Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell ; 186(21): 4710-4727.e35, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37774705

RESUMEN

Polarized cells rely on a polarized cytoskeleton to function. Yet, how cortical polarity cues induce cytoskeleton polarization remains elusive. Here, we capitalized on recently established designed 2D protein arrays to ectopically engineer cortical polarity of virtually any protein of interest during mitosis in various cell types. This enables direct manipulation of polarity signaling and the identification of the cortical cues sufficient for cytoskeleton polarization. Using this assay, we dissected the logic of the Par complex pathway, a key regulator of cytoskeleton polarity during asymmetric cell division. We show that cortical clustering of any Par complex subunit is sufficient to trigger complex assembly and that the primary kinetic barrier to complex assembly is the relief of Par6 autoinhibition. Further, we found that inducing cortical Par complex polarity induces two hallmarks of asymmetric cell division in unpolarized mammalian cells: spindle orientation, occurring via Par3, and central spindle asymmetry, depending on aPKC activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Polaridad Celular , Técnicas Citológicas , Mitosis , Animales , Citoesqueleto/metabolismo , Mamíferos/metabolismo , Microtúbulos/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
2.
Nat Rev Mol Cell Biol ; 24(1): 5, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35835877
3.
Development ; 150(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36930528

RESUMEN

The Company of Biologists' 2022 workshop on 'Cell State Transitions: Approaches, Experimental Systems and Models' brought together an international and interdisciplinary team of investigators spanning the fields of cell and developmental biology, stem cell biology, physics, mathematics and engineering to tackle the question of how cells precisely navigate between distinct identities and do so in a dynamic manner. This second edition of the workshop was organized after a successful virtual workshop on the same topic that took place in 2021.


Asunto(s)
Células Madre , Congresos como Asunto , Biología Celular , Biología Evolutiva
5.
Development ; 147(14)2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32680920

RESUMEN

Gene regulatory networks and tissue morphogenetic events drive the emergence of shape and function: the pillars of embryo development. Although model systems offer a window into the molecular biology of cell fate and tissue shape, mechanistic studies of our own development have so far been technically and ethically challenging. However, recent technical developments provide the tools to describe, manipulate and mimic human embryos in a dish, thus opening a new avenue to exploring human development. Here, I discuss the evidence that supports a role for the crosstalk between cell fate and tissue shape during early human embryogenesis. This is a critical developmental period, when the body plan is laid out and many pregnancies fail. Dissecting the basic mechanisms that coordinate cell fate and tissue shape will generate an integrated understanding of early embryogenesis and new strategies for therapeutic intervention in early pregnancy loss.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Aneuploidia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Morfogénesis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Nature ; 552(7684): 239-243, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29186120

RESUMEN

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.


Asunto(s)
Embrión de Mamíferos/citología , Morfogénesis , Células Madre Pluripotentes/citología , Amnios/citología , Animales , Tipificación del Cuerpo , Colágeno , Combinación de Medicamentos , Femenino , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Glicosilación , Células Madre Embrionarias Humanas/citología , Humanos , Laminina , Masculino , Ratones , Células Madre Embrionarias de Ratones/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteoglicanos , Sialomucinas/metabolismo , Esferoides Celulares/citología
8.
J Cell Sci ; 130(4): 683-688, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28069833

RESUMEN

Epidermal homeostasis is tightly controlled by a balancing act of self-renewal or terminal differentiation of proliferating basal keratinocytes. An increase in DNA content as a consequence of a mitotic block is a recognized mechanism underlying keratinocyte differentiation, but the molecular mechanisms involved in this process are not yet fully understood. Using cultured primary keratinocytes, here we report that the expression of the mammalian microtubule and kinetochore-associated protein Clasp2 is intimately associated with the basal proliferative makeup of keratinocytes, and its deficiency leads to premature differentiation. Clasp2-deficient keratinocytes exhibit increased centrosomal numbers and numerous mitotic alterations, including multipolar spindles and chromosomal misalignments that overall result in mitotic stress and a high DNA content. Such mitotic block prompts premature keratinocyte differentiation in a p53-dependent manner in the absence of cell death. Our findings reveal a new role for Clasp2 in governing keratinocyte undifferentiated features and highlight the presence of surveillance mechanisms that prevent cell cycle entry in cells that have alterations in the DNA content.


Asunto(s)
Diferenciación Celular , Células Epidérmicas , Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis , Animales , Línea Celular Transformada , Células Cultivadas , Daño del ADN , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/metabolismo
11.
Cell Stem Cell ; 31(10): 1398-1418, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39366361

RESUMEN

The use of stem cells to model the early human embryo promises to transform our understanding of developmental biology and human reproduction. In this review, we present our current knowledge of the first 2 weeks of human embryo development. We first focus on the distinct cell lineages of the embryo and the derivation of stem cell lines. We then discuss the intercellular crosstalk that guides early embryo development and how this crosstalk is recapitulated in vitro to generate stem cell-based embryo models. We highlight advances in this fast-developing field, discuss current limitations, and provide a vision for the future.


Asunto(s)
Desarrollo Embrionario , Humanos , Modelos Biológicos , Embrión de Mamíferos/citología , Linaje de la Célula , Células Madre/citología , Animales
12.
STAR Protoc ; 5(4): 103347, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39340775

RESUMEN

Mouse gastrulation entails concomitant changes in cell fate, tissue shape, and embryo size. The use of a reproducible in vitro system is crucial for dissecting the mechanisms that coordinate these events. Here, we present a protocol for generating a 3D culture of epiblast stem cells (3D EpiSCs), which grow as epithelial spheroids mimicking key features of the gastrulating mouse embryonic epiblast. We describe steps for spheroid formation, growth, and passaging, followed by imaging or further downstream analyses. For complete details on the use and execution of this protocol, please refer to Sato et al.1.

13.
Sci Adv ; 10(36): eadk2252, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39231227

RESUMEN

Primordial germ cells (PGCs) are the precursors of gametes and the sole mechanism by which animals transmit genetic information across generations. In the mouse embryo, the transcriptional and epigenetic regulation of PGC specification has been extensively characterized. However, the initial event that triggers the soma-germline segregation remains poorly understood. Here, we uncover a critical role for the basement membrane in regulating germline entry. We show that PGCs arise in a region of the mouse embryo that lacks contact with the basement membrane, and the addition of exogenous extracellular matrix (ECM) inhibits both PGC and PGC-like cell (PGCLC) specification in mouse embryos and stem cell models, respectively. Mechanistically, we demonstrate that the engagement of ß1 integrin with laminin blocks PGCLC specification by preventing the Wnt signaling-dependent down-regulation of the PGC transcriptional repressor, Otx2. In this way, the physical segregation of cells away from the basement membrane acts as a morphogenetic fate switch that controls the soma-germline bifurcation.


Asunto(s)
Células Germinativas , Células Madre Pluripotentes , Animales , Ratones , Células Germinativas/metabolismo , Células Germinativas/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Transducción de Señal , Integrinas/metabolismo , Integrinas/genética , Membrana Basal/metabolismo , Vía de Señalización Wnt , Diferenciación Celular , Matriz Extracelular/metabolismo , Laminina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrina beta1/metabolismo , Integrina beta1/genética , Factores de Transcripción Otx/metabolismo , Factores de Transcripción Otx/genética , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología
14.
Nat Commun ; 15(1): 7364, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39198421

RESUMEN

During the epithelial-mesenchymal transition driving mouse embryo gastrulation, cells divide more frequently at the primitive streak, and half of those divisions happen away from the apical pole. These observations suggest that non-apical mitoses might play a role in cell delamination. We aim to uncover and challenge the molecular determinants of mitosis position in different regions of the epiblast through computational modeling and pharmacological treatments of embryos and stem cell-based epiblast spheroids. Blocking basement membrane degradation at the streak has no impact on the asymmetry in mitosis frequency and position. By contrast, disturbance of the actomyosin cytoskeleton or cell cycle dynamics elicits ectopic non-apical mitosis and shows that the streak region is characterized by local relaxation of the actomyosin cytoskeleton and less stringent regulation of cell division. These factors are essential for normal dynamics at the streak and favor cell delamination from the epiblast.


Asunto(s)
Actomiosina , Gastrulación , Estratos Germinativos , Mitosis , Animales , Ratones , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Actomiosina/metabolismo , Transición Epitelial-Mesenquimal , Embrión de Mamíferos/citología , Línea Primitiva/citología , Línea Primitiva/metabolismo , Femenino , Membrana Basal/metabolismo , Citoesqueleto/metabolismo , Ciclo Celular
15.
Dev Cell ; 59(10): 1252-1268.e13, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38579720

RESUMEN

The blueprint of the mammalian body plan is laid out during gastrulation, when a trilaminar embryo is formed. This process entails a burst of proliferation, the ingression of embryonic epiblast cells at the primitive streak, and their priming toward primitive streak fates. How these different events are coordinated remains unknown. Here, we developed and characterized a 3D culture of self-renewing mouse embryonic cells that captures the main transcriptional and architectural features of the early gastrulating mouse epiblast. Using this system in combination with microfabrication and in vivo experiments, we found that proliferation-induced crowding triggers delamination of cells that express high levels of the apical polarity protein aPKC. Upon delamination, cells become more sensitive to Wnt signaling and upregulate the expression of primitive streak markers such as Brachyury. This mechanistic coupling between ingression and differentiation ensures that the right cell types become specified at the right place during embryonic development.


Asunto(s)
Diferenciación Celular , Gastrulación , Estratos Germinativos , Animales , Ratones , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Línea Primitiva/metabolismo , Proteínas Fetales/metabolismo , Proteínas Fetales/genética , Vía de Señalización Wnt , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo
16.
Nat Commun ; 15(1): 7404, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39191776

RESUMEN

Human development relies on the correct replication, maintenance and segregation of our genetic blueprints. How these processes are monitored across embryonic lineages, and why genomic mosaicism varies during development remain unknown. Using pluripotent stem cells, we identify that several patterning signals-including WNT, BMP, and FGF-converge into the modulation of DNA replication stress and damage during S-phase, which in turn controls chromosome segregation fidelity in mitosis. We show that the WNT and BMP signals protect from excessive origin firing, DNA damage and chromosome missegregation derived from stalled forks in pluripotency. Cell signalling control of chromosome segregation declines during lineage specification into the three germ layers, but re-emerges in neural progenitors. In particular, we find that the neurogenic factor FGF2 induces DNA replication stress-mediated chromosome missegregation during the onset of neurogenesis, which could provide a rationale for the elevated chromosomal mosaicism of the developing brain. Our results highlight roles for morphogens and cellular identity in genome maintenance that contribute to somatic mosaicism during mammalian development.


Asunto(s)
Segregación Cromosómica , Replicación del ADN , Neurogénesis , Neurogénesis/genética , Animales , Humanos , Ratones , Daño del ADN , Transducción de Señal , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Mitosis , Mosaicismo
17.
Cell Stem Cell ; 29(1): 8-10, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34995497

RESUMEN

Studying human embryo development is technically and ethically challenging. An improved protocol to generate human embryo-like structures (blastoids) from human pluripotent stem cells (PSCs) (Kagawa et al., 2021) offers innovative opportunities to dissect the mechanisms of human embryogenesis.


Asunto(s)
Células Madre Pluripotentes , Embrión de Mamíferos , Desarrollo Embrionario , Humanos
18.
Cell Stem Cell ; 28(5): 796-798, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33961762

RESUMEN

Over the past year, Cell Stem Cell has introduced early-career researchers impacted by the COVID-19 pandemic and subsequent closures to our readers. One year since our first introductions, we've invited several participants to reflect on their experiences and key issues. In this Story, Marta Shahbazi discusses the meaning of identity while balancing running a lab with motherhood.


Asunto(s)
COVID-19 , Pandemias , Femenino , Humanos , Madres , Investigadores , SARS-CoV-2
19.
Stem Cell Reports ; 16(5): 1182-1196, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33035465

RESUMEN

Mammalian embryogenesis is a paradigm of regulative development as mouse embryos show plasticity in the regulation of cell fate, cell number, and tissue morphogenesis. However, the mechanisms behind embryo plasticity remain largely unknown. Here, we determine how mouse embryos respond to an increase in cell numbers to regulate the timing and mechanism of embryonic morphogenesis, leading to the formation of the pro-amniotic cavity. Using embryos and embryonic stem cell aggregates of different size, we show that while pro-amniotic cavity formation in normal-sized embryos is achieved through basement membrane-induced polarization and exocytosis, cavity formation of increased-size embryos is delayed and achieved through apoptosis of cells that lack contact with the basement membrane. Importantly, blocking apoptosis, both genetically and pharmacologically, alters pro-amniotic cavity formation but does not affect size regulation in enlarged embryos. We conclude that the regulation of embryonic size and morphogenesis, albeit concomitant, have distinct molecular underpinnings.


Asunto(s)
Embrión de Mamíferos/anatomía & histología , Morfogénesis , Amnios/embriología , Animales , Apoptosis , Agregación Celular , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Tamaño de los Órganos , Factores de Tiempo
20.
Elife ; 102021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34403333

RESUMEN

Human embryogenesis entails complex signalling interactions between embryonic and extra-embryonic cells. However, how extra-embryonic cells direct morphogenesis within the human embryo remains largely unknown due to a lack of relevant stem cell models. Here, we have established conditions to differentiate human pluripotent stem cells (hPSCs) into yolk sac-like cells (YSLCs) that resemble the post-implantation human hypoblast molecularly and functionally. YSLCs induce the expression of pluripotency and anterior ectoderm markers in human embryonic stem cells (hESCs) at the expense of mesoderm and endoderm markers. This activity is mediated by the release of BMP and WNT signalling pathway inhibitors, and, therefore, resembles the functioning of the anterior visceral endoderm signalling centre of the mouse embryo, which establishes the anterior-posterior axis. Our results implicate the yolk sac in epiblast cell fate specification in the human embryo and propose YSLCs as a tool for studying post-implantation human embryo development in vitro.


Asunto(s)
Estratos Germinativos/crecimiento & desarrollo , Células Madre Pluripotentes/metabolismo , Saco Vitelino/crecimiento & desarrollo , Animales , Línea Celular , Ectodermo/crecimiento & desarrollo , Desarrollo Embrionario , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda