Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Publication year range
1.
Cell ; 165(4): 1012-26, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27062923

ABSTRACT

Mouse studies have been instrumental in forming our current understanding of early cell-lineage decisions; however, similar insights into the early human development are severely limited. Here, we present a comprehensive transcriptional map of human embryo development, including the sequenced transcriptomes of 1,529 individual cells from 88 human preimplantation embryos. These data show that cells undergo an intermediate state of co-expression of lineage-specific genes, followed by a concurrent establishment of the trophectoderm, epiblast, and primitive endoderm lineages, which coincide with blastocyst formation. Female cells of all three lineages achieve dosage compensation of X chromosome RNA levels prior to implantation. However, in contrast to the mouse, XIST is transcribed from both alleles throughout the progression of this expression dampening, and X chromosome genes maintain biallelic expression while dosage compensation proceeds. We envision broad utility of this transcriptional atlas in future studies on human development as well as in stem cell research.


Subject(s)
Blastocyst/metabolism , Chromosomes, Human, X , Single-Cell Analysis , Blastocyst Inner Cell Mass/metabolism , Dosage Compensation, Genetic , Female , Humans , Male , RNA, Long Noncoding/genetics , Sequence Analysis, RNA , Sex Characteristics , Transcriptome
2.
Nature ; 622(7983): 562-573, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37673118

ABSTRACT

The ability to study human post-implantation development remains limited owing to ethical and technical challenges associated with intrauterine development after implantation1. Embryo-like models with spatially organized morphogenesis and structure of all defining embryonic and extra-embryonic tissues of the post-implantation human conceptus (that is, the embryonic disc, the bilaminar disc, the yolk sac, the chorionic sac and the surrounding trophoblast layer) remain lacking1,2. Mouse naive embryonic stem cells have recently been shown to give rise to embryonic and extra-embryonic stem cells capable of self-assembling into post-gastrulation structured stem-cell-based embryo models with spatially organized morphogenesis (called SEMs)3. Here we extend those findings to humans using only genetically unmodified human naive embryonic stem cells (cultured in human enhanced naive stem cell medium conditions)4. Such human fully integrated and complete SEMs recapitulate the organization of nearly all known lineages and compartments of post-implantation human embryos, including the epiblast, the hypoblast, the extra-embryonic mesoderm and the trophoblast layer surrounding the latter compartments. These human complete SEMs demonstrated developmental growth dynamics that resemble key hallmarks of post-implantation stage embryogenesis up to 13-14 days after fertilization (Carnegie stage 6a). These include embryonic disc and bilaminar disc formation, epiblast lumenogenesis, polarized amniogenesis, anterior-posterior symmetry breaking, primordial germ-cell specification, polarized yolk sac with visceral and parietal endoderm formation, extra-embryonic mesoderm expansion that defines a chorionic cavity and a connecting stalk, and a trophoblast-surrounding compartment demonstrating syncytium and lacunae formation. This SEM platform will probably enable the experimental investigation of previously inaccessible windows of human early post implantation up to peri-gastrulation development.


Subject(s)
Embryo Implantation , Embryo, Mammalian , Embryonic Development , Human Embryonic Stem Cells , Humans , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Fertilization , Gastrulation , Germ Layers/cytology , Germ Layers/embryology , Human Embryonic Stem Cells/cytology , Trophoblasts/cytology , Yolk Sac/cytology , Yolk Sac/embryology , Giant Cells/cytology
4.
Genome Res ; 2022 Aug 10.
Article in English | MEDLINE | ID: mdl-35948369

ABSTRACT

The preconceptual, intrauterine, and early life environments can have a profound and long-lasting impact on the developmental trajectories and health outcomes of the offspring. Given the relatively low success rates of assisted reproductive technologies (ART; ∼25%), additives and adjuvants, such as glucocorticoids, are used to improve the success rate. Considering the dynamic developmental events that occur during this window, these exposures may alter blastocyst formation at a molecular level, and as such, affect not only the viability of the embryo and the ability of the blastocyst to implant, but also the developmental trajectory of the first three cell lineages, ultimately influencing the physiology of the embryo. In this study, we present a comprehensive single-cell transcriptome, methylome, and small RNA atlas in the day 7 human embryo. We show that, despite no change in morphology and developmental features, preimplantation glucocorticoid exposure reprograms the molecular profile of the TE lineage, and these changes are associated with an altered metabolic and inflammatory response. Our data also suggest that glucocorticoids can precociously mature the TE sublineages, supported by the presence of extravillous trophoblast markers in the polar sublineage and presence of X Chromosome dosage compensation. Further, we have elucidated that epigenetic regulation-DNA methylation and microRNAs (miRNAs)-likely underlies the transcriptional changes observed. This study suggests that exposures to exogenous compounds during preimplantation may unintentionally reprogram the human embryo, possibly leading to suboptimal development and longer-term health outcomes.

5.
Stem Cells ; 41(2): 105-110, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36153824

ABSTRACT

Ovaries are central to development, fertility, and reproduction of women. A particularly interesting feature of ovaries is their accelerated aging compared to other tissues, leading to loss of function far before other organs senesce. The limited pool of ovarian follicles is generated before birth and once exhausted, menopause will inevitably commence around the age of 50 years marking the end of fertility. Yet, there are reports suggesting the presence of germline stem cells and neo-oogenesis in adult human ovaries. These observations have fueled a long debate, created experimental fertility treatments, and opened business opportunities. Our recent analysis of cell types in the ovarian cortex of women of fertile age could not find evidence of germline stem cells. Like before, our work has been met with critique suggesting methodological shortcomings. We agree that excellence starts with methods and welcome discussion on the pros and cons of different protocols. In this commentary, we discuss the recent re-interpretation of our work.


Subject(s)
Oogenesis , Ovary , Adult , Female , Humans , Middle Aged , Oogenesis/physiology , Ovarian Follicle , Germ Cells , Stem Cells/metabolism
6.
Reproduction ; 165(4): R103-R116, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36700623

ABSTRACT

In brief: Human embryogenesis still remains largely unexplored. This review helps identify some of our current gaps in knowledge pertaining to preimplantation development, which may have implications for understanding fundamental aspects of human development, assisted reproductive technologies, and stem cell biology. Abstract: Preimplantation development is arguably one of the most critical stages of embryogenesis. Beginning with the formation of the totipotent zygote post-fertilization, a series of cell divisions, and a complex coordination of physical cues, molecular signals and changes in gene expression lead to the formation of the blastocyst, a structure capable of implanting into the uterine wall. The blastocyst is composed of more specified cellular lineages, which will give rise to every tissue of the developing organism as well as the extra-embryonic lineages which support fetal growth. While the mouse has been used as a model to understand the events of preimplantation development for decades, in recent years, an expanding body of work has been conducted using the human embryo. These studies have identified some crucial species differences, particularly in the transcriptional and spatio-temporal expression of lineage markers and responses to cell signaling perturbations. This review compares recent findings on preimplantation development in mouse and human, with a focus on the specification of the first cellular lineages. Highlighting differences and noting mechanisms that require further examination in the human embryo is of critical importance for both the accurate translation of results from the mouse model and our overall understanding of mammalian development. We further highlight the latest advancement in reproductive research, the development of the 3D stem cell-based models known as 'blastoids'. The knowledge discussed in this review has major clinical implications for assisted reproductive technologies such as in vitro fertilization and for applications in stem cell biology.


Subject(s)
Embryonic Development , Zygote , Animals , Humans , Embryonic Development/genetics , Cell Lineage , Zygote/metabolism , Embryo, Mammalian , Blastocyst , Gene Expression Regulation, Developmental , Mammals/genetics
7.
Yale J Biol Med ; 96(4): 481-494, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38161584

ABSTRACT

Proper formation of the blastocyst, including the specification of the first embryonic cellular lineages, is required to ensure healthy embryo development and can significantly impact the success of assisted reproductive technologies (ARTs). However, the regulatory role of microRNAs in early development, particularly in the context of preimplantation lineage specification, remains largely unknown. Taking a cross-species approach, this review aims to summarize the expression dynamics and functional significance of microRNAs in the differentiation and maintenance of lineage identity in both the mouse and the human. Findings are consolidated from studies conducted using in vitro embryonic stem cell models representing the epiblast, trophectoderm, and primitive endoderm lineages (modeled by naïve embryonic stem cells, trophoblast stem cells, and extraembryonic endoderm stem cells, respectively) to provide insight on what may be occurring in the embryo. Additionally, studies directly conducted in both mouse and human embryos are discussed, emphasizing similarities to the stem cell models and the gaps in our understanding, which will hopefully lead to further investigation of these areas. By unraveling the intricate mechanisms by which microRNAs regulate the specification and maintenance of cellular lineages in the blastocyst, we can leverage this knowledge to further optimize stem cell-based models such as the blastoids, enhance embryo competence, and develop methods of non-invasive embryo selection, which can potentially increase the success rates of assisted reproductive technologies and improve the experiences of those receiving fertility treatments.


Subject(s)
MicroRNAs , Animals , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Lineage/genetics , Embryonic Development/genetics , Embryo, Mammalian/metabolism , Cell Differentiation/genetics
8.
Genome Res ; 26(10): 1342-1354, 2016 10.
Article in English | MEDLINE | ID: mdl-27486082

ABSTRACT

Pluripotency, differentiation, and X Chromosome inactivation (XCI) are key aspects of embryonic development. However, the underlying relationship and mechanisms among these processes remain unclear. Here, we systematically dissected these features along developmental progression using mouse embryonic stem cells (mESCs) and single-cell RNA sequencing with allelic resolution. We found that mESCs grown in a ground state 2i condition displayed transcriptomic profiles diffused from preimplantation mouse embryonic cells, whereas EpiStem cells closely resembled the post-implantation epiblast. Sex-related gene expression varied greatly across distinct developmental states. We also identified novel markers that were highly enriched in each developmental state. Moreover, we revealed that several novel pathways, including PluriNetWork and Focal Adhesion, were responsible for the delayed progression of female EpiStem cells. Importantly, we "digitalized" XCI progression using allelic expression of active and inactive X Chromosomes and surprisingly found that XCI states exhibited profound variability in each developmental state, including the 2i condition. XCI progression was not tightly synchronized with loss of pluripotency and increase of differentiation at the single-cell level, although these processes were globally correlated. In addition, highly expressed genes, including core pluripotency factors, were in general biallelically expressed. Taken together, our study sheds light on the dynamics of XCI progression and the asynchronicity between pluripotency, differentiation, and XCI.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , X Chromosome Inactivation , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells/metabolism , Single-Cell Analysis , Transcriptome
9.
Development ; 143(19): 3459-3469, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27578796

ABSTRACT

Leucine twenty homeobox (LEUTX) is a paired (PRD)-like homeobox gene that is expressed almost exclusively in human embryos during preimplantation development. We previously identified a novel transcription start site for the predicted human LEUTX gene based on the transcriptional analysis of human preimplantation embryos. The novel variant encodes a protein with a complete homeodomain. Here, we provide a detailed description of the molecular cloning of the complete homeodomain-containing LEUTX Using a human embryonic stem cell overexpression model we show that the complete homeodomain isoform is functional and sufficient to activate the transcription of a large proportion of the genes that are upregulated in human embryo genome activation (EGA), whereas the previously predicted partial homeodomain isoform is largely inactive. Another PRD-like transcription factor, DPRX, is then upregulated as a powerful repressor of transcription. We propose a two-stage model of human EGA in which LEUTX acts as a transcriptional activator at the 4-cell stage, and DPRX as a balancing repressor at the 8-cell stage. We conclude that LEUTX is a candidate regulator of human EGA.


Subject(s)
Blastocyst/metabolism , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Protein Isoforms/metabolism , Animals , Cell Line , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Mice , Polymerase Chain Reaction , Protein Isoforms/genetics
11.
Cell Physiol Biochem ; 41(3): 1044-1050, 2017.
Article in English | MEDLINE | ID: mdl-28222448

ABSTRACT

BACKGROUND/AIMS: Viral infection during pregnancy is known to affect the fetal brain. The toll-like receptor (TLR)-3 is a pattern recognition receptor activated by viruses known to elicit adverse fetal neurological outcomes. The P-glycoprotein (P-gp) efflux transporter protects the developing fetus by limiting the transfer of substrates across both the placenta and the fetal blood-brain barrier (BBB). As such, inhibition of P-gp at these blood-barrier sites may result in increased exposure of the developing fetus to environmental toxins and xenobiotics present in the maternal circulation. We hypothesized that viral exposure during pregnancy would impair P-gp function in the placenta and in the developing BBB. Here we investigated whether the TLR-3 ligand, polyinosinic:polycytidylic acid (PolyI:C), increased accumulation of one P-gp substrate in the fetus and in the developing fetal brain. METHODS: Pregnant C57BL/6 mice (GD15.5) were injected (i.p.) with PolyI:C (5 mg/kg or 10 mg/kg) or vehicle (saline). [3H]digoxin (P-gp substrate) was injected (i.v.) 3 or 23h post-treatment and animals were euthanized 1h later. Maternal plasma, 'fetal-units' (fetal membranes, amniotic fluid and whole fetus), and fetal brains were collected. RESULTS: PolyI:C exposure (4h) significantly elevated maternal plasma IL-6 (P<0.001) and increased [3H]digoxin accumulation in the fetal brain (P<0.05). In contrast, 24h after PolyI:C exposure, no effect on IL-6 or fetal brain accumulation of P-gp substrate was observed. CONCLUSION: Viral infection modeled by PolyI:C causes acute increases in fetal brain accumulation of P-gp substrates and by doing so, may increase fetal brain exposure to xenobiotics and environmental toxins present in the maternal circulation.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Fetus/metabolism , Poly I-C/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Biological Transport , Digoxin/metabolism , Disease Models, Animal , Female , Fetus/blood supply , Gene Expression , Interleukin-6/biosynthesis , Interleukin-6/genetics , Maternal-Fetal Exchange , Mice , Mice, Inbred C57BL , Placenta/metabolism , Pregnancy , Tritium , Virus Diseases/metabolism
12.
Biol Reprod ; 90(2): 43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24451982

ABSTRACT

Synthetic glucocorticoids (sGCs) are commonly prescribed for the management of inflammatory and endocrine disorders. However, nothing is known regarding the effects of sGC on adult germline methylome and whether these effects can be transmitted to the next generation. We hypothesized that administration of sGC to adult male mice alters DNA methylation in mature sperm and modifies the transcription and methylation of steroid receptors in male F1 offspring. Adult C57BL/6 males (n = 10/group) were injected on five consecutive days with 1 mg/kg sGC (i.e., dexamethasone) or vehicle and euthanized 35 or 60 days after initial treatment or bred with control females (60 days postinitial treatment; n = 5/group). A significant increase in global non-CpG methylation was observed in F0 sperm 60 days following sGC treatment. In the hippocampus and kidney of Postnatal Day 50 (PND50) and PND240 male offspring derived from fathers exposed to sGC, significant differences in mineralocorticoid receptor (Nr3c2; Mr), estrogen alpha receptor (Nr3a1; Ers1), and glucocorticoid receptor (Nr3c1; Gr) expression were observed. Furthermore, significant demethylation in regulatory regions of Mr, Gr, and Esr1 was observed in the PND50 kidney derived from fathers exposed to sGC. This is the first demonstration that paternal pharmacological exposure to sGC can alter the expression and DNA methylation of nuclear steroid receptors in brain and somatic tissues of offspring. These findings provide proof of principle that adult male exposure to sGC can affect DNA methylation and gene expression in offspring, indicating the possibility that adult experiences that evoke increases in endogenous glucocorticoid (i.e., stress) might have similar effects.


Subject(s)
DNA Methylation/drug effects , Glucocorticoids/pharmacology , Hippocampus/metabolism , Prenatal Exposure Delayed Effects/genetics , Receptors, Steroid/genetics , Transcription, Genetic/drug effects , Adrenal Cortex/drug effects , Adrenal Cortex/physiology , Animals , Female , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Paternal Exposure , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Receptors, Steroid/metabolism , Spermatozoa/drug effects , Spermatozoa/physiology
13.
Methods Mol Biol ; 2767: 275-292, 2024.
Article in English | MEDLINE | ID: mdl-37284942

ABSTRACT

Studying various animal models is important for comparative biology and to better understand evolutionary development. Furthermore, when aiming to translate findings to human development, it is crucial to select an appropriate animal model that closely resembles the specific aspect of development under study. The guinea pig is highlighted as a useful platform for reproductive studies due to similarities in in utero development and general physiology with the human. This chapter outlines the methods required for guinea pig mating and collection of embryos for in vitro culture and molecular characterization. Specifically, this chapter provides detailed guidance on monitoring the estrus cycle to determine the mating time, performing a vaginal flush and smear to confirm successful mating, performing euthanasia of the guinea pig, and flushing in vivo embryos. Once collected, the embryos can be utilized for numerous downstream applications. Here we will cover embryo culturing and processing embryos for immunofluorescence.


Subject(s)
Estrus , Reproduction , Humans , Female , Guinea Pigs , Animals , Vagina , Blastocyst , Fluorescent Antibody Technique
14.
Methods Mol Biol ; 2767: 293-305, 2024.
Article in English | MEDLINE | ID: mdl-37418145

ABSTRACT

Single-cell genomics allow the characterization and quantification of molecular heterogeneity from a wide variety of tissues. Here, we describe the manual dissociation and collection of single cells, a method adapted for the characterization of precious small tissues like preimplantation embryos. We also describe the acquisition of mouse embryos by flushing of the oviducts. The cells can then be used in multiple sequencing protocols, for example, Smart-seq2, Smart-seq3, smallseq, and scBSseq.


Subject(s)
Blastocyst , Embryo, Mammalian , Mice , Animals , Genomics , Mammals
15.
Methods Mol Biol ; 2767: 307-320, 2024.
Article in English | MEDLINE | ID: mdl-37261674

ABSTRACT

Fluorescence in situ hybridization (FISH) provides a valuable tool for studying the spatial localization of and expression level of genes and cell function in diverse biological contexts. In this chapter, we describe a protocol for the simultaneous detection of RNA (including single-molecule (sm)RNA) and DNA in mammalian embryos using FISH. RNA FISH is a technique that enables the detection and visualization of specific RNA molecules within cells. With advancements in technology, the sensitivity and specificity of RNA FISH has been improved to allow the detection of individual mRNA molecules. Both RNA and smRNA are detected using a set of fluorescent-labeled probes, which are complementary to a specific nucleotide sequence corresponding to the gene of interest. These probes hybridize to the target RNA molecules, enabling the simultaneous detection of multiple RNAs within the same cell or tissue. DNA FISH is performed using probes directed at the DNA sequence to detect the genome region of interest. In this chapter, we provide a protocol to process mammalian embryos for FISH with probe examples specifically for studying X-Chromosome activity. By utilizing other probe designs, this protocol can be adapted for the visualization and quantification of other genes and chromosomal regions of interest.


Subject(s)
Embryo, Mammalian , RNA , Animals , RNA/genetics , In Situ Hybridization, Fluorescence/methods , RNA, Messenger/genetics , DNA/genetics , Mammals/genetics
16.
Methods Mol Biol ; 2767: 189-212, 2024.
Article in English | MEDLINE | ID: mdl-37278916

ABSTRACT

The development of single-cell multiomics has provided the ability to systematically investigate cellular diversity and heterogeneity in different biological systems via comprehensive delineations of individual cellular states. Single-cell RNA sequencing in particular has served as a powerful tool to the study of the molecular circuitries underlying preimplantation embryonic development in both the mouse and human. Here we describe a method to elucidate the cellular dynamics of the embryo further by performing both single-cell RNA sequencing (Smart-Seq2) and single-cell small non-coding RNA sequencing (Small-Seq) on the same individual embryonic cell.


Subject(s)
RNA, Small Untranslated , Humans , Pregnancy , Female , Mice , Animals , Blastocyst , Embryo, Mammalian , Embryonic Development/genetics , RNA, Messenger
17.
ACS Cent Sci ; 10(1): 184-198, 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38292600

ABSTRACT

Nonhealing skin wounds are a problematic complication associated with diabetes. Therapeutic gases delivered by biomaterials have demonstrated powerful wound healing capabilities. However, the cellular responses and heterogeneity in the skin regeneration process after gas therapy remain elusive. Here, we display the benefit of the carbon monoxide (CO)-releasing hyaluronan hydrogel (CO@HAG) in promoting diabetic wound healing and investigate the cellular responses through single-cell transcriptomic analysis. The presented CO@HAG demonstrates wound microenvironment responsive gas releasing properties and accelerates the diabetic wound healing process in vivo. It is found that a new cluster of Cxcl14+ fibroblasts with progenitor property is accumulated in the CO@HAG-treated wound. This cluster of Cxcl14+ fibroblasts is yet unreported in the skin regeneration process. CO@HAG-treated wound macrophages feature a decrease in pro-inflammatory property, while their anti-inflammatory property increases. Moreover, the TGF-ß signal between the pro-inflammatory (M1) macrophage and the Cxcl14+ fibroblast in the CO@HAG-treated wound is attenuated based on cell-cell interaction analysis. Our study provides a useful hydrogel-mediated gas therapy method for diabetic wounds and new insights into cellular events in the skin regeneration process after gas-releasing biomaterials therapy.

18.
Nat Commun ; 15(1): 167, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167821

ABSTRACT

Primordial germ cells (PGCs) are the embryonic precursors of sperm and eggs. They transmit genetic and epigenetic information across generations. Given the prominent role of germline defects in diseases such as infertility, detailed understanding of human PGC (hPGC) development has important implications in reproductive medicine and studying human evolution. Yet, hPGC specification remains an elusive process. Here, we report the induction of hPGC-like cells (hPGCLCs) in a bioengineered human pluripotent stem cell (hPSC) culture that mimics peri-implantation human development. In this culture, amniotic ectoderm-like cells (AMLCs), derived from hPSCs, induce hPGCLC specification from hPSCs through paracrine signaling downstream of ISL1. Our data further show functional roles of NODAL, WNT, and BMP signaling in hPGCLC induction. hPGCLCs are successfully derived from eight non-obstructive azoospermia (NOA) participant-derived hPSC lines using this biomimetic platform, demonstrating its promise for screening applications.


Subject(s)
Pluripotent Stem Cells , Semen , Humans , Male , Germ Cells/metabolism , Cell Line , Signal Transduction , Cell Differentiation
19.
Cell Rep ; 42(1): 112021, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36848234

ABSTRACT

Different formative pluripotent stem cells harboring similar functional properties have been recently established to be lineage neutral and germline competent yet have distinct molecular identities. Here, we show that WNT/ß-catenin signaling activation sustains transient mouse epiblast-like cells as epiblast-like stem cells (EpiLSCs). EpiLSCs display metastable formative pluripotency with bivalent cellular energy metabolism and unique transcriptomic features and chromatin accessibility. We develop single-cell stage label transfer (scSTALT) to study the formative pluripotency continuum and reveal that EpiLSCs recapitulate a unique developmental period in vivo, filling the gap of the formative pluripotency continuum between other published formative stem cells. WNT/ß-catenin signaling activation counteracts differentiation effects of activin A and bFGF by preventing complete dissolution of naive pluripotency regulatory network. Moreover, EpiLSCs have direct competence toward germline specification, which is further matured by an FGF receptor inhibitor. Our EpiLSCs can serve as an in vitro model for mimicking and studying early post-implantation development and pluripotency transition.


Subject(s)
Pluripotent Stem Cells , Wnt Signaling Pathway , Animals , Mice , beta Catenin , Cell Differentiation , Germ Cells , Germ Layers
20.
Nat Cell Biol ; 24(6): 845-857, 2022 06.
Article in English | MEDLINE | ID: mdl-35637409

ABSTRACT

The first lineage choice in human embryo development separates trophectoderm from the inner cell mass. Naïve human embryonic stem cells are derived from the inner cell mass and offer possibilities to explore how lineage integrity is maintained. Here, we discover that polycomb repressive complex 2 (PRC2) maintains naïve pluripotency and restricts differentiation to trophectoderm and mesoderm lineages. Through quantitative epigenome profiling, we found that a broad gain of histone H3 lysine 27 trimethylation (H3K27me3) is a distinct feature of naïve pluripotency. We define shared and naïve-specific bivalent promoters featuring PRC2-mediated H3K27me3 concomitant with H3K4me3. Naïve bivalency maintains key trophectoderm and mesoderm transcription factors in a transcriptionally poised state. Inhibition of PRC2 forces naïve human embryonic stem cells into an 'activated' state, characterized by co-expression of pluripotency and lineage-specific transcription factors, followed by differentiation into either trophectoderm or mesoderm lineages. In summary, PRC2-mediated repression provides a highly adaptive mechanism to restrict lineage potential during early human development.


Subject(s)
Human Embryonic Stem Cells , Polycomb Repressive Complex 2 , Cell Differentiation/genetics , Embryonic Development , Histones/genetics , Human Embryonic Stem Cells/metabolism , Humans , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL