Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
Open Biol ; 12(11): 220193, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36382369

RESUMO

The preimplantation mammalian embryo has the potential to self-organize, allowing the formation of a correctly patterned embryo despite experimental perturbation. To better understand the mechanisms controlling the developmental plasticity of the early mouse embryo, we used chimaeras composed of an embryonic day (E)3.5 or E4.5 inner cell mass (ICM) and cleaving 8-cell embryo. We revealed that the restricted potential of the ICM can be compensated for by uncommitted 8-cell embryo-derived blastomeres, thus leading to the formation of a normal chimaeric blastocyst that can undergo full development. However, whether such chimaeras maintain developmental competence depends on the presence or specific orientation of the polarized primitive endoderm layer in the ICM component. We also demonstrated that downregulated FGFR1 and FGFR2 expression in 8-cell embryos disturbs intercellular interactions between both components and results in an inverse proportion of primitive endoderm and epiblast within the resulting ICM and abnormal embryo development. This finding suggests that FGF signalling is a key part of the regulatory mechanism that assigns cells to a given lineage and ensures the proper composition of the blastocyst, which is a prerequisite for its successful implantation in the uterus and for further development.


Assuntos
Blastocisto , Endoderma , Feminino , Camundongos , Animais , Linhagem da Célula/fisiologia , Diferenciação Celular/fisiologia , Blastocisto/metabolismo , Camadas Germinativas/fisiologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mamíferos
2.
Biochem Soc Trans ; 50(6): 1619-1631, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36398790

RESUMO

The interplay of signalling input and downstream transcriptional activity is the key molecular attribute driving the differentiation of germ layer tissue and the specification of cell lineages within each germ layer during gastrulation. This review delves into the current understanding of signalling and transcriptional control of lineage development in the germ layers of mouse embryo and non-human primate embryos during gastrulation and highlights the inter-species conservation and divergence of the cellular and molecular mechanisms of germ layer development in the human embryo.


Assuntos
Gastrulação , Camadas Germinativas , Camundongos , Animais , Linhagem da Célula , Camadas Germinativas/fisiologia , Diferenciação Celular , Embrião de Mamíferos , Mamíferos
3.
Elife ; 112022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35088714

RESUMO

The epiblast of vertebrate embryos is comprised of neural and non-neural ectoderm, with the border territory at their intersection harboring neural crest and cranial placode progenitors. Here, we a generate single-cell atlas of the developing chick epiblast from late gastrulation through early neurulation stages to define transcriptional changes in the emerging 'neural plate border' as well as other regions of the epiblast. Focusing on the border territory, the results reveal gradual establishment of heterogeneous neural plate border signatures, including novel genes that we validate by fluorescent in situ hybridization. Developmental trajectory analysis infers that segregation of neural plate border lineages only commences at early neurulation, rather than at gastrulation as previously predicted. We find that cells expressing the prospective neural crest marker Pax7 contribute to multiple lineages, and a subset of premigratory neural crest cells shares a transcriptional signature with their border precursors. Together, our results suggest that cells at the neural plate border remain heterogeneous until early neurulation, at which time progenitors become progressively allocated toward defined neural crest and placode lineages. The data also can be mined to reveal changes throughout the developing epiblast.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/embriologia , Placa Neural/embriologia , Neurulação/fisiologia , Animais , Embrião de Galinha/citologia , Galinhas/fisiologia , Camadas Germinativas/fisiologia , Hibridização in Situ Fluorescente , Fator de Transcrição PAX7/análise
4.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445681

RESUMO

Parthenogenetic embryos have been widely studied as an effective tool related to paternal and maternal imprinting genes and reproductive problems for a long time. In this study, we established a parthenogenetic epiblast-like stem cell line through culturing parthenogenetic diploid blastocysts in a chemically defined medium containing activin A and bFGF named paAFSCs. The paAFSCs expressed pluripotent marker genes and germ-layer-related genes, as well as being alkaline-phosphatase-positive, which is similar to epiblast stem cells (EpiSCs). We previously showed that advanced embryonic stem cells (ASCs) represent hypermethylated naive pluripotent embryonic stem cells (ESCs). Here, we converted paAFSCs to ASCs by replacing bFGF with bone morphogenetic protein 4 (BMP4), CHIR99021, and leukemia inhibitory factor (LIF) in a culture medium, and we obtained parthenogenetic advanced stem cells (paASCs). The paASCs showed similar morphology with ESCs and also displayed a stronger developmental potential than paAFSCs in vivo by producing chimaeras. Our study demonstrates that maternal genes could support parthenogenetic EpiSCs derived from blastocysts and also have the potential to convert primed state paAFSCs to naive state paASCs.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Partenogênese/fisiologia , Ativinas/metabolismo , Animais , Blastocisto/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Técnicas de Cultura Embrionária/métodos , Feminino , Fatores de Crescimento de Fibroblastos/farmacologia , Camadas Germinativas/metabolismo , Camadas Germinativas/fisiologia , Fator Inibidor de Leucemia/farmacologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos ICR , Células-Tronco Embrionárias Murinas/citologia , Partenogênese/genética , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia
5.
PLoS Comput Biol ; 17(3): e1008571, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33684098

RESUMO

During early mammalian embryo development, a small number of cells make robust fate decisions at particular spatial locations in a tight time window to form inner cell mass (ICM), and later epiblast (Epi) and primitive endoderm (PE). While recent single-cell transcriptomics data allows scrutinization of heterogeneity of individual cells, consistent spatial and temporal mechanisms the early embryo utilize to robustly form the Epi/PE layers from ICM remain elusive. Here we build a multiscale three-dimensional model for mammalian embryo to recapitulate the observed patterning process from zygote to late blastocyst. By integrating the spatiotemporal information reconstructed from multiple single-cell transcriptomic datasets, the data-informed modeling analysis suggests two major processes critical to the formation of Epi/PE layers: a selective cell-cell adhesion mechanism (via EphA4/EphrinB2) for fate-location coordination and a temporal attenuation mechanism of cell signaling (via Fgf). Spatial imaging data and distinct subsets of single-cell gene expression data are then used to validate the predictions. Together, our study provides a multiscale framework that incorporates single-cell gene expression datasets to analyze gene regulations, cell-cell communications, and physical interactions among cells in complex geometries at single-cell resolution, with direct application to late-stage development of embryogenesis.


Assuntos
Desenvolvimento Embrionário/genética , Camadas Germinativas , Modelos Biológicos , Transcriptoma/genética , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/fisiologia , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Camadas Germinativas/fisiologia , Camundongos , Análise de Célula Única
7.
Nat Genet ; 52(8): 819-827, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32514123

RESUMO

Mammalian cells stably maintain high levels of DNA methylation despite expressing both positive (DNMT3A/B) and negative (TET1-3) regulators. Here, we analyzed the independent and combined effects of these regulators on the DNA methylation landscape using a panel of knockout human embryonic stem cell (ESC) lines. The greatest impact on global methylation levels was observed in DNMT3-deficient cells, including reproducible focal demethylation at thousands of normally methylated loci. Demethylation depends on TET expression and occurs only when both DNMT3s are absent. Dynamic loci are enriched for hydroxymethylcytosine and overlap with subsets of putative somatic enhancers that are methylated in ESCs and can be activated upon differentiation. We observe similar dynamics in mouse ESCs that were less frequent in epiblast stem cells (EpiSCs) and scarce in somatic tissues, suggesting a conserved pluripotency-linked mechanism. Taken together, our data reveal tightly regulated competition between DNMT3s and TETs at thousands of somatic regulatory sequences within pluripotent cells.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Elementos Facilitadores Genéticos/genética , Oxigenases de Função Mista/genética , Células-Tronco Pluripotentes/fisiologia , Proteínas Proto-Oncogênicas/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , DNA Metiltransferase 3A , Células-Tronco Embrionárias/fisiologia , Epigênese Genética/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camadas Germinativas/fisiologia , Humanos , Camundongos , Camundongos Knockout
8.
Aging (Albany NY) ; 12(10): 8790-8819, 2020 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-32474458

RESUMO

Heterochronic blood sharing rejuvenates old tissues, and most of the studies on how this works focus on young plasma, its fractions, and a few youthful systemic candidates. However, it was not formally established that young blood is necessary for this multi-tissue rejuvenation. Here, using our recently developed small animal blood exchange process, we replaced half of the plasma in mice with saline containing 5% albumin (terming it a "neutral" age blood exchange, NBE) thus diluting the plasma factors and replenishing the albumin that would be diminished if only saline was used. Our data demonstrate that a single NBE suffices to meet or exceed the rejuvenative effects of enhancing muscle repair, reducing liver adiposity and fibrosis, and increasing hippocampal neurogenesis in old mice, all the key outcomes seen after blood heterochronicity. Comparative proteomic analysis on serum from NBE, and from a similar human clinical procedure of therapeutic plasma exchange (TPE), revealed a molecular re-setting of the systemic signaling milieu, interestingly, elevating the levels of some proteins, which broadly coordinate tissue maintenance and repair and promote immune responses. Moreover, a single TPE yielded functional blood rejuvenation, abrogating the typical old serum inhibition of progenitor cell proliferation. Ectopically added albumin does not seem to be the sole determinant of such rejuvenation, and levels of albumin do not decrease with age nor are increased by NBE/TPE. A model of action (supported by a large body of published data) is that significant dilution of autoregulatory proteins that crosstalk to multiple signaling pathways (with their own feedback loops) would, through changes in gene expression, have long-lasting molecular and functional effects that are consistent with our observations. This work improves our understanding of the systemic paradigms of multi-tissue rejuvenation and suggest a novel and immediate use of the FDA approved TPE for improving the health and resilience of older people.


Assuntos
Albuminas/farmacologia , Camadas Germinativas , Troca Plasmática , Plasma/fisiologia , Rejuvenescimento/fisiologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Células Cultivadas , Camadas Germinativas/citologia , Camadas Germinativas/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Solução Salina/farmacologia
9.
Proc Natl Acad Sci U S A ; 117(21): 11503-11512, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32398375

RESUMO

To explain the evolutionary origin of vertebrate teeth from odontodes, it has been proposed that competent epithelium spread into the oropharyngeal cavity via the mouth and other possible channels such as the gill slits [Huysseune et al., 2009, J. Anat. 214, 465-476]. Whether tooth formation deep inside the pharynx in extant vertebrates continues to require external epithelia has not been addressed so far. Using zebrafish we have previously demonstrated that cells derived from the periderm penetrate the oropharyngeal cavity via the mouth and via the endodermal pouches and connect to periderm-like cells that subsequently cover the entire endoderm-derived pharyngeal epithelium [Rosa et al., 2019, Sci. Rep. 9, 10082]. We now provide conclusive evidence that the epithelial component of pharyngeal teeth in zebrafish (the enamel organ) is derived from medial endoderm, as hitherto assumed based on position deep in the pharynx. Yet, dental morphogenesis starts only after the corresponding endodermal pouch (pouch 6) has made contact with the skin ectoderm, and only after periderm-like cells have covered the prospective tooth-forming endodermal epithelium. Manipulation of signaling pathways shown to adversely affect tooth development indicates they act downstream of these events. We demonstrate that pouch-ectoderm contact and the presence of a periderm-like layer are both required, but not sufficient, for tooth initiation in the pharynx. We conclude that the earliest interactions to generate pharyngeal teeth encompass those between different epithelial populations (skin ectoderm, endoderm, and periderm-like cells in zebrafish), in addition to the epithelial-mesenchymal interactions that govern the formation of all vertebrate teeth.


Assuntos
Epitélio/fisiologia , Camadas Germinativas , Odontogênese/fisiologia , Faringe/fisiologia , Dente/crescimento & desenvolvimento , Animais , Evolução Biológica , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camadas Germinativas/citologia , Camadas Germinativas/fisiologia , Transdução de Sinais/fisiologia , Peixe-Zebra
10.
Nat Cell Biol ; 22(5): 534-545, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32367046

RESUMO

Following implantation, the naive pluripotent epiblast of the mouse blastocyst generates a rosette, undergoes lumenogenesis and forms the primed pluripotent egg cylinder, which is able to generate the embryonic tissues. How pluripotency progression and morphogenesis are linked and whether intermediate pluripotent states exist remain controversial. We identify here a rosette pluripotent state defined by the co-expression of naive factors with the transcription factor OTX2. Downregulation of blastocyst WNT signals drives the transition into rosette pluripotency by inducing OTX2. The rosette then activates MEK signals that induce lumenogenesis and drive progression to primed pluripotency. Consequently, combined WNT and MEK inhibition supports rosette-like stem cells, a self-renewing naive-primed intermediate. Rosette-like stem cells erase constitutive heterochromatin marks and display a primed chromatin landscape, with bivalently marked primed pluripotency genes. Nonetheless, WNT induces reversion to naive pluripotency. The rosette is therefore a reversible pluripotent intermediate whereby control over both pluripotency progression and morphogenesis pivots from WNT to MEK signals.


Assuntos
Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Blastocisto/metabolismo , Blastocisto/fisiologia , Diferenciação Celular/fisiologia , Cromatina/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camadas Germinativas/metabolismo , Camadas Germinativas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/fisiologia , Fatores de Transcrição Otx/metabolismo , Células-Tronco Pluripotentes/metabolismo
11.
Curr Top Dev Biol ; 136: 343-375, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31959295

RESUMO

Gastrulation entails specification and formation of three embryonic germ layers-ectoderm, mesoderm and endoderm-thereby establishing the basis for the future body plan. In zebrafish embryos, germ layer specification occurs during blastula and early gastrula stages (Ho & Kimmel, 1993), a period when the main morphogenetic movements underlying gastrulation are initiated. Hence, the signals driving progenitor cell fate specification, such as Nodal ligands from the TGF-ß family, also play key roles in regulating germ layer progenitor cell segregation (Carmany-Rampey & Schier, 2001; David & Rosa, 2001; Feldman et al., 2000; Gritsman et al., 1999; Keller et al., 2008). In this review, we summarize and discuss the main signaling pathways involved in germ layer progenitor cell fate specification and segregation, specifically focusing on recent advances in understanding the interplay between mesoderm and endoderm specification and the internalization movements at the onset of zebrafish gastrulation.


Assuntos
Padronização Corporal , Embrião não Mamífero/fisiologia , Gástrula/fisiologia , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia , Animais , Blástula , Embrião não Mamífero/citologia , Gástrula/citologia , Camadas Germinativas/citologia , Camadas Germinativas/fisiologia , Transdução de Sinais , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
12.
Curr Top Dev Biol ; 136: 377-407, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31959296

RESUMO

Gastrulation is the period of development when the three germ layers, mesoderm, endoderm and ectoderm, are not only formed, but also shaped into a rudimentary body plan. An elongated anteroposterior (AP) axis is a key feature of all vertebrate body plans, and it forms during gastrulation through the highly conserved morphogenetic mechanism of convergence & extension (C&E). As the name suggests, this process requires that cells within each germ layer converge toward the dorsal midline to narrow the tissue in the mediolateral (ML) dimension and concomitantly extend it in the AP dimension. In a number of vertebrate species, C&E is driven primarily by mediolateral intercalation behavior (MIB), during which cells elongate, align, and extend protrusions in the ML direction and interdigitate between their neighbors. MIB is only one of many complex cellular mechanisms that contributes to C&E in zebrafish embryos, however, where a combination of individual cell migration, collective migration, random walk, radial intercalation, epiboly movements, and MIB all act together to shape the nascent germ layers. Each of these diverse cell movements is driven by a distinct suite of dynamic cellular properties/activities, such as actin-rich protrusions, myosin contractility, and blebbing. Here, we discuss the spatiotemporal patterns of cellular behaviors underlying C&E gastrulation movements within each germ layer of zebrafish embryos. These behaviors must be coordinated with the embryonic axes, and we highlight the roles of Planar Cell Polarity (PCP) in orienting and BMP signaling in patterning C&E cell behaviors with respect to the AP and dorsoventral axes. Finally, we address the role of GPCR signaling, extracellular matrix, and mechanical signals in coordination of C&E movements between adjacent germ layers.


Assuntos
Padronização Corporal , Embrião não Mamífero/fisiologia , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia , Animais , Embrião não Mamífero/citologia , Camadas Germinativas/citologia , Morfogênese , Transdução de Sinais , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
13.
Curr Top Dev Biol ; 136: 429-454, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31959298

RESUMO

Gastrulation is a central process in mammalian development in which a spatiotemporally coordinated series of events driven by cross-talk between adjacent embryonic and extra-embryonic tissues results in stereotypical morphogenetic cell behaviors, massive cell proliferation and the acquisition of distinct cell identities. Gastrulation provides the blueprint of the body plan of the embryo, as well as generating extra-embryonic cell types of the embryo to make a connection with its mother. Gastrulation involves the specification of mesoderm and definitive endoderm from pluripotent epiblast, concomitant with a highly ordered elongation of tissue along the anterior-posterior (AP) axis. Interestingly, cells with an endoderm identity arise twice during mouse development. Cells with a primitive endoderm identity are specified in the preimplantation blastocyst, and which at gastrulation intercalate with the emergent definitive endoderm to form a mosaic tissue, referred to as the gut endoderm. The gut endoderm gives rise to the gut tube, which will subsequently become patterned along its AP axis into domains possessing unique visceral organ identities, such as thyroid, lung, liver and pancreas. In this way, proper endoderm development is essential for vital organismal functions, including the absorption of nutrients, gas exchange, detoxification and glucose homeostasis.


Assuntos
Embrião de Mamíferos/fisiologia , Endoderma/fisiologia , Trato Gastrointestinal/fisiologia , Gastrulação , Camadas Germinativas/fisiologia , Mesoderma/fisiologia , Morfogênese , Animais , Embrião de Mamíferos/citologia , Endoderma/citologia , Trato Gastrointestinal/citologia , Camadas Germinativas/citologia , Mesoderma/citologia , Camundongos
14.
Nat Cell Biol ; 21(12): 1518-1531, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31792383

RESUMO

The first lineage specification of pluripotent mouse epiblast segregates neuroectoderm (NE) from mesoderm and definitive endoderm (ME) by mechanisms that are not well understood. Here we demonstrate that the induction of ME gene programs critically relies on the T-box transcription factors Eomesodermin (also known as Eomes) and Brachyury, which concomitantly repress pluripotency and NE gene programs. Cells deficient in these T-box transcription factors retain pluripotency and differentiate to NE lineages despite the presence of ME-inducing signals transforming growth factor ß (TGF-ß)/Nodal and Wnt. Pluripotency and NE gene networks are additionally repressed by ME factors downstream of T-box factor induction, demonstrating a redundancy in program regulation to safeguard mutually exclusive lineage specification. Analyses of chromatin revealed that accessibility of ME enhancers depends on T-box factor binding, whereas NE enhancers are accessible and already activation primed at pluripotency. This asymmetry of the chromatin landscape thus explains the default differentiation of pluripotent cells to NE in the absence of ME induction that depends on activating and repressive functions of Eomes and Brachyury.


Assuntos
Cromatina/genética , Proteínas Fetais/genética , Camadas Germinativas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Proteínas com Domínio T/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Separação Celular/métodos , Endoderma/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Masculino , Camundongos , Placa Neural/fisiologia , Fator de Crescimento Transformador beta/genética
15.
Development ; 146(24)2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31740534

RESUMO

Embryonic stem cells (ESCs) exist in at least two states that transcriptionally resemble different stages of embryonic development. Naïve ESCs resemble peri-implantation stages and primed ESCs the pre-gastrulation epiblast. In mouse, primed ESCs give rise to definitive endoderm in response to the pathways downstream of Nodal and Wnt signalling. However, when these pathways are activated in naïve ESCs, they differentiate to a cell type resembling early primitive endoderm (PrE), the blastocyst-stage progenitor of the extra-embryonic endoderm. Here, we apply this context dependency to human ESCs, showing that activation of Nodal and Wnt signalling drives the differentiation of naïve pluripotent cells toward extra-embryonic PrE, or hypoblast, and these can be expanded as an in vitro model for naïve extra-embryonic endoderm (nEnd). Consistent with observations made in mouse, human PrE differentiation is dependent on FGF signalling in vitro, and we show that, by inhibiting FGF receptor signalling, we can simplify naïve pluripotent culture conditions, such that the inhibitor requirements closer resemble those used in mouse. The expandable nEnd cultures reported here represent stable extra-embryonic endoderm, or human hypoblast, cell lines.This article has an associated 'The people behind the papers' interview.


Assuntos
Endoderma/embriologia , Fator Inibidor de Leucemia/fisiologia , Ligantes da Sinalização Nodal/fisiologia , Células-Tronco Pluripotentes/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Endoderma/citologia , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camadas Germinativas/fisiologia , Humanos , Fator Inibidor de Leucemia/metabolismo , Camundongos , Ligantes da Sinalização Nodal/metabolismo , Transdução de Sinais/fisiologia
16.
Development ; 146(21)2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31601548

RESUMO

A switch from E- to N-cadherin regulates the transition from pluripotency to neural identity, but the mechanism by which cadherins regulate differentiation was previously unknown. Here, we show that the acquisition of N-cadherin stabilises neural identity by dampening anti-neural signals. We use quantitative image analysis to show that N-cadherin promotes neural differentiation independently of its effects on cell cohesiveness. We reveal that cadherin switching diminishes the level of nuclear ß-catenin, and that N-cadherin also dampens FGF activity and consequently stabilises neural fate. Finally, we compare the timing of cadherin switching and differentiation in vivo and in vitro, and find that this process becomes dysregulated during in vitro differentiation. We propose that N-cadherin helps to propagate a stable neural identity throughout the emerging neuroepithelium, and that dysregulation of this process contributes to asynchronous differentiation in culture.


Assuntos
Caderinas/fisiologia , Células-Tronco Embrionárias/citologia , Neurônios/citologia , beta Catenina/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Núcleo Celular/fisiologia , Células Cultivadas , Fatores de Crescimento de Fibroblastos/fisiologia , Camadas Germinativas/fisiologia , Camundongos , Camundongos Transgênicos , Células-Tronco Pluripotentes/citologia
17.
Cell Stem Cell ; 25(3): 388-406.e8, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31422912

RESUMO

Understanding how cell identity transitions occur and whether there are multiple paths between the same beginning and end states are questions of wide interest. Here we show that acquisition of naive pluripotency can follow transcriptionally and mechanistically distinct routes. Starting from post-implantation epiblast stem cells (EpiSCs), one route advances through a mesodermal state prior to naive pluripotency induction, whereas another transiently resembles the early inner cell mass and correspondingly gains greater developmental potency. These routes utilize distinct signaling networks and transcription factors but subsequently converge on the same naive endpoint, showing surprising flexibility in mechanisms underlying identity transitions and suggesting that naive pluripotency is a multidimensional attractor state. These route differences are reconciled by precise expression of Oct4 as a unifying, essential, and sufficient feature. We propose that fine-tuned regulation of this "transition factor" underpins multidimensional access to naive pluripotency, offering a conceptual framework for understanding cell identity transitions.


Assuntos
Massa Celular Interna do Blastocisto/fisiologia , Camadas Germinativas/fisiologia , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Plasticidade Celular , Reprogramação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Camundongos , Camundongos Endogâmicos C57BL , Fator 3 de Transcrição de Octâmero/genética , Transdução de Sinais
18.
Mol Hum Reprod ; 25(9): 519-526, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31211841

RESUMO

Our current knowledge of the mechanisms leading to human primordial germ cell (PGC) specification stems solely from differentiation experiments starting from human pluripotent stem cells. However, information regarding the origin of PGCs in vivo remains obscure. Here we apply an improved system for extended in vitro culture of human embryos to investigate the presence of PGC-like cells (PGCLCs) 12 days post fertilization (dpf). Good quality blastocysts (n = 141) were plated at 6 dpf and maintained in hypoxia, in medium supplemented with Activin A until 12 dpf. We primarily reveal that 12 dpf outgrowths recapitulate human peri-implantation events and demonstrate that blastocyst quality significantly impacts both embryo viability at 12 dpf, as well as the presence of POU5F1+ cells within viable outgrowths. Moreover, detailed examination of 12 dpf blastocyst outgrowths revealed a population of POU5F1+, SOX2- and SOX17+ cells that may correspond to PGCLCs, alongside POU5F1+ epiblast-like cells and GATA6+ endoderm-like cells. Our findings suggest that, in human, PGC precursors may become specified within the epiblast and migrate either transiently to the extra-embryonic mesoderm or directly to the dorsal part of the yolk sac endoderm around 12 dpf. This is a descriptive analysis and as such the conclusion that POU5F1+ and SOX17+ cells represent bona fide PGCs can only be considered as preliminary. In the future, other PGC markers may be used to further validate the observed cell populations. Overall, our findings provide insights into the origin of the human germline and may serve as a foundation to further unravel the molecular mechanisms governing PGC specification in human.


Assuntos
Blastocisto/citologia , Blastocisto/fisiologia , Linhagem da Célula/fisiologia , Células Germinativas/citologia , Células Germinativas/fisiologia , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Técnicas de Cultura Embrionária , Implantação do Embrião/fisiologia , Embrião de Mamíferos , Camadas Germinativas/citologia , Camadas Germinativas/fisiologia , Humanos , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Pseudópodes/fisiologia
19.
Nat Commun ; 10(1): 2175, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31092818

RESUMO

Clinical applications of human induced pluripotent stem cells (hiPSCs) are expected, but hiPSC lines vary in their differentiation propensity. For efficient selection of hiPSC lines suitable for differentiation into desired cell lineages, here we identify SALL3 as a marker to predict differentiation propensity. SALL3 expression in hiPSCs correlates positively with ectoderm differentiation capacity and negatively with mesoderm/endoderm differentiation capacity. Without affecting self-renewal of hiPSCs, SALL3 knockdown inhibits ectoderm differentiation and conversely enhances mesodermal/endodermal differentiation. Similarly, loss- and gain-of-function studies reveal that SALL3 inversely regulates the differentiation of hiPSCs into cardiomyocytes and neural cells. Mechanistically, SALL3 modulates DNMT3B function and DNA methyltransferase activity, and influences gene body methylation of Wnt signaling-related genes in hiPSCs. These findings suggest that SALL3 switches the differentiation propensity of hiPSCs toward distinct cell lineages by changing the epigenetic profile and serves as a marker for evaluating the hiPSC differentiation propensity.


Assuntos
Linhagem da Célula/fisiologia , Camadas Germinativas/fisiologia , Proteínas de Homeodomínio/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Fatores de Transcrição/fisiologia , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos
20.
Stem Cell Reports ; 12(2): 305-318, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30713040

RESUMO

Although pluripotent stem cells can generate various types of differentiated cells, it is unclear why lineage-committed stem/progenitor cells derived from pluripotent stem cells are decelerated and why the differentiation-resistant propensity of embryonic stem cell (ESC)/induced pluripotent stem cell (iPSC)-derived cells is predominant compared with the in vivo equivalents derived from embryonic/adult tissues. In this study, we demonstrated that iPSCs reprogrammed and maintained with three chemical inhibitors of the fibroblast growth factor 4-mitogen-activated protein kinase cascade and GSK3ß (3i) could be differentiated into all three germ layers more efficiently than the iPSCs reprogrammed without the 3i chemicals, even though they were maintained with 3i chemicals once they were reprogrammed. Although the iPSCs reprogrammed with 3i had increased numbers of Zscan4-positive cells, the Zscan4-positive cells among iPSCs that were reprogrammed without 3i did not have an accelerated differentiation ability. These observations suggest that 3i exposure during the reprogramming period determines the accelerated differentiation/maturation potentials of iPSCs that are stably maintained at the distinct state.


Assuntos
Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Animais , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Fator 4 de Crescimento de Fibroblastos/metabolismo , Camadas Germinativas/metabolismo , Camadas Germinativas/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...