Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Cell ; 187(13): 3224-3228, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38906097

RESUMO

The next 50 years of developmental biology will illuminate exciting new discoveries but are also poised to provide solutions to important problems society faces. Ten scientists whose work intersects with developmental biology in various capacities tell us about their vision for the future.


Assuntos
Biologia do Desenvolvimento , Biologia do Desenvolvimento/tendências , Humanos , Células-Tronco/citologia , Animais , Pesquisa com Células-Tronco
2.
Cell ; 187(11): 2838-2854.e17, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38744282

RESUMO

Retrospective lineage reconstruction of humans predicts that dramatic clonal imbalances in the body can be traced to the 2-cell stage embryo. However, whether and how such clonal asymmetries arise in the embryo is unclear. Here, we performed prospective lineage tracing of human embryos using live imaging, non-invasive cell labeling, and computational predictions to determine the contribution of each 2-cell stage blastomere to the epiblast (body), hypoblast (yolk sac), and trophectoderm (placenta). We show that the majority of epiblast cells originate from only one blastomere of the 2-cell stage embryo. We observe that only one to three cells become internalized at the 8-to-16-cell stage transition. Moreover, these internalized cells are more frequently derived from the first cell to divide at the 2-cell stage. We propose that cell division dynamics and a cell internalization bottleneck in the early embryo establish asymmetry in the clonal composition of the future human body.


Assuntos
Blastômeros , Linhagem da Célula , Embrião de Mamíferos , Feminino , Humanos , Blastômeros/citologia , Blastômeros/metabolismo , Divisão Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Masculino , Animais , Camundongos
3.
Cell ; 183(6): 1467-1478, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33306953

RESUMO

Early embryogenesis is a conserved and self-organized process. In the mammalian embryo, the potential for self-organization is manifested in its extraordinary developmental plasticity, allowing a correctly patterned embryo to arise despite experimental perturbation. The underlying mechanisms enabling such regulative development have long been a topic of study. In this Review, we summarize our current understanding of the self-organizing principles behind the regulative nature of the early mammalian embryo. We argue that geometrical constraints, feedback between mechanical and biochemical factors, and cellular heterogeneity are all required to ensure the developmental plasticity of mammalian embryo development.


Assuntos
Embrião de Mamíferos/fisiologia , Mamíferos/embriologia , Animais , Blastocisto/citologia , Padronização Corporal , Linhagem da Célula , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário
4.
Cell ; 175(7): 1902-1916.e13, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30550788

RESUMO

Nuclear architecture has never been carefully examined during early mammalian development at the stages leading to establishment of the embryonic and extra-embryonic lineages. Heterogeneous activity of the methyltransferase CARM1 during these stages results in differential methylation of histone H3R26 to modulate establishment of these two lineages. Here we show that CARM1 accumulates in nuclear granules at the 2- to 4-cell stage transition in the mouse embryo, with the majority corresponding to paraspeckles. The paraspeckle component Neat1 and its partner p54nrb are required for CARM1's association with paraspeckles and for H3R26 methylation. Conversely, CARM1 also influences paraspeckle organization. Depletion of Neat1 or p54nrb results in arrest at the 16- to 32-cell stage, with elevated expression of transcription factor Cdx2, promoting differentiation into the extra-embryonic lineage. This developmental arrest occurs at an earlier stage than following CARM1 depletion, indicating that paraspeckles act upstream of CARM1 but also have additional earlier roles in fate choice.


Assuntos
Blastocisto/metabolismo , Diferenciação Celular , Linhagem da Célula , Desenvolvimento Embrionário , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Blastocisto/citologia , Pontos de Checagem do Ciclo Celular , Camundongos , Proteínas Associadas à Matriz Nuclear/genética , Proteína-Arginina N-Metiltransferases/genética , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA/genética
5.
Cell ; 165(1): 61-74, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27015307

RESUMO

The major and essential objective of pre-implantation development is to establish embryonic and extra-embryonic cell fates. To address when and how this fundamental process is initiated in mammals, we characterize transcriptomes of all individual cells throughout mouse pre-implantation development. This identifies targets of master pluripotency regulators Oct4 and Sox2 as being highly heterogeneously expressed between blastomeres of the 4-cell embryo, with Sox21 showing one of the most heterogeneous expression profiles. Live-cell tracking demonstrates that cells with decreased Sox21 yield more extra-embryonic than pluripotent progeny. Consistently, decreasing Sox21 results in premature upregulation of the differentiation regulator Cdx2, suggesting that Sox21 helps safeguard pluripotency. Furthermore, Sox21 is elevated following increased expression of the histone H3R26-methylase CARM1 and is lowered following CARM1 inhibition, indicating the importance of epigenetic regulation. Therefore, our results indicate that heterogeneous gene expression, as early as the 4-cell stage, initiates cell-fate decisions by modulating the balance of pluripotency and differentiation.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição SOXB2/metabolismo , Animais , Blastocisto/metabolismo , Fator de Transcrição CDX2 , Epigênese Genética , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Proteínas de Homeodomínio/genética , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Análise de Célula Única , Fatores de Transcrição/genética
6.
Cell ; 156(5): 1032-44, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24529478

RESUMO

Transformation of pluripotent epiblast cells into a cup-shaped epithelium as the mouse blastocyst implants is a poorly understood and yet key developmental step. Studies of morphogenesis in embryoid bodies led to the current belief that it is programmed cell death that shapes the epiblast. However, by following embryos developing in vivo and in vitro, we demonstrate that not cell death but a previously unknown morphogenetic event transforms the amorphous epiblast into a rosette of polarized cells. This transformation requires basal membrane-stimulated integrin signaling that coordinates polarization of epiblast cells and their apical constriction, a prerequisite for lumenogenesis. We show that basal membrane function can be substituted in vitro by extracellular matrix (ECM) proteins and that ES cells can be induced to form similar polarized rosettes that initiate lumenogenesis. Together, these findings lead to a completely revised model for peri-implantation morphogenesis in which ECM triggers the self-organization of the embryo's stem cells.


Assuntos
Blastocisto/citologia , Implantação do Embrião , Embrião de Mamíferos/citologia , Camadas Germinativas/citologia , Animais , Apoptose , Blastocisto/metabolismo , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Matriz Extracelular/metabolismo , Feminino , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Morfogênese , Transdução de Sinais
7.
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
8.
Nature ; 610(7930): 143-153, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36007540

RESUMO

Embryonic stem (ES) cells can undergo many aspects of mammalian embryogenesis in vitro1-5, but their developmental potential is substantially extended by interactions with extraembryonic stem cells, including trophoblast stem (TS) cells, extraembryonic endoderm stem (XEN) cells and inducible XEN (iXEN) cells6-11. Here we assembled stem cell-derived embryos in vitro from mouse ES cells, TS cells and iXEN cells and showed that they recapitulate the development of whole natural mouse embryo in utero up to day 8.5 post-fertilization. Our embryo model displays headfolds with defined forebrain and midbrain regions and develops a beating heart-like structure, a trunk comprising a neural tube and somites, a tail bud containing neuromesodermal progenitors, a gut tube, and primordial germ cells. This complete embryo model develops within an extraembryonic yolk sac that initiates blood island development. Notably, we demonstrate that the neurulating embryo model assembled from Pax6-knockout ES cells aggregated with wild-type TS cells and iXEN cells recapitulates the ventral domain expansion of the neural tube that occurs in natural, ubiquitous Pax6-knockout embryos. Thus, these complete embryoids are a powerful in vitro model for dissecting the roles of diverse cell lineages and genes in development. Our results demonstrate the self-organization ability of ES cells and two types of extraembryonic stem cells to reconstitute mammalian development through and beyond gastrulation to neurulation and early organogenesis.


Assuntos
Embrião de Mamíferos , Gastrulação , Modelos Biológicos , Neurulação , Organogênese , Animais , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Endoderma/embriologia , Coração/embriologia , Mesencéfalo/embriologia , Camundongos , Tubo Neural/embriologia , Fator de Transcrição PAX6/deficiência , Fator de Transcrição PAX6/genética , Prosencéfalo/embriologia , Somitos/embriologia
9.
EMBO J ; 41(24): e111021, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-35993232

RESUMO

Individual cells within de novo polarising tubes and cavities must integrate their forming apical domains into a centralised apical membrane initiation site (AMIS). This is necessary to enable organised lumen formation within multi-cellular tissue. Despite the well-documented importance of cell division in localising the AMIS, we have found a division-independent mechanism of AMIS localisation that relies instead on Cadherin-mediated cell-cell adhesion. Our study of de novo polarising mouse embryonic stem cells (mESCs) cultured in 3D suggests that cell-cell adhesion localises apical proteins such as PAR-6 to a centralised AMIS. Unexpectedly, we also found that mESC clusters lacking functional E-cadherin still formed a lumen-like cavity in the absence of AMIS localisation but did so at a later stage of development via a "closure" mechanism, instead of via hollowing. This work suggests that there are two, interrelated mechanisms of apical polarity localisation: cell adhesion and cell division. Alignment of these mechanisms in space allows for redundancy in the system and ensures the development of a coherent epithelial structure within a growing organ.


Assuntos
Caderinas , Polaridade Celular , Animais , Camundongos , Caderinas/genética , Caderinas/metabolismo , Membrana Celular/metabolismo , Adesão Celular , Células Epiteliais/metabolismo
10.
Nature ; 582(7811): 253-258, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32523119

RESUMO

Tissue sculpting during development has been attributed mainly to cellular events through processes such as convergent extension or apical constriction1,2. However, recent work has revealed roles for basement membrane remodelling in global tissue morphogenesis3-5. Upon implantation, the epiblast and extraembryonic ectoderm of the mouse embryo become enveloped by a basement membrane. Signalling between the basement membrane and these tissues is critical for cell polarization and the ensuing morphogenesis6,7. However, the mechanical role of the basement membrane in post-implantation embryogenesis remains unknown. Here we demonstrate the importance of spatiotemporally regulated basement membrane remodelling during early embryonic development. Specifically, we show that Nodal signalling directs the generation and dynamic distribution of perforations in the basement membrane by regulating the expression of matrix metalloproteinases. This basement membrane remodelling facilitates embryo growth before gastrulation. The establishment of the anterior-posterior axis8,9 further regulates basement membrane remodelling by localizing Nodal signalling-and therefore the activity of matrix metalloproteinases and basement membrane perforations-to the posterior side of the embryo. Perforations on the posterior side are essential for primitive-streak extension during gastrulation by rendering the basement membrane of the prospective primitive streak more prone to breaching. Thus spatiotemporally regulated basement membrane remodelling contributes to the coordination of embryo growth, morphogenesis and gastrulation.


Assuntos
Membrana Basal/embriologia , Membrana Basal/metabolismo , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Animais , Membrana Basal/citologia , Blastocisto/citologia , Blastocisto/metabolismo , Embrião de Mamíferos/citologia , Matriz Extracelular/metabolismo , Feminino , Gástrula/embriologia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Ligantes da Sinalização Nodal/metabolismo , Linha Primitiva/citologia , Linha Primitiva/embriologia , Linha Primitiva/metabolismo
14.
Nature ; 552(7684): 239-243, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29186120

RESUMO

The foundations of mammalian development lie in a cluster of embryonic epiblast stem cells. In response to extracellular matrix signalling, these cells undergo epithelialization and create an apical surface in contact with a cavity, a fundamental event for all subsequent development. Concomitantly, epiblast cells transit through distinct pluripotent states, before lineage commitment at gastrulation. These pluripotent states have been characterized at the molecular level, but their biological importance remains unclear. Here we show that exit from an unrestricted naive pluripotent state is required for epiblast epithelialization and generation of the pro-amniotic cavity in mouse embryos. Embryonic stem cells locked in the naive state are able to initiate polarization but fail to undergo lumenogenesis. Mechanistically, exit from naive pluripotency activates an Oct4-governed transcriptional program that results in expression of glycosylated sialomucin proteins and the vesicle tethering and fusion events of lumenogenesis. Similarly, exit of epiblasts from naive pluripotency in cultured human post-implantation embryos triggers amniotic cavity formation and developmental progression. Our results add tissue-level architecture as a new criterion for the characterization of different pluripotent states, and show the relevance of transitions between these states during development of the mammalian embryo.


Assuntos
Embrião de Mamíferos/citologia , Morfogênese , Células-Tronco Pluripotentes/citologia , Âmnio/citologia , Animais , Padronização Corporal , Colágeno , Combinação de Medicamentos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Glicosilação , Células-Tronco Embrionárias Humanas/citologia , Humanos , Laminina , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Proteoglicanas , Sialomucinas/metabolismo , Esferoides Celulares/citologia
15.
Dev Biol ; 474: 82-90, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33333067

RESUMO

Breaking embryonic symmetry is an essential prerequisite to shape the initially symmetric embryo into a highly organized body plan that serves as the blueprint of the adult organism. This critical process is driven by morphogen signaling gradients that instruct anteroposterior axis specification. Despite its fundamental importance, what triggers symmetry breaking and how the signaling gradients are established in time and space in the mammalian embryo remain largely unknown. Stem cell-based in vitro models of embryogenesis offer an unprecedented opportunity to quantitatively dissect the multiple physical and molecular processes that shape the mammalian embryo. Here we review biochemical mechanisms governing early mammalian patterning in vivo and highlight recent advances to recreate this in vitro using stem cells. We discuss how the novel insights from these model systems extend previously proposed concepts to illuminate the extent to which embryonic cells have the intrinsic capability to generate specific, reproducible patterns during embryogenesis.


Assuntos
Desenvolvimento Embrionário , Morfogênese , Animais , Padronização Corporal , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Humanos , Camundongos , Transdução de Sinais , Células-Tronco/citologia
16.
Dev Biol ; 470: 84-94, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33217407

RESUMO

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


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Ectoderma/embriologia , Implantação do Embrião , Desenvolvimento Embrionário , Transdução de Sinais , Animais , Morte Celular , Linhagem da Célula , Ectoderma/citologia , Técnicas de Cultura Embrionária , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Camundongos , Morfogênese , Trofoblastos/citologia
17.
Dev Biol ; 474: 91-99, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33333069

RESUMO

Early human post-implantation development involves extensive growth combined with a series of complex morphogenetic events. The lack of precise spatial and temporal control over these processes leads to pregnancy loss. Given the ethical and technical limitations in studying the natural human embryo, alternative approaches are needed to investigate mechanisms underlying this critical stage of human development. Here, we present an overview of the different stem cells and stem cell-derived models which serve as useful, albeit imperfect, tools in understanding human embryogenesis. Current models include stem cells that represent each of the three earliest lineages: human embryonic stem cells corresponding to the epiblast, hypoblast-like stem cells and trophoblast stem cells. We also review the use of human embryonic stem cells to model complex aspects of epiblast morphogenesis and differentiation. Additionally, we propose that the combination of both embryonic and extra-embryonic stem cells to form three-dimensional embryo models will provide valuable insights into cell-cell chemical and mechanical interactions that are essential for natural embryogenesis.


Assuntos
Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/metabolismo , Células-Tronco/metabolismo , Animais , Embrião de Mamíferos/citologia , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Humanos , Células-Tronco/citologia , Trofoblastos/citologia
18.
Nature ; 533(7602): 251-4, 2016 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-27144363

RESUMO

Implantation of the blastocyst is a developmental milestone in mammalian embryonic development. At this time, a coordinated program of lineage diversification, cell-fate specification, and morphogenetic movements establishes the generation of extra-embryonic tissues and the embryo proper, and determines the conditions for successful pregnancy and gastrulation. Despite its basic and clinical importance, this process remains mysterious in humans. Here we report the use of a novel in vitro system to study the post-implantation development of the human embryo. We unveil the self-organizing abilities and autonomy of in vitro attached human embryos. We find human-specific molecular signatures of early cell lineage, timing, and architecture. Embryos display key landmarks of normal development, including epiblast expansion, lineage segregation, bi-laminar disc formation, amniotic and yolk sac cavitation, and trophoblast diversification. Our findings highlight the species-specificity of these developmental events and provide a new understanding of early human embryonic development beyond the blastocyst stage. In addition, our study establishes a new model system relevant to early human pregnancy loss. Finally, our work will also assist in the rational design of differentiation protocols of human embryonic stem cells to specific cell types for disease modelling and cell replacement therapy.


Assuntos
Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Âmnio/citologia , Âmnio/embriologia , Animais , Diferenciação Celular , Linhagem da Célula , Perda do Embrião/patologia , Embrião de Mamíferos/anatomia & histologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/patologia , Células-Tronco Embrionárias/transplante , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Humanos , Técnicas In Vitro , Camundongos , Modelos Biológicos , Especificidade da Espécie , Trofoblastos/citologia , Saco Vitelino/citologia , Saco Vitelino/embriologia
19.
Nat Methods ; 20(12): 1844-1848, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38057512
20.
Dev Biol ; 450(2): 132-140, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30940540

RESUMO

Migration of Anterior Visceral Endoderm (AVE) is a critical symmetry breaking event in the early post-implantation embryo development and is essential for establishing the correct body plan. Despite much effort, cellular and molecular events influencing AVE migration are only partially understood. Here, using time-lapse live imaging of mouse embryos, we demonstrate that cell division in the embryonic visceral endoderm is coordinated with AVE migration. Moreover, we demonstrate that temporal inhibition of FGF signalling during the pre-implantation specification of embryonic visceral endoderm perturbs cell cycle progression, thus affecting AVE migration. These findings demonstrate that coordinated cell cycle progression during the implantation stages of development is important for post-implantation morphogenesis in the mouse embryo.


Assuntos
Blastocisto/metabolismo , Ciclo Celular , Movimento Celular , Desenvolvimento Embrionário , Endoderma/embriologia , Animais , Blastocisto/citologia , Endoderma/citologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA