Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 686
Filtrar
1.
Development ; 151(8)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38563517

RESUMO

The lineage decision that generates the epiblast and primitive endoderm from the inner cell mass (ICM) is a paradigm for cell fate specification. Recent mathematics has formalized Waddington's landscape metaphor and proven that lineage decisions in detailed gene network models must conform to a small list of low-dimensional stereotypic changes called bifurcations. The most plausible bifurcation for the ICM is the so-called heteroclinic flip that we define and elaborate here. Our re-analysis of recent data suggests that there is sufficient cell movement in the ICM so the FGF signal, which drives the lineage decision, can be treated as spatially uniform. We thus extend the bifurcation model for a single cell to the entire ICM by means of a self-consistently defined time-dependent FGF signal. This model is consistent with available data and we propose additional dynamic experiments to test it further. This demonstrates that simplified, quantitative and intuitively transparent descriptions are possible when attention is shifted from specific genes to lineages. The flip bifurcation is a very plausible model for any situation where the embryo needs control over the relative proportions of two fates by a morphogen feedback.


Assuntos
Blastocisto , Diferenciação Celular , Linhagem da Célula , Modelos Biológicos , Animais , Camundongos , Blastocisto/metabolismo , Blastocisto/citologia , Transdução de Sinais , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Endoderma/citologia , Endoderma/metabolismo , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo
2.
Nature ; 626(7998): 357-366, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38052228

RESUMO

Recently, several studies using cultures of human embryos together with single-cell RNA-seq analyses have revealed differences between humans and mice, necessitating the study of human embryos1-8. Despite the importance of human embryology, ethical and legal restrictions have limited post-implantation-stage studies. Thus, recent efforts have focused on developing in vitro self-organizing models using human stem cells9-17. Here, we report genetic and non-genetic approaches to generate authentic hypoblast cells (naive hPSC-derived hypoblast-like cells (nHyCs))-known to give rise to one of the two extraembryonic tissues essential for embryonic development-from naive human pluripotent stem cells (hPSCs). Our nHyCs spontaneously assemble with naive hPSCs to form a three-dimensional bilaminar structure (bilaminoids) with a pro-amniotic-like cavity. In the presence of additional naive hPSC-derived analogues of the second extraembryonic tissue, the trophectoderm, the efficiency of bilaminoid formation increases from 20% to 40%, and the epiblast within the bilaminoids continues to develop in response to trophectoderm-secreted IL-6. Furthermore, we show that bilaminoids robustly recapitulate the patterning of the anterior-posterior axis and the formation of cells reflecting the pregastrula stage, the emergence of which can be shaped by genetically manipulating the DKK1/OTX2 hypoblast-like domain. We have therefore successfully modelled and identified the mechanisms by which the two extraembryonic tissues efficiently guide the stage-specific growth and progression of the epiblast as it establishes the post-implantation landmarks of human embryogenesis.


Assuntos
Desenvolvimento Embrionário , Camadas Germinativas , Células-Tronco Pluripotentes , Humanos , Diferenciação Celular , Implantação do Embrião , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Células-Tronco Pluripotentes/citologia , Interleucina-6/metabolismo , Gástrula/citologia , Gástrula/embriologia , Âmnio/citologia , Âmnio/embriologia , Âmnio/metabolismo , Ectoderma/citologia , Ectoderma/embriologia , Ectoderma/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo
3.
Nature ; 626(7998): 367-376, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38092041

RESUMO

Implantation of the human embryo begins a critical developmental stage that comprises profound events including axis formation, gastrulation and the emergence of haematopoietic system1,2. Our mechanistic knowledge of this window of human life remains limited due to restricted access to in vivo samples for both technical and ethical reasons3-5. Stem cell models of human embryo have emerged to help unlock the mysteries of this stage6-16. Here we present a genetically inducible stem cell-derived embryoid model of early post-implantation human embryogenesis that captures the reciprocal codevelopment of embryonic tissue and the extra-embryonic endoderm and mesoderm niche with early haematopoiesis. This model is produced from induced pluripotent stem cells and shows unanticipated self-organizing cellular programmes similar to those that occur in embryogenesis, including the formation of amniotic cavity and bilaminar disc morphologies as well as the generation of an anterior hypoblast pole and posterior domain. The extra-embryonic layer in these embryoids lacks trophoblast and shows advanced multilineage yolk sac tissue-like morphogenesis that harbours a process similar to distinct waves of haematopoiesis, including the emergence of erythroid-, megakaryocyte-, myeloid- and lymphoid-like cells. This model presents an easy-to-use, high-throughput, reproducible and scalable platform to probe multifaceted aspects of human development and blood formation at the early post-implantation stage. It will provide a tractable human-based model for drug testing and disease modelling.


Assuntos
Desenvolvimento Embrionário , Camadas Germinativas , Hematopoese , Saco Vitelino , Humanos , Implantação do Embrião , Endoderma/citologia , Endoderma/embriologia , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Saco Vitelino/citologia , Saco Vitelino/embriologia , Mesoderma/citologia , Mesoderma/embriologia , Células-Tronco Pluripotentes Induzidas/citologia , Âmnio/citologia , Âmnio/embriologia , Corpos Embrioides/citologia , Linhagem da Célula , Biologia do Desenvolvimento/métodos , Biologia do Desenvolvimento/tendências
4.
Nature ; 622(7983): 562-573, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37673118

RESUMO

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.


Assuntos
Implantação do Embrião , Embrião de Mamíferos , Desenvolvimento Embrionário , Células-Tronco Embrionárias Humanas , Humanos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Fertilização , Gastrulação , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Células-Tronco Embrionárias Humanas/citologia , Trofoblastos/citologia , Saco Vitelino/citologia , Saco Vitelino/embriologia , Células Gigantes/citologia
5.
Nature ; 622(7983): 584-593, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37369347

RESUMO

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


Assuntos
Implantação do Embrião , Embrião de Mamíferos , Desenvolvimento Embrionário , Modelos Biológicos , Células-Tronco Pluripotentes , Feminino , Humanos , Gravidez , Proteínas Morfogenéticas Ósseas , Diferenciação Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Corpos Embrioides/citologia , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Células-Tronco Embrionárias Humanas/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células-Tronco Pluripotentes/citologia
6.
Nature ; 622(7983): 574-583, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37369348

RESUMO

Investigating human development is a substantial scientific challenge due to the technical and ethical limitations of working with embryonic samples. In the face of these difficulties, stem cells have provided an alternative to experimentally model inaccessible stages of human development in vitro1-13. Here we show that human pluripotent stem cells can be triggered to self-organize into three-dimensional structures that recapitulate some key spatiotemporal events of early human post-implantation embryonic development. Our system reproducibly captures spontaneous differentiation and co-development of embryonic epiblast-like and extra-embryonic hypoblast-like lineages, establishes key signalling hubs with secreted modulators and undergoes symmetry breaking-like events. Single-cell transcriptomics confirms differentiation into diverse cell states of the perigastrulating human embryo14,15 without establishing placental cell types, including signatures of post-implantation epiblast, amniotic ectoderm, primitive streak, mesoderm, early extra-embryonic endoderm, as well as initial yolk sac induction. Collectively, our system captures key features of human embryonic development spanning from Carnegie stage16 4-7, offering a reproducible, tractable and scalable experimental platform to understand the basic cellular and molecular mechanisms that underlie human development, including new opportunities to dissect congenital pathologies with high throughput.


Assuntos
Linhagem da Célula , Implantação do Embrião , Desenvolvimento Embrionário , Células-Tronco Pluripotentes , Feminino , Humanos , Gravidez , Diferenciação Celular , Camadas Germinativas/citologia , Camadas Germinativas/enzimologia , Células-Tronco Embrionárias Humanas/citologia , Placenta/citologia , Células-Tronco Pluripotentes/citologia , Linha Primitiva/citologia , Linha Primitiva/embriologia , Saco Vitelino/citologia , Saco Vitelino/embriologia
7.
Nature ; 609(7925): 136-143, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35709828

RESUMO

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.


Assuntos
Callithrix , Gastrulação , Útero , Animais , Callithrix/embriologia , Diferenciação Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Endoderma/citologia , Endoderma/embriologia , Feminino , Perfilação da Expressão Gênica , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Humanos , Células-Tronco Pluripotentes/citologia
8.
Cell Stem Cell ; 29(3): 347-348, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35245463

RESUMO

Human naive pluripotent stem cells have the remarkable ability to generate blastoids comprising trophectoderm, epiblast, and hypoblast-like cells. In this issue, Taubenschmid-Stowers et al. (2022) show that human naive pluripotent stem cell cultures contain cells that resemble the 8-cell human embryo, providing a model to study zygotic genome activation.


Assuntos
Camadas Germinativas , Células-Tronco Pluripotentes , Embrião de Mamíferos/citologia , Camadas Germinativas/citologia , Humanos , Células-Tronco Pluripotentes/citologia
9.
Science ; 375(6580): 574-578, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-35113719

RESUMO

The mammalian blastocyst consists of three distinct cell types: epiblast, trophoblast (TB), and primitive endoderm (PrE). Although embryonic stem cells (ESCs) and trophoblast stem cells (TSCs) retain the functional properties of epiblast and TB, respectively, stem cells that fully recapitulate the developmental potential of PrE have not been established. Here, we report derivation of primitive endoderm stem cells (PrESCs) in mice. PrESCs recapitulate properties of embryonic day 4.5 founder PrE, are efficiently incorporated into PrE upon blastocyst injection, generate functionally competent PrE-derived tissues, and support fetal development of PrE-depleted blastocysts in chimeras. Furthermore, PrESCs can establish interactions with ESCs and TSCs and generate descendants with yolk sac-like structures in utero. Establishment of PrESCs will enable the elucidation of the mechanisms for PrE specification and subsequent pre- and postimplantation development.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Endoderma/citologia , Endoderma/embriologia , Animais , Blastocisto/citologia , Blastocisto/fisiologia , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Quimera , Desenvolvimento Embrionário , Endoderma/crescimento & desenvolvimento , Desenvolvimento Fetal , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Trofoblastos/citologia , Trofoblastos/fisiologia
10.
Development ; 149(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35179180

RESUMO

Specification of primordial germ cells requires a proportion of the cells in the posterior of the epiblast to reacquire pluripotency. A new paper in Development describes how OVOL2 is involved in regulating the balance between mesodermal fate and germ cell fate during gastrulation. We caught up with the first author, Yuki Naitou, and corresponding author, Katsuhiko Hayashi (Osaka University), to find out more about the paper and their future research.


Assuntos
Células Germinativas/metabolismo , Pesquisadores/psicologia , Fatores de Transcrição/metabolismo , Animais , Autoria , Transição Epitelial-Mesenquimal , Gastrulação , Células Germinativas/citologia , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Humanos , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
11.
Stem Cell Reports ; 17(2): 427-442, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35090587

RESUMO

Elucidating regulatory relationships between transcription factors (TFs) and target genes is fundamental to understanding how cells control their identity and behavior. Unfortunately, existing computational gene regulatory network (GRN) reconstruction methods are imprecise, computationally burdensome, and fail to reveal dynamic regulatory topologies. Here, we present Epoch, a reconstruction tool that uses single-cell transcriptomics to accurately infer dynamic networks. We apply Epoch to identify the dynamic networks underpinning directed differentiation of mouse embryonic stem cells (ESCs) guided by multiple signaling pathways, and we demonstrate that modulating these pathways drives topological changes that bias cell fate potential. We also find that Peg3 rewires the pluripotency network to favor mesoderm specification. By integrating signaling pathways with GRNs, we trace how Wnt activation and PI3K suppression govern mesoderm and endoderm specification, respectively. Finally, we identify regulatory circuits of patterning and axis formation that distinguish in vitro and in vivo mesoderm specification.


Assuntos
Redes Reguladoras de Genes/genética , Camadas Germinativas/metabolismo , Animais , Diferenciação Celular , Endoderma/citologia , Endoderma/metabolismo , Camadas Germinativas/citologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/genética , Análise de Célula Única , Proteínas Wnt/metabolismo
12.
Biochem Biophys Res Commun ; 590: 125-131, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-34974300

RESUMO

Embryonic stem cells (ESCs) are derived from the inner cell mass of developing blastocysts, which have self-renewal ability and have the potential to develop or reconstitute into all embryonic lineages. Selenophosphate synthetase 1 (SEPHS1) is an essential protein in mouse early embryo development. However, the role of SEPHS1 in mouse ESCs remains to be elucidated. In this study, we generated Sephs1 KO ESCs and found that deficiency of SEPSH1 has little effect on pluripotency maintenance and proliferation. Notably, SEPHS1 deficiency impaired differentiation into three germ layers and gastruloid aggregation in vitro. RNA-seq analysis showed SEPHS1 is involved in cardiogenesis, verified by no beating signal in Sephs1 KO embryoid body at d10 and low expression of cardiac-related and contraction markers. Taken together, our results suggest that SPEHS1 is dispensable in ESC self-renewal, but indispensable in subsequent germ layer differentiation especially for functional cardiac lineage.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Miocárdio/citologia , Fosfotransferases/metabolismo , Animais , Diferenciação Celular/genética , Corpos Embrioides/citologia , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfotransferases/deficiência , Transcrição Gênica
13.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34921114

RESUMO

N6-methyladenosine (m6A) deposition on messenger RNA (mRNA) controls embryonic stem cell (ESC) fate by regulating the mRNA stabilities of pluripotency and lineage transcription factors (TFs) [P. J. Batista et al., Cell Stem Cell 15, 707-719 (2014); Y. Wang et al., Nat. Cell Biol. 16, 191-198 (2014); and S. Geula et al., Science 347, 1002-1006 (2015)]. If the mRNAs of these two TF groups become stabilized, it remains unclear how the pluripotency or lineage commitment decision is implemented. We performed noninvasive quantification of Nanog and Oct4 TF protein levels in reporter ESCs to define cell-state dynamics at single-cell resolution. Long-term single-cell tracking shows that immediate m6A depletion by Mettl3 knock-down in serum/leukemia inhibitory factor supports both pluripotency maintenance and its departure. This is mediated by differential and opposing signaling pathways. Increased FGF5 mRNA stability activates pErk, leading to Nanog down-regulation. FGF5-mediated coactivation of pAkt reenforces Nanog expression. In formative stem cells poised toward differentiation, m6A depletion activates both pErk and pAkt, increasing the propensity for mesendodermal lineage induction. Stable m6A depletion by Mettl3 knock-out also promotes pErk activation. Higher pErk counteracts the pluripotency exit delay exhibited by stably m6A-depleted cells upon differentiation. At single-cell resolution, we illustrate that decreasing m6A abundances activates pErk and pAkt-signaling, regulating pluripotency departure.


Assuntos
Adenosina/análogos & derivados , Células-Tronco Embrionárias/fisiologia , Sistema de Sinalização das MAP Quinases , Adenosina/metabolismo , Animais , Linhagem Celular , Camadas Germinativas/citologia , Camundongos
14.
Development ; 148(23)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34874452

RESUMO

Despite four decades of effort, robust propagation of pluripotent stem cells from livestock animals remains challenging. The requirements for self-renewal are unclear and the relationship of cultured stem cells to pluripotent cells resident in the embryo uncertain. Here, we avoided using feeder cells or serum factors to provide a defined culture microenvironment. We show that the combination of activin A, fibroblast growth factor and the Wnt inhibitor XAV939 (AFX) supports establishment and continuous expansion of pluripotent stem cell lines from porcine, ovine and bovine embryos. Germ layer differentiation was evident in teratomas and readily induced in vitro. Global transcriptome analyses highlighted commonality in transcription factor expression across the three species, while global comparison with porcine embryo stages showed proximity to bilaminar disc epiblast. Clonal genetic manipulation and gene targeting were exemplified in porcine stem cells. We further demonstrated that genetically modified AFX stem cells gave rise to cloned porcine foetuses by nuclear transfer. In summary, for major livestock mammals, pluripotent stem cells related to the formative embryonic disc are reliably established using a common and defined signalling environment. This article has an associated 'The people behind the papers' interview.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/metabolismo , Camadas Germinativas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Bovinos , Embrião de Mamíferos/citologia , Camadas Germinativas/citologia , Gado , Células-Tronco Pluripotentes/citologia , Ovinos , Especificidade da Espécie , Suínos
15.
Cell Rep ; 37(6): 109990, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34758327

RESUMO

As pluripotent human embryonic stem cells progress toward one germ layer fate, they lose the ability to adopt alternative fates. Using a low-dimensional reaction coordinate to monitor progression toward ectoderm, we show that a differentiating stem cell's probability of adopting a mesendodermal fate given appropriate signals falls sharply at a point along the ectoderm trajectory. We use this reaction coordinate to prospectively isolate and profile differentiating cells based on their mesendoderm competence and analyze their RNA sequencing (RNA-seq) and assay for transposase-accessible chromatin using sequencing (ATAC-seq) profiles to identify transcription factors that control the cell's mesendoderm competence. By modulating these key transcription factors, we can expand or contract the window of competence to adopt the mesendodermal fate along the ectodermal differentiation trajectory. The ability of the underlying gene regulatory network to modulate competence is essential for understanding human development and controlling the fate choices of stem cells in vitro.


Assuntos
Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Células-Tronco Embrionárias Humanas/citologia , Mesoderma/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Diferenciação Celular , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Camadas Germinativas/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Mesoderma/metabolismo , Fator 3 de Transcrição de Octâmero/genética , RNA-Seq , Fatores de Transcrição SOXB1/genética
16.
Cell Rep ; 37(2): 109812, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34644585

RESUMO

Rabbit embryos develop as bilaminar discs at gastrulation as in humans and most other mammals, whereas rodents develop as egg cylinders. Primordial germ cells (PGCs) appear to originate during gastrulation according to many systematic studies on mammalian embryos. Here, we show that rabbit PGC (rbPGC) specification occurs at the posterior epiblast at the onset of gastrulation. Using newly derived rabbit pluripotent stem cells, we show robust and rapid induction of rbPGC-like cells in vitro with WNT and BMP morphogens, which reveals SOX17 as the critical regulator of rbPGC fate as in several non-rodent mammals. We posit that development as a bilaminar disc is a crucial determinant of the PGC regulators, regardless of the highly diverse development of extraembryonic tissues, including the amnion. We propose that investigations on rabbits with short gestation, large litters, and where gastrulation precedes implantation can contribute significantly to advances in early mammalian development.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/fisiologia , Gastrulação , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Movimento Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Pluripotentes/metabolismo , Coelhos , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
17.
Development ; 148(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34608934

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of the CAG repeats in the huntingtin gene (HTT). Although HD has been shown to have a developmental component, how early during human embryogenesis the HTT-CAG expansion can cause embryonic defects remains unknown. Here, we demonstrate a specific and highly reproducible CAG length-dependent phenotypic signature in a synthetic model for human gastrulation derived from human embryonic stem cells (hESCs). Specifically, we observed a reduction in the extension of the ectodermal compartment that is associated with enhanced activin signaling. Surprisingly, rather than a cell-autonomous effect, tracking the dynamics of TGFß signaling demonstrated that HTT-CAG expansion perturbs the spatial restriction of activin response. This is due to defects in the apicobasal polarization in the context of the polarized epithelium of the 2D gastruloid, leading to ectopic subcellular localization of TGFß receptors. This work refines the earliest developmental window for the prodromal phase of HD to the first 2 weeks of human development, as modeled by our 2D gastruloids.


Assuntos
Linhagem da Célula , Polaridade Celular , Camadas Germinativas/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Proteína Huntingtina/metabolismo , Ativinas/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Células-Tronco Embrionárias Humanas/citologia , Humanos , Proteína Huntingtina/genética , Camundongos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Expansão das Repetições de Trinucleotídeos
18.
Nat Commun ; 12(1): 5771, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599190

RESUMO

Germline specification in mammals occurs through an inductive process whereby competent cells in the post-implantation epiblast differentiate into primordial germ cells (PGC). The intrinsic factors that endow epiblast cells with the competence to respond to germline inductive signals remain unknown. Single-cell RNA sequencing across multiple stages of an in vitro PGC-like cells (PGCLC) differentiation system shows that PGCLC genes initially expressed in the naïve pluripotent stage become homogeneously dismantled in germline competent epiblast like-cells (EpiLC). In contrast, the decommissioning of enhancers associated with these germline genes is incomplete. Namely, a subset of these enhancers partly retain H3K4me1, accumulate less heterochromatic marks and remain accessible and responsive to transcriptional activators. Subsequently, as in vitro germline competence is lost, these enhancers get further decommissioned and lose their responsiveness to transcriptional activators. Importantly, using H3K4me1-deficient cells, we show that the loss of this histone modification reduces the germline competence of EpiLC and decreases PGCLC differentiation efficiency. Our work suggests that, although H3K4me1 might not be essential for enhancer function, it can facilitate the (re)activation of enhancers and the establishment of gene expression programs during specific developmental transitions.


Assuntos
Elementos Facilitadores Genéticos , Células Germinativas/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Animais , Diferenciação Celular , Cromatina/metabolismo , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica , Células Germinativas/citologia , Camadas Germinativas/citologia , Masculino , Metilação , Camundongos , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia , Mutação/genética , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , RNA-Seq , Análise de Célula Única , Sítio de Iniciação de Transcrição , Transcrição Gênica
19.
Stem Cell Reports ; 16(11): 2628-2641, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34678211

RESUMO

Quantitative analysis of human induced pluripotent stem cell (iPSC) lines from healthy donors is a powerful tool for uncovering the relationship between genetic variants and cellular behavior. We previously identified rare, deleterious non-synonymous single nucleotide variants (nsSNVs) in cell adhesion genes that are associated with outlier iPSC phenotypes in the pluripotent state. Here, we generated micropatterned colonies of iPSCs to test whether nsSNVs influence patterning of radially ordered germ layers. Using a custom-built image analysis pipeline, we quantified the differentiation phenotypes of 13 iPSC lines that harbor nsSNVs in genes related to cell adhesion or germ layer development. All iPSC lines differentiated into the three germ layers; however, there was donor-specific variation in germ layer patterning. We identified one line that presented an outlier phenotype of expanded endodermal differentiation, which was associated with a nsSNV in ITGB1. Our study establishes a platform for investigating the impact of nsSNVs on differentiation.


Assuntos
Diferenciação Celular/genética , Endoderma/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Integrina beta1/genética , Polimorfismo de Nucleotídeo Único , Adesão Celular/genética , Linhagem Celular , Endoderma/citologia , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Perfilação da Expressão Gênica/métodos , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fenótipo , Proteômica/métodos , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
20.
Development ; 148(21)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34651174

RESUMO

During embryonic development and tissue homeostasis, reproducible proportions of differentiated cell types are specified from populations of multipotent precursor cells. Molecular mechanisms that enable both robust cell-type proportioning despite variable initial conditions in the precursor cells, and the re-establishment of these proportions upon perturbations in a developing tissue remain to be characterized. Here, we report that the differentiation of robust proportions of epiblast-like and primitive endoderm-like cells in mouse embryonic stem cell cultures emerges at the population level through cell-cell communication via a short-range fibroblast growth factor 4 (FGF4) signal. We characterize the molecular and dynamical properties of the communication mechanism and show how it controls both robust cell-type proportioning from a wide range of experimentally controlled initial conditions, as well as the autonomous re-establishment of these proportions following the isolation of one cell type. The generation and maintenance of reproducible proportions of discrete cell types is a new function for FGF signaling that might operate in a range of developing tissues.


Assuntos
Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Fator 4 de Crescimento de Fibroblastos/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Animais , Padronização Corporal , Desenvolvimento Embrionário , Endoderma/citologia , Endoderma/embriologia , Endoderma/metabolismo , Fator 4 de Crescimento de Fibroblastos/genética , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Comunicação Parácrina/fisiologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...