Mol Sci ; 24(14)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37511125

ABSTRACT

In vitro modeling of human peri-gastrulation development is a valuable tool for understanding embryogenetic mechanisms. The extraembryonic mesoderm (ExM) is crucial in supporting embryonic development by forming tissues such as the yolk sac, allantois, and chorionic villi. However, the origin of human ExM remains only partially understood. While evidence suggests a primitive endoderm (PrE) origin based on morphological findings, current in vitro models use epiblast-like cells. To address this gap, we developed a protocol to generate ExM-like cells from PrE-like cell line called naïve extraembryonic endoderm (nEnd). We identified the ExM-like cells by specific markers (LUM and ANXA1). Moreover, these in vitro-produced ExM cells displayed angiogenic potential on a soft matrix, mirroring their physiological role in vasculogenesis. By integrating single-cell RNA sequencing (scRNAseq) data, we found that the ExM-like cells clustered with the LUM/ANXA1-rich cell populations of the gastrulating embryo, indicating similarity between in vitro and ex utero cell populations. This study confirms the derivation of ExM from PrE and establishes a cell culture system that can be utilized to investigate ExM during human peri-gastrulation development, both in monolayer cultures and more complex models.


Subject(s)
Endoderm , Mesoderm , Pregnancy , Female , Humans , Endoderm/metabolism , Mesoderm/metabolism , Cell Differentiation/physiology , Embryo, Mammalian , Embryonic Development
19.
Dev Biol ; 470: 84-94, 2021 02.
Article in English | MEDLINE | ID: mdl-33217407

ABSTRACT

At implantation, the mouse embryo undergoes a critical transformation which requires the precise spatiotemporal control of signalling pathways necessary for morphogenesis and developmental progression. The role played by such signalling pathways during this transition are largely unexplored, due to the inaccessibility of the embryo during the implantation when it becomes engulfed by uterine tissues. Genetic studies demonstrate that mutant embryos for BMPs die around gastrulation. Here we have aimed to dissect the role of BMPs during pre-to post-implantation transition by using a protocol permitting the development of the embryo beyond implantation stages in vitro and using stem cells to mimic post-implantation tissue organisation. By assessing both the canonical and non-canonical mechanisms of BMP, we show that the loss of canonical BMP activity compromises the extra-embryonic ectoderm development. Our analyses demonstrate that BMP signalling maintains stem cell populations within both embryonic/extra-embryonic tissues during pre-to post-implantation development. These results may provide insight into the role played by BMP signalling in controlling early embryogenesis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Ectoderm/embryology , Embryo Implantation , Embryonic Development , Signal Transduction , Animals , Cell Death , Cell Lineage , Ectoderm/cytology , Embryo Culture Techniques , Embryonic Stem Cells/cytology , Germ Layers/cytology , Germ Layers/embryology , Mice , Morphogenesis , Trophoblasts/cytology
20.
Biol Reprod ; 106(3): 540-550, 2022 03 19.
Article in English | MEDLINE | ID: mdl-34791028

ABSTRACT

The Bone Morphogenetic Protein (BMP) pathway is involved in numerous developmental processes, including cell growth, apoptosis, and differentiation. In mouse embryogenesis, BMP signaling is a well-known morphogen for both mesoderm induction and germ cell development. Recent evidence points to a potential role in development of the extraembryonic compartment, including trophectoderm-derived tissues. In this study, we investigated the effect of BMP signaling in both mouse and human trophoblast stem cells (TSC) in vitro, evaluating the expression and activation of the BMP signaling response machinery, and the effect of BMP signaling manipulation during TSC maintenance and differentiation. Both mouse trophoblast stem cells (mTSC) and human trophoblast stem cells (hTSC) expressed various BMP ligands and the receptors BMPR1A and BMPR2, necessary for BMP response, and displayed maximal active BMP signaling when undifferentiated. We also observed a conserved modulatory role of BMP signaling during trophoblast differentiation, whereby maintenance of active BMP signaling blunted differentiation of TSC in both species. Conversely, the effect of BMP signaling on the undifferentiated state of TSC appeared to be species-specific, with SMAD-independent signaling important in maintenance of mTSC, and a more subtle role for both SMAD-dependent and -independent BMP signaling in hTSC. Altogether, these data establish an autocrine role for the BMP pathway in the trophoblast compartment. As specification and correct differentiation of the extraembryonic compartment are fundamental for implantation and early placental development, insights on the role of the BMP signaling in early development might prove useful in the setting of in vitro fertilization as well as targeting trophoblast-associated placental dysfunction.


Subject(s)
Placenta , Trophoblasts , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Female , Humans , Mice , Placenta/metabolism , Pregnancy , Stem Cells/metabolism , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL