Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.930
Filtrar
1.
Phys Rev Lett ; 132(24): 248401, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38949349

RESUMO

Cellular Potts models are broadly applied across developmental biology and cancer research. We overcome limitations of the traditional approach, which reinterprets a modified Metropolis sampling as ad hoc dynamics, by introducing a physical timescale through Poissonian kinetics and by applying principles of stochastic thermodynamics to separate thermal and relaxation effects from athermal noise and nonconservative forces. Our method accurately describes cell-sorting dynamics in mouse-embryo development and identifies the distinct contributions of nonequilibrium processes, e.g., cell growth and active fluctuations.


Assuntos
Modelos Biológicos , Processos Estocásticos , Animais , Camundongos , Cinética , Termodinâmica , Desenvolvimento Embrionário/fisiologia , Embrião de Mamíferos/citologia
3.
Methods Mol Biol ; 2805: 171-186, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39008182

RESUMO

Biophysical factors, including changes in mechanical stiffness, have been shown to influence the morphogenesis of developing organs. There is a lack of experimental techniques, however, that can probe the mechanical properties of embryonic tissues-especially those which are not mechanically or optically accessible, such as the visceral organs of the developing mouse embryo. Here, using the embryonic kidney as a model system, we describe a method to use microindentation to quantify tissue-level regional differences in the mechanical properties of an embryonic organ. This technique is generalizable and can be used to quantify patterns of tissue stiffness within other developing organ systems. Going forward, these data will enable new experimental studies of the role of biophysical cues during organogenesis.


Assuntos
Rim , Animais , Camundongos , Rim/embriologia , Rim/citologia , Fenômenos Biomecânicos , Organogênese , Embrião de Mamíferos/citologia , Embrião de Mamíferos/fisiologia
4.
Curr Top Dev Biol ; 160: 31-64, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38937030

RESUMO

Biomechanics in embryogenesis is a dynamic field intertwining the physical forces and biological processes that shape the first days of a mammalian embryo. From the first cell fate bifurcation during blastulation to the complex symmetry breaking and tissue remodeling in gastrulation, mechanical cues appear critical in cell fate decisions and tissue patterning. Recent strides in mouse and human embryo culture, stem cell modeling of mammalian embryos, and biomaterial design have shed light on the role of cellular forces, cell polarization, and the extracellular matrix in influencing cell differentiation and morphogenesis. This chapter highlights the essential functions of biophysical mechanisms in blastocyst formation, embryo implantation, and early gastrulation where the interplay between the cytoskeleton and extracellular matrix stiffness orchestrates the intricacies of embryogenesis and placenta specification. The advancement of in vitro models like blastoids, gastruloids, and other types of embryoids, has begun to faithfully recapitulate human development stages, offering new avenues for exploring the biophysical underpinnings of early development. The integration of synthetic biology and advanced biomaterials is enhancing the precision with which we can mimic and study these processes. Looking ahead, we emphasize the potential of CRISPR-mediated genomic perturbations coupled with live imaging to uncover new mechanosensitive pathways and the application of engineered biomaterials to fine-tune the mechanical conditions conducive to embryonic development. This synthesis not only bridges the gap between experimental models and in vivo conditions to advancing fundamental developmental biology of mammalian embryogenesis, but also sets the stage for leveraging biomechanical insights to inform regenerative medicine.


Assuntos
Desenvolvimento Embrionário , Animais , Humanos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Fenômenos Biomecânicos
5.
Cells ; 13(12)2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38920627

RESUMO

Preimplantation embryo culture, pivotal in assisted reproductive technology (ART), has lagged in innovation compared to embryo selection advancements. This review examines the persisting gap between in vivo and in vitro embryo development, emphasizing the need for improved culture conditions. While in humans this gap is hardly estimated, animal models, particularly bovines, reveal clear disparities in developmental competence, cryotolerance, pregnancy and live birth rates between in vitro-produced (IVP) and in vivo-derived (IVD) embryos. Molecular analyses unveil distinct differences in morphology, metabolism, and genomic stability, underscoring the need for refining culture conditions for better ART outcomes. To this end, a deeper comprehension of oviduct physiology and embryo transport is crucial for grasping embryo-maternal interactions' mechanisms. Research on autocrine and paracrine factors, and extracellular vesicles in embryo-maternal tract interactions, elucidates vital communication networks for successful implantation and pregnancy. In vitro, confinement, and embryo density are key factors to boost embryo development. Advanced dynamic culture systems mimicking fluid mechanical stimulation in the oviduct, through vibration, tilting, and microfluidic methods, and the use of innovative softer substrates, hold promise for optimizing in vitro embryo development.


Assuntos
Técnicas de Cultura Embrionária , Embrião de Mamíferos , Animais , Humanos , Técnicas de Cultura Embrionária/métodos , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Gravidez , Feminino , Blastocisto/citologia , Blastocisto/metabolismo
6.
Nat Cell Biol ; 26(6): 868-877, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38849542

RESUMO

Despite a distinct developmental origin, extraembryonic cells in mice contribute to gut endoderm and converge to transcriptionally resemble their embryonic counterparts. Notably, all extraembryonic progenitors share a non-canonical epigenome, raising several pertinent questions, including whether this landscape is reset to match the embryonic regulation and if extraembryonic cells persist into later development. Here we developed a two-colour lineage-tracing strategy to track and isolate extraembryonic cells over time. We find that extraembryonic gut cells display substantial memory of their developmental origin including retention of the original DNA methylation landscape and resulting transcriptional signatures. Furthermore, we show that extraembryonic gut cells undergo programmed cell death and neighbouring embryonic cells clear their remnants via non-professional phagocytosis. By midgestation, we no longer detect extraembryonic cells in the wild-type gut, whereas they persist and differentiate further in p53-mutant embryos. Our study provides key insights into the molecular and developmental fate of extraembryonic cells inside the embryo.


Assuntos
Apoptose , Linhagem da Célula , Metilação de DNA , Endoderma , Regulação da Expressão Gênica no Desenvolvimento , Animais , Endoderma/citologia , Endoderma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Fagocitose , Camundongos Endogâmicos C57BL , Camundongos , Diferenciação Celular , Feminino , Desenvolvimento Embrionário , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Camundongos Transgênicos , Trato Gastrointestinal/citologia , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo
7.
Nat Commun ; 15(1): 5055, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38871742

RESUMO

The anterior-posterior axis of the mammalian embryo is laid down by the anterior visceral endoderm (AVE), an extraembryonic signaling center that is specified within the visceral endoderm. Current models posit that AVE differentiation is promoted globally by epiblast-derived Nodal signals, and spatially restricted by a BMP gradient established by the extraembryonic ectoderm. Here, we report spatially restricted AVE differentiation in bilayered embryo-like aggregates made from mouse embryonic stem cells that lack an extraembryonic ectoderm. Notably, clusters of AVE cells also form in pure visceral endoderm cultures upon activation of Nodal signaling, indicating that tissue-intrinsic factors can restrict AVE differentiation. We identify ß-catenin activity as a tissue-intrinsic factor that antagonizes AVE-inducing Nodal signals. Together, our results show how an AVE-like population can arise through interactions between epiblast and visceral endoderm alone. This mechanism may be a flexible solution for axis patterning in a wide range of embryo geometries, and provide robustness to axis patterning when coupled with signal gradients.


Assuntos
Padronização Corporal , Diferenciação Celular , Endoderma , Proteína Nodal , Transdução de Sinais , beta Catenina , Animais , Endoderma/citologia , Endoderma/metabolismo , Endoderma/embriologia , beta Catenina/metabolismo , Camundongos , Proteína Nodal/metabolismo , Proteína Nodal/genética , Camadas Germinativas/metabolismo , Camadas Germinativas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Regulação da Expressão Gênica no Desenvolvimento , Embrião de Mamíferos/citologia
8.
Nat Commun ; 15(1): 5381, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918406

RESUMO

During human embryonic development, early cleavage-stage embryos are more susceptible to errors. Studies have shown that many problems occur during the first mitosis, such as direct cleavage, chromosome segregation errors, and multinucleation. However, the mechanisms whereby these errors occur during the first mitosis in human embryos remain unknown. To clarify this aspect, in the present study, we image discarded living human two-pronuclear stage zygotes using fluorescent labeling and confocal microscopy without microinjection of DNA or mRNA and investigate the association between spindle shape and nuclear abnormality during the first mitosis. We observe that the first mitotic spindles vary, and low-aspect-ratio-shaped spindles tend to lead to the formation of multiple nuclei at the 2-cell stage. Moreover, we observe defocusing poles in many of the first mitotic spindles, which are strongly associated with multinucleation. Additionally, we show that differences in the positions of the centrosomes cause spindle abnormality in the first mitosis. Furthermore, many multinuclei are modified to form mononuclei after the second mitosis because the occurrence of pole defocusing is firmly reduced. Our study will contribute markedly to research on the occurrence of mitotic errors during the early cleavage of human embryos.


Assuntos
Núcleo Celular , Mitose , Fuso Acromático , Humanos , Fuso Acromático/metabolismo , Núcleo Celular/metabolismo , Zigoto/citologia , Zigoto/metabolismo , Embrião de Mamíferos/citologia , Microscopia Confocal , Centrossomo/metabolismo , Desenvolvimento Embrionário/fisiologia , Feminino
9.
Nature ; 630(8017): 720-727, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38839949

RESUMO

Spermatozoa harbour a complex and environment-sensitive pool of small non-coding RNAs (sncRNAs)1, which influences offspring development and adult phenotypes1-7. Whether spermatozoa in the epididymis are directly susceptible to environmental cues is not fully understood8. Here we used two distinct paradigms of preconception acute high-fat diet to dissect epididymal versus testicular contributions to the sperm sncRNA pool and offspring health. We show that epididymal spermatozoa, but not developing germ cells, are sensitive to the environment and identify mitochondrial tRNAs (mt-tRNAs) and their fragments (mt-tsRNAs) as sperm-borne factors. In humans, mt-tsRNAs in spermatozoa correlate with body mass index, and paternal overweight at conception doubles offspring obesity risk and compromises metabolic health. Sperm sncRNA sequencing of mice mutant for genes involved in mitochondrial function, and metabolic phenotyping of their wild-type offspring, suggest that the upregulation of mt-tsRNAs is downstream of mitochondrial dysfunction. Single-embryo transcriptomics of genetically hybrid two-cell embryos demonstrated sperm-to-oocyte transfer of mt-tRNAs at fertilization and suggested their involvement in the control of early-embryo transcription. Our study supports the importance of paternal health at conception for offspring metabolism, shows that mt-tRNAs are diet-induced and sperm-borne and demonstrates, in a physiological setting, father-to-offspring transfer of sperm mitochondrial RNAs at fertilization.


Assuntos
Dieta Hiperlipídica , Epigênese Genética , Mitocôndrias , RNA Mitocondrial , Espermatozoides , Animais , Feminino , Humanos , Masculino , Camundongos , Dieta Hiperlipídica/efeitos adversos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Epididimo/citologia , Epigênese Genética/genética , Fertilização/genética , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/genética , Obesidade/genética , Obesidade/metabolismo , Obesidade/etiologia , Oócitos/metabolismo , Sobrepeso/genética , Sobrepeso/metabolismo , Herança Paterna/genética , RNA Mitocondrial/genética , RNA Mitocondrial/metabolismo , Pequeno RNA não Traduzido/genética , Pequeno RNA não Traduzido/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , Espermatozoides/metabolismo , Testículo/citologia , Regulação da Expressão Gênica no Desenvolvimento , Perfilação da Expressão Gênica , Transcrição Gênica , Índice de Massa Corporal
10.
Cell ; 187(13): 3194-3219, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38906095

RESUMO

Developing functional organs from stem cells remains a challenging goal in regenerative medicine. Existing methodologies, such as tissue engineering, bioprinting, and organoids, only offer partial solutions. This perspective focuses on two promising approaches emerging for engineering human organs from stem cells: stem cell-based embryo models and interspecies organogenesis. Both approaches exploit the premise of guiding stem cells to mimic natural development. We begin by summarizing what is known about early human development as a blueprint for recapitulating organogenesis in both embryo models and interspecies chimeras. The latest advances in both fields are discussed before highlighting the technological and knowledge gaps to be addressed before the goal of developing human organs could be achieved using the two approaches. We conclude by discussing challenges facing embryo modeling and interspecies organogenesis and outlining future prospects for advancing both fields toward the generation of human tissues and organs for basic research and translational applications.


Assuntos
Quimera , Organogênese , Animais , Humanos , Quimera/embriologia , Implantação do Embrião , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Células-Tronco Embrionárias , Modelos Biológicos , Organoides , Medicina Regenerativa , Engenharia Tecidual/métodos
11.
Dev Cell ; 59(12): 1487-1488, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38889690

RESUMO

In this issue of Developmental Cell, Bolondi et al. systematically assesses neuro-mesodermal progenitor (NMP) dynamics by combining a mouse stem-cell-based embryo model with molecular recording of single cells, shedding light on the dynamics of neural tube and paraxial mesoderm formation during mammalian development.


Assuntos
Mesoderma , Animais , Camundongos , Mesoderma/citologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Tubo Neural/citologia , Tubo Neural/embriologia , Diferenciação Celular/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Padronização Corporal
13.
Nat Commun ; 15(1): 5210, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890321

RESUMO

Cell-fate decisions during mammalian gastrulation are poorly understood outside of rodent embryos. The embryonic disc of pig embryos mirrors humans, making them a useful proxy for studying gastrulation. Here we present a single-cell transcriptomic atlas of pig gastrulation, revealing cell-fate emergence dynamics, as well as conserved and divergent gene programs governing early porcine, primate, and murine development. We highlight heterochronicity in extraembryonic cell-types, despite the broad conservation of cell-type-specific transcriptional programs. We apply these findings in combination with functional investigations, to outline conserved spatial, molecular, and temporal events during definitive endoderm specification. We find early FOXA2 + /TBXT- embryonic disc cells directly form definitive endoderm, contrasting later-emerging FOXA2/TBXT+ node/notochord progenitors. Unlike mesoderm, none of these progenitors undergo epithelial-to-mesenchymal transition. Endoderm/Node fate hinges on balanced WNT and hypoblast-derived NODAL, which is extinguished upon endodermal differentiation. These findings emphasise the interplay between temporal and topological signalling in fate determination during gastrulation.


Assuntos
Embrião de Mamíferos , Endoderma , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Análise de Célula Única , Animais , Endoderma/citologia , Endoderma/metabolismo , Endoderma/embriologia , Suínos , Camundongos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Diferenciação Celular , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Transcriptoma , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Linhagem da Célula , Proteínas com Domínio T/metabolismo , Proteínas com Domínio T/genética , Transição Epitelial-Mesenquimal/genética
15.
Nat Struct Mol Biol ; 31(6): 964-976, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38789684

RESUMO

The mouse and human embryo gradually loses totipotency before diversifying into the inner cell mass (ICM, future organism) and trophectoderm (TE, future placenta). The transcription factors TFAP2C and TEAD4 with activated RHOA accelerate embryo polarization. Here we show that these factors also accelerate the loss of totipotency. TFAP2C and TEAD4 paradoxically promote and inhibit Hippo signaling before lineage diversification: they drive expression of multiple Hippo regulators while also promoting apical domain formation, which inactivates Hippo. Each factor activates TE specifiers in bipotent cells, while TFAP2C also activates specifiers of the ICM fate. Asymmetric segregation of the apical domain reconciles the opposing regulation of Hippo signaling into Hippo OFF and the TE fate, or Hippo ON and the ICM fate. We propose that the bistable switch established by TFAP2C and TEAD4 is exploited to trigger robust lineage diversification in the developing embryo.


Assuntos
Proteínas de Ligação a DNA , Fatores de Transcrição de Domínio TEA , Fator de Transcrição AP-2 , Fatores de Transcrição , Fator de Transcrição AP-2/metabolismo , Fator de Transcrição AP-2/genética , Animais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Humanos , Transdução de Sinais , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/citologia , Via de Sinalização Hippo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Desenvolvimento Embrionário/genética
16.
Nature ; 629(8012): 646-651, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38693259

RESUMO

The shaping of human embryos begins with compaction, during which cells come into close contact1,2. Assisted reproductive technology studies indicate that human embryos fail compaction primarily because of defective adhesion3,4. On the basis of our current understanding of animal morphogenesis5,6, other morphogenetic engines, such as cell contractility, could be involved in shaping human embryos. However, the molecular, cellular and physical mechanisms driving human embryo morphogenesis remain uncharacterized. Using micropipette aspiration on human embryos donated to research, we have mapped cell surface tensions during compaction. This shows a fourfold increase of tension at the cell-medium interface whereas cell-cell contacts keep a steady tension. Therefore, increased tension at the cell-medium interface drives human embryo compaction, which is qualitatively similar to compaction in mouse embryos7. Further comparison between human and mouse shows qualitatively similar but quantitively different mechanical strategies, with human embryos being mechanically least efficient. Inhibition of cell contractility and cell-cell adhesion in human embryos shows that, whereas both cellular processes are required for compaction, only contractility controls the surface tensions responsible for compaction. Cell contractility and cell-cell adhesion exhibit distinct mechanical signatures when faulty. Analysing the mechanical signature of naturally failing embryos, we find evidence that non-compacting or partially compacting embryos containing excluded cells have defective contractility. Together, our study shows that an evolutionarily conserved increase in cell contractility is required to generate the forces driving the first morphogenetic movement shaping the human body.


Assuntos
Adesão Celular , Embrião de Mamíferos , Desenvolvimento Embrionário , Animais , Feminino , Humanos , Masculino , Camundongos , Fenômenos Biomecânicos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Tensão Superficial , Adulto
17.
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
18.
Dev Cell ; 59(10): 1252-1268.e13, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38579720

RESUMO

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.


Assuntos
Diferenciação Celular , Gastrulação , Camadas Germinativas , Animais , Camundongos , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Proteínas com Domínio T/metabolismo , Proteínas com Domínio T/genética , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Linha Primitiva/citologia , Linha Primitiva/metabolismo , Proteínas Fetais/metabolismo , Proteínas Fetais/genética , Via de Sinalização Wnt , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo
19.
Dev Cell ; 59(13): 1689-1706.e8, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38636517

RESUMO

During enteric nervous system (ENS) development, pioneering wavefront enteric neural crest cells (ENCCs) initiate gut colonization. However, the molecular mechanisms guiding their specification and niche interaction are not fully understood. We used single-cell RNA sequencing and spatial transcriptomics to map the spatiotemporal dynamics and molecular landscape of wavefront ENCCs in mouse embryos. Our analysis shows a progressive decline in wavefront ENCC potency during migration and identifies transcription factors governing their specification and differentiation. We further delineate key signaling pathways (ephrin-Eph, Wnt-Frizzled, and Sema3a-Nrp1) utilized by wavefront ENCCs to interact with their surrounding cells. Disruptions in these pathways are observed in human Hirschsprung's disease gut tissue, linking them to ENS malformations. Additionally, we observed region-specific and cell-type-specific transcriptional changes in surrounding gut tissues upon wavefront ENCC arrival, suggesting their role in shaping the gut microenvironment. This work offers a roadmap of ENS development, with implications for understanding ENS disorders.


Assuntos
Movimento Celular , Sistema Nervoso Entérico , Crista Neural , Transdução de Sinais , Animais , Crista Neural/metabolismo , Crista Neural/citologia , Camundongos , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/embriologia , Sistema Nervoso Entérico/citologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/citologia , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Doença de Hirschsprung/patologia , Humanos
20.
Nat Cell Biol ; 26(5): 719-730, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38594587

RESUMO

During embryonic development, blood cells emerge from specialized endothelial cells, named haemogenic endothelial cells (HECs). As HECs are rare and only transiently found in early developing embryos, it remains difficult to distinguish them from endothelial cells. Here we performed transcriptomic analysis of 28- to 32-day human embryos and observed that the expression of Fc receptor CD32 (FCGR2B) is highly enriched in the endothelial cell population that contains HECs. Functional analyses using human embryonic and human pluripotent stem cell-derived endothelial cells revealed that robust multilineage haematopoietic potential is harboured within CD32+ endothelial cells and showed that 90% of CD32+ endothelial cells are bona fide HECs. Remarkably, these analyses indicated that HECs progress through different states, culminating in FCGR2B expression, at which point cells are irreversibly committed to a haematopoietic fate. These findings provide a precise method for isolating HECs from human embryos and human pluripotent stem cell cultures, thus allowing the efficient generation of haematopoietic cells in vitro.


Assuntos
Desenvolvimento Embrionário , Receptores de IgG , Humanos , Desenvolvimento Embrionário/genética , Receptores de IgG/metabolismo , Receptores de IgG/genética , Hemangioblastos/metabolismo , Hemangioblastos/citologia , Diferenciação Celular , Células Endoteliais/metabolismo , Células Endoteliais/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Linhagem da Célula , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Transcriptoma , Perfilação da Expressão Gênica , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...