Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 164(4): 668-80, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26871632

ABSTRACT

Mouse embryonic stem cells (ESCs) are maintained in a naive ground state of pluripotency in the presence of MEK and GSK3 inhibitors. Here, we show that ground-state ESCs express low Myc levels. Deletion of both c-myc and N-myc (dKO) or pharmacological inhibition of Myc activity strongly decreases transcription, splicing, and protein synthesis, leading to proliferation arrest. This process is reversible and occurs without affecting pluripotency, suggesting that Myc-depleted stem cells enter a state of dormancy similar to embryonic diapause. Indeed, c-Myc is depleted in diapaused blastocysts, and the differential expression signatures of dKO ESCs and diapaused epiblasts are remarkably similar. Following Myc inhibition, pre-implantation blastocysts enter biosynthetic dormancy but can progress through their normal developmental program after transfer into pseudo-pregnant recipients. Our study shows that Myc controls the biosynthetic machinery of stem cells without affecting their potency, thus regulating their entry and exit from the dormant state.


Subject(s)
Embryonic Stem Cells/cytology , Genes, myc , Proto-Oncogene Proteins c-myc/genetics , Animals , Blastocyst/metabolism , Cell Proliferation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Female , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL
2.
Nature ; 609(7925): 136-143, 2022 09.
Article in English | MEDLINE | ID: mdl-35709828

ABSTRACT

Gastrulation controls the emergence of cellular diversity and axis patterning in the early embryo. In mammals, this transformation is orchestrated by dynamic signalling centres at the interface of embryonic and extraembryonic tissues1-3. Elucidating the molecular framework of axis formation in vivo is fundamental for our understanding of human development4-6 and to advance stem-cell-based regenerative approaches7. Here we illuminate early gastrulation of marmoset embryos in utero using spatial transcriptomics and stem-cell-based embryo models. Gaussian process regression-based 3D transcriptomes delineate the emergence of the anterior visceral endoderm, which is hallmarked by conserved (HHEX, LEFTY2, LHX1) and primate-specific (POSTN, SDC4, FZD5) factors. WNT signalling spatially coordinates the formation of the primitive streak in the embryonic disc and is counteracted by SFRP1 and SFRP2 to sustain pluripotency in the anterior domain. Amnion specification occurs at the boundaries of the embryonic disc through ID1, ID2 and ID3 in response to BMP signalling, providing a developmental rationale for amnion differentiation of primate pluripotent stem cells (PSCs). Spatial identity mapping demonstrates that primed marmoset PSCs exhibit the highest similarity to the anterior embryonic disc, whereas naive PSCs resemble the preimplantation epiblast. Our 3D transcriptome models reveal the molecular code of lineage specification in the primate embryo and provide an in vivo reference to decipher human development.


Subject(s)
Callithrix , Gastrulation , Uterus , Animals , Callithrix/embryology , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Endoderm/cytology , Endoderm/embryology , Female , Gene Expression Profiling , Germ Layers/cytology , Germ Layers/embryology , Humans , Pluripotent Stem Cells/cytology
3.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35792865

ABSTRACT

The trophoblast lineage safeguards fetal development by mediating embryo implantation, immune tolerance, nutritional supply and gas exchange. Human trophoblast stem cells (hTSCs) provide a platform to study lineage specification of placental tissues; however, the regulatory network controlling self-renewal remains elusive. Here, we present a single-cell atlas of human trophoblast development from zygote to mid-gestation together with single-cell profiling of hTSCs. We determine the transcriptional networks of trophoblast lineages in vivo and leverage probabilistic modelling to identify a role for MAPK signalling in trophoblast differentiation. Placenta- and blastoid-derived hTSCs consistently map between late trophectoderm and early cytotrophoblast, in contrast to blastoid-trophoblast, which correspond to trophectoderm. We functionally assess the requirement of the predicted cytotrophoblast network in an siRNA-screen and reveal 15 essential regulators for hTSC self-renewal, including MAZ, NFE2L3, TFAP2C, NR2F2 and CTNNB1. Our human trophoblast atlas provides a powerful analytical resource to delineate trophoblast cell fate acquisition, to elucidate transcription factors required for hTSC self-renewal and to gauge the developmental stage of in vitro cultured cells.


Subject(s)
Placentation , Trophoblasts , Basic-Leucine Zipper Transcription Factors , Cell Differentiation/genetics , Female , Humans , Placenta , Pregnancy , Stem Cells
4.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-36125063

ABSTRACT

The early specification and rapid growth of extraembryonic membranes are distinctive hallmarks of primate embryogenesis. These complex tasks are resolved through an intricate combination of signals controlling the induction of extraembryonic lineages and, at the same time, safeguarding the pluripotent epiblast. Here, we delineate the signals orchestrating primate epiblast and amnion identity. We encapsulated marmoset pluripotent stem cells into agarose microgels and identified culture conditions for the development of epiblast- and amnion-spheroids. Spatial identity mapping authenticated spheroids generated in vitro by comparison with marmoset embryos in vivo. We leveraged the microgel system to functionally interrogate the signalling environment of the post-implantation primate embryo. Single-cell profiling of the resulting spheroids demonstrated that activin/nodal signalling is required for embryonic lineage identity. BMP4 promoted amnion formation and maturation, which was counteracted by FGF signalling. Our combination of microgel culture, single-cell profiling and spatial identity mapping provides a powerful approach to decipher the essential cues for embryonic and extraembryonic lineage formation in primate embryogenesis.


Subject(s)
Microgels , Activins , Amnion , Animals , Callithrix , Cell Differentiation , Germ Layers , Sepharose
5.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33452132

ABSTRACT

OCT4 is a fundamental component of the molecular circuitry governing pluripotency in vivo and in vitro. To determine how OCT4 establishes and protects the pluripotent lineage in the embryo, we used comparative single-cell transcriptomics and quantitative immunofluorescence on control and OCT4 null blastocyst inner cell masses at two developmental stages. Surprisingly, activation of most pluripotency-associated transcription factors in the early mouse embryo occurs independently of OCT4, with the exception of the JAK/STAT signaling machinery. Concurrently, OCT4 null inner cell masses ectopically activate a subset of trophectoderm-associated genes. Inspection of metabolic pathways implicates the regulation of rate-limiting glycolytic enzymes by OCT4, consistent with a role in sustaining glycolysis. Furthermore, up-regulation of the lysosomal pathway was specifically detected in OCT4 null embryos. This finding implicates a requirement for OCT4 in the production of normal trophectoderm. Collectively, our findings uncover regulation of cellular metabolism and biophysical properties as mechanisms by which OCT4 instructs pluripotency.


Subject(s)
Cell Lineage/genetics , Embryonic Development/immunology , Octamer Transcription Factor-3/genetics , STAT3 Transcription Factor/genetics , Animals , Blastocyst Inner Cell Mass/metabolism , Embryo, Mammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Glycolysis/genetics , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/genetics , Single-Cell Analysis
6.
Development ; 145(3)2018 02 07.
Article in English | MEDLINE | ID: mdl-29361568

ABSTRACT

Single-cell profiling techniques create opportunities to delineate cell fate progression in mammalian development. Recent studies have provided transcriptome data from human pre-implantation embryos, in total comprising nearly 2000 individual cells. Interpretation of these data is confounded by biological factors, such as variable embryo staging and cell-type ambiguity, as well as technical challenges in the collective analysis of datasets produced with different sample preparation and sequencing protocols. Here, we address these issues to assemble a complete gene expression time course spanning human pre-implantation embryogenesis. We identify key transcriptional features over developmental time and elucidate lineage-specific regulatory networks. We resolve post-hoc cell-type assignment in the blastocyst, and define robust transcriptional prototypes that capture epiblast and primitive endoderm lineages. Examination of human pluripotent stem cell transcriptomes in this framework identifies culture conditions that sustain a naïve state pertaining to the inner cell mass. Our approach thus clarifies understanding both of lineage segregation in the early human embryo and of in vitro stem cell identity, and provides an analytical resource for comparative molecular embryology.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cell Line , Cell Lineage/genetics , Cell Lineage/physiology , Chromosome Mapping , Embryo Culture Techniques , Embryonic Development/genetics , Embryonic Development/physiology , Gene Expression Profiling , Genetic Markers , Germ Layers/cytology , Germ Layers/embryology , Germ Layers/metabolism , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Primates , Single-Cell Analysis
7.
Development ; 145(21)2018 11 09.
Article in English | MEDLINE | ID: mdl-30413530

ABSTRACT

The mouse embryo is the canonical model for mammalian preimplantation development. Recent advances in single cell profiling allow detailed analysis of embryogenesis in other eutherian species, including human, to distinguish conserved from divergent regulatory programs and signalling pathways in the rodent paradigm. Here, we identify and compare transcriptional features of human, marmoset and mouse embryos by single cell RNA-seq. Zygotic genome activation correlates with the presence of polycomb repressive complexes in all three species, while ribosome biogenesis emerges as a predominant attribute in primate embryos, supporting prolonged translation of maternally deposited RNAs. We find that transposable element expression signatures are species, stage and lineage specific. The pluripotency network in the primate epiblast lacks certain regulators that are operative in mouse, but encompasses WNT components and genes associated with trophoblast specification. Sequential activation of GATA6, SOX17 and GATA4 markers of primitive endoderm identity is conserved in primates. Unexpectedly, OTX2 is also associated with primitive endoderm specification in human and non-human primate blastocysts. Our cross-species analysis demarcates both conserved and primate-specific features of preimplantation development, and underscores the molecular adaptability of early mammalian embryogenesis.


Subject(s)
Callithrix/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Single-Cell Analysis , Transcriptome/genetics , Animals , Conserved Sequence/genetics , Endoderm/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Germ Layers/metabolism , Humans , Mice , Otx Transcription Factors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics , Transcription, Genetic
8.
Development ; 144(2): 175-186, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28096211

ABSTRACT

Naïve pluripotent mouse embryonic stem cells (ESCs) resemble the preimplantation epiblast and efficiently contribute to chimaeras. Primate ESCs correspond to the postimplantation embryo and fail to resume development in chimaeric assays. Recent data suggest that human ESCs can be 'reset' to an earlier developmental stage, but their functional capacity remains ill defined. Here, we discuss how the naïve state is inherently linked to preimplantation epiblast identity in the embryo. We hypothesise that distinctive features of primate development provide stringent criteria to evaluate naïve pluripotency in human and other primate cells. Based on our hypothesis, we define 12 key hallmarks of naïve pluripotency, five of which are specific to primates. These hallmarks may serve as a functional framework to assess human naïve ESCs.


Subject(s)
Embryonic Development/physiology , Pluripotent Stem Cells/physiology , Primates/embryology , Animals , Blastocyst/cytology , Cell Differentiation , Cells, Cultured , Chimera/embryology , Embryo, Mammalian , Embryonic Stem Cells/physiology , Germ Layers/cytology , Humans , Mice , Rats
10.
Aging Cell ; : e14200, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38757354

ABSTRACT

The sperm epigenome is thought to affect the developmental programming of the resulting embryo, influencing health and disease in later life. Age-related methylation changes in the sperm of old fathers may mediate the increased risks for reproductive and offspring medical problems. The impact of paternal age on sperm methylation has been extensively studied in humans and, to a lesser extent, in rodents and cattle. Here, we performed a comparative analysis of paternal age effects on protein-coding genes in the human and marmoset sperm methylomes. The marmoset has gained growing importance as a non-human primate model of aging and age-related diseases. Using reduced representation bisulfite sequencing, we identified age-related differentially methylated transcription start site (ageTSS) regions in 204 marmoset and 27 human genes. The direction of methylation changes was the opposite, increasing with age in marmosets and decreasing in humans. None of the identified ageTSS was differentially methylated in both species. Although the average methylation levels of all TSS regions were highly correlated between marmosets and humans, with the majority of TSS being hypomethylated in sperm, more than 300 protein-coding genes were endowed with species-specifically (hypo)methylated TSS. Several genes of the glycosphingolipid (GSL) biosynthesis pathway, which plays a role in embryonic stem cell differentiation and regulation of development, were hypomethylated (<5%) in human and fully methylated (>95%) in marmoset sperm. The expression levels and patterns of defined sets of GSL genes differed considerably between human and marmoset pre-implantation embryo stages and blastocyst tissues, respectively.

11.
Biol Open ; 12(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36504370

ABSTRACT

We previously demonstrated gradual loss of epiblast during diapause in embryos lacking components of the LIF/IL6 receptor. Here, we explore the requirement for the downstream signalling transducer andactivator of transcription STAT3 and its target, TFCP2L1, in maintenance of naïve pluripotency. Unlike conventional markers, such as NANOG, which remains high in epiblast until implantation, both STAT3 and TFCP2L1 proteins decline during blastocyst expansion, but intensify in the embryonic region after induction of diapause, as observed visually and confirmed using our image-analysis pipeline, consistent with our previous transcriptional expression data. Embryos lacking STAT3 or TFCP2L1 underwent catastrophic loss of most of the inner cell mass during the first few days of diapause, indicating involvement of signals in addition to LIF/IL6 for sustaining naïve pluripotency in vivo. By blocking MEK/ERK signalling from the morula stage, we could derive embryonic stem cells with high efficiency from STAT3 null embryos, but not those lacking TFCP2L1, suggesting a hitherto unknown additional role for this essential STAT3 target in transition from embryo to embryonic stem cells in vitro. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Pluripotent Stem Cells , Repressor Proteins , STAT3 Transcription Factor , Mice , Blastocyst/metabolism , Embryonic Stem Cells/metabolism , Leukemia Inhibitory Factor/metabolism , Pluripotent Stem Cells/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Animals
12.
Life Sci Alliance ; 6(8)2023 08.
Article in English | MEDLINE | ID: mdl-37217306

ABSTRACT

Human germline-soma segregation occurs during weeks 2-3 in gastrulating embryos. Although direct studies are hindered, here, we investigate the dynamics of human primordial germ cell (PGCs) specification using in vitro models with temporally resolved single-cell transcriptomics and in-depth characterisation using in vivo datasets from human and nonhuman primates, including a 3D marmoset reference atlas. We elucidate the molecular signature for the transient gain of competence for germ cell fate during peri-implantation epiblast development. Furthermore, we show that both the PGCs and amnion arise from transcriptionally similar TFAP2A-positive progenitors at the posterior end of the embryo. Notably, genetic loss of function experiments shows that TFAP2A is crucial for initiating the PGC fate without detectably affecting the amnion and is subsequently replaced by TFAP2C as an essential component of the genetic network for PGC fate. Accordingly, amniotic cells continue to emerge from the progenitors in the posterior epiblast, but importantly, this is also a source of nascent PGCs.


Subject(s)
Embryo, Mammalian , Gene Regulatory Networks , Animals , Humans , Gene Regulatory Networks/genetics , Cell Differentiation/genetics , Germ Layers , Germ Cells
13.
Stem Cell Reports ; 18(10): 1987-2002, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37683645

ABSTRACT

Primate germ cell development remains largely unexplored due to limitations in sample collection and the long duration of development. In mice, primordial germ cell-like cells (PGCLCs) derived from pluripotent stem cells (PSCs) can develop into functional gametes by in vitro culture or in vivo transplantation. Such PGCLC-mediated induction of mature gametes in primates is highly useful for understanding human germ cell development. Since marmosets generate functional sperm earlier than other species, recapitulating the whole male germ cell development process is technically more feasible. Here, we induced the differentiation of iPSCs into gonocyte-like cells via PGCLCs in marmosets. First, we developed an mRNA transfection-based method to efficiently generate PGCLCs. Subsequently, to promote PGCLC differentiation, xenoreconstituted testes (xrtestes) were generated in the mouse kidney capsule. PGCLCs show progressive DNA demethylation and stepwise expression of developmental marker genes. This study provides an efficient platform for the study of marmoset germ cell development.


Subject(s)
Callithrix , Semen , Humans , Male , Animals , Mice , Germ Cells , Cell Differentiation/genetics , RNA, Messenger/genetics
14.
Nat Commun ; 14(1): 4022, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37419903

ABSTRACT

Biomechanical cues are instrumental in guiding embryonic development and cell differentiation. Understanding how these physical stimuli translate into transcriptional programs will provide insight into mechanisms underlying mammalian pre-implantation development. Here, we explore this type of regulation by exerting microenvironmental control over mouse embryonic stem cells. Microfluidic encapsulation of mouse embryonic stem cells in agarose microgels stabilizes the naive pluripotency network and specifically induces expression of Plakoglobin (Jup), a vertebrate homolog of ß-catenin. Overexpression of Plakoglobin is sufficient to fully re-establish the naive pluripotency gene regulatory network under metastable pluripotency conditions, as confirmed by single-cell transcriptome profiling. Finally, we find that, in the epiblast, Plakoglobin was exclusively expressed at the blastocyst stage in human and mouse embryos - further strengthening the link between Plakoglobin and naive pluripotency in vivo. Our work reveals Plakoglobin as a mechanosensitive regulator of naive pluripotency and provides a paradigm to interrogate the effects of volumetric confinement on cell-fate transitions.


Subject(s)
Embryonic Development , Germ Layers , Animals , Mice , Humans , gamma Catenin/genetics , gamma Catenin/metabolism , Cell Differentiation/genetics , Germ Layers/metabolism , Embryonic Development/genetics , Gene Expression Profiling , Blastocyst/metabolism , Mammals/genetics
15.
Stem Cells ; 29(2): 193-205, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21732478

ABSTRACT

ESCs undergoing neural differentiation in vitro display an intrinsic heterogeneity with a large subset of the cells forming polarized neural rosettes that maintain the neural progenitor microenvironment. This heterogeneity is not only necessary for normal development but also causes substantial technical challenges for practical applications. Here, we report a novel regulator of early neural progenitors, the apical polarity protein Crb2 (Crumbs homologue 2). Employing monolayer differentiation of mouse ESCs to model neurogenesis in vitro, we find that Crb2 is upregulated with Sox1 and Musashi at the onset of neuroepithelial specification and localizes to the apical side of neural rosettes. Stable Crb2-knockdown (KD) lines die at the onset of neural specification and fail to stabilize several apical polarity proteins. However, these cells are able to proliferate under self-renewing conditions and can be differentiated into mesodermal and endodermal lineages. Conversely, Crb2 overexpression during neural differentiation results in elevated levels of other apical polarity proteins and increases proliferation. Additionally, sustained overexpression of Crb2 reduces terminal differentiation into TuJ1-positive neurons. Furthermore, we demonstrate that Crb2 overexpression under self-renewing conditions increases glycogen synthase kinase (GSK)-3ß inhibition, correlating with an increase in clonogenicity. To confirm the importance of GSK-3ß inhibition downstream of Crb2, we show that Crb2-KD cells can be forced into neural lineages by blocking GSK-3ß function and supplementing Epidermal Growth Factor (EGF) and basic Fibroblast Growth Factor (bFGF). Thus, this is the first demonstration that a member of the Crumbs family is essential for survival and differentiation of ESC-derived neural progenitors.


Subject(s)
Cell Polarity , Embryonic Stem Cells/metabolism , Membrane Proteins/biosynthesis , Neural Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/cytology , Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 2/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Rosette Formation , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Tubulin/biosynthesis
16.
Cell Stem Cell ; 29(6): 869-870, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35659870

ABSTRACT

In this issue of Cell Stem Cell, Simunovic et al. (2022) establish embryoids by combining embryonic and extraembryonic components derived from human pluripotent stem cells. The embryoids resemble human embryos cultured to post-implantation stages in vitro with regard to morphology, symmetry breaking, and the formation of primitive streak-like cell types.


Subject(s)
Embryo, Mammalian , Pluripotent Stem Cells , Embryo Implantation , Humans
17.
Philos Trans R Soc Lond B Biol Sci ; 377(1865): 20210256, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36252209

ABSTRACT

Implantation of the conceptus into the uterus is absolutely essential for successful embryo development. In humans, our understanding of this process has remained rudimentary owing to the inaccessibility of early implantation stages. Non-human primates recapitulate many aspects of human embryo development and provide crucial insights into trophoblast development, uterine receptivity and embryo invasion. Moreover, primate species exhibit a variety of implantation strategies and differ in embryo invasion depths. This review examines conservation and divergence of the key processes required for embryo implantation in different primates and in comparison with the canonical rodent model. We discuss trophectoderm compartmentalization, endometrial remodelling and embryo adhesion and invasion. Finally, we propose that studying the mechanism controlling invasion depth between different primate species may provide new insights and treatment strategies for placentation disorders in humans. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.


Subject(s)
Embryo Implantation , Primates , Animals , Female , Pregnancy , Endometrium/embryology , Primates/embryology , Trophoblasts , Uterus , Humans
18.
Nat Commun ; 13(1): 3407, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35710749

ABSTRACT

Mammalian embryogenesis relies on glycolysis and oxidative phosphorylation to balance the generation of biomass with energy production. However, the dynamics of metabolic regulation in the postimplantation embryo in vivo have remained elusive due to the inaccessibility of the implanted conceptus for biochemical studies. To address this issue, we compiled single-cell embryo profiling data in six mammalian species and determined their metabolic dynamics through glycolysis and oxidative phosphorylation associated gene expression. Strikingly, we identify a conserved switch from bivalent respiration in the late blastocyst towards a glycolytic metabolism in early gastrulation stages across species, which is independent of embryo implantation. Extraembryonic lineages followed the dynamics of the embryonic lineage, except visceral endoderm. Finally, we demonstrate that in vitro primate embryo culture substantially impacts metabolic gene regulation by comparison to in vivo samples. Our work reveals a conserved metabolic programme despite different implantation modes and highlights the need to optimise postimplantation embryo culture protocols.


Subject(s)
Embryo, Mammalian , Transcriptome , Animals , Blastocyst/metabolism , Cell Lineage/genetics , Embryo Implantation/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Mammals/genetics , Transcriptome/genetics
19.
Commun Biol ; 4(1): 749, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140619

ABSTRACT

The uterus is the organ for embryo implantation and fetal development. Most current models of the uterus are centred around capturing its function during later stages of pregnancy to increase the survival in pre-term births. However, in vitro models focusing on the uterine tissue itself would allow modelling of pathologies including endometriosis and uterine cancers, and open new avenues to investigate embryo implantation and human development. Motivated by these key questions, we discuss how stem cell-based uteri may be engineered from constituent cell parts, either as advanced self-organising cultures, or by controlled assembly through microfluidic and print-based technologies.


Subject(s)
Stem Cells/physiology , Tissue Engineering/methods , Uterus/cytology , Uterus/physiology , Animals , Embryo Implantation/physiology , Female , Fetal Development/physiology , Humans , Models, Biological , Placenta/physiology , Pregnancy , Primates , Tissue Scaffolds
20.
Stem Cell Reports ; 16(5): 1347-1362, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979603

ABSTRACT

Human periimplantation development requires the transformation of the naive pluripotent epiblast into a polarized epithelium. Lumenogenesis plays a critical role in this process, as the epiblast undergoes rosette formation and lumen expansion to form the amniotic cavity. Here, we present a high-throughput in vitro model for epiblast morphogenesis. We established a microfluidic workflow to encapsulate human pluripotent stem cells (hPSCs) into monodisperse agarose microgels. Strikingly, hPSCs self-organized into polarized epiblast spheroids that could be maintained in self-renewing and differentiating conditions. Encapsulated primed hPSCs required Rho-associated kinase inhibition, in contrast to naive hPSCs. We applied microgel suspension culture to examine the lumen-forming capacity of hPSCs and reveal an increase in lumenogenesis during the naive-to-primed transition. Finally, we demonstrate the feasibility of co-encapsulating cell types across different lineages and species. Our work provides a foundation for stem cell-based embryo models to interrogate the critical components of human epiblast self-organization and morphogenesis.


Subject(s)
Cell Culture Techniques , Induced Pluripotent Stem Cells/cytology , Microgels/chemistry , Morphogenesis , Sepharose/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Survival/drug effects , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Germ Layers/cytology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Morphogenesis/drug effects , Protein Kinase Inhibitors/pharmacology , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL