Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Cell Syst ; 15(5): 445-461.e4, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38692274

RESUMEN

BMP signaling is essential for mammalian gastrulation, as it initiates a cascade of signals that control self-organized patterning. As development is highly dynamic, it is crucial to understand how time-dependent combinatorial signaling affects cellular differentiation. Here, we show that BMP signaling duration is a crucial control parameter that determines cell fates upon the exit from pluripotency through its interplay with the induced secondary signal WNT. BMP signaling directly converts cells from pluripotent to extraembryonic fates while simultaneously upregulating Wnt signaling, which promotes primitive streak and mesodermal specification. Using live-cell imaging of signaling and cell fate reporters together with a simple mathematical model, we show that this circuit produces a temporal morphogen effect where, once BMP signal duration is above a threshold for differentiation, intermediate and long pulses of BMP signaling produce specification of mesoderm and extraembryonic fates, respectively. Our results provide a systems-level picture of how these signaling pathways control the landscape of early human development.


Asunto(s)
Proteínas Morfogenéticas Óseas , Diferenciación Celular , Línea Primitiva , Transducción de Señal , Línea Primitiva/metabolismo , Línea Primitiva/embriología , Proteínas Morfogenéticas Óseas/metabolismo , Humanos , Transducción de Señal/fisiología , Animales , Mesodermo/metabolismo , Mesodermo/embriología , Vía de Señalización Wnt/fisiología , Proteínas Wnt/metabolismo , Regulación del Desarrollo de la Expresión Génica , Gastrulación/fisiología
2.
bioRxiv ; 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38559061

RESUMEN

The Wnt pathway is essential for inducing the primitive streak, the precursor of the mesendoderm, as well as setting anterior-posterior coordinates. How Wnt coordinates these diverse activities remains incompletely understood. Here, we show that in Wnt-treated human pluripotent cells, endogenous Nodal signaling is a crucial switch between posteriorizing and primitive streak-including activities. While treatment with Wnt posteriorizes cells in standard culture, in micropatterned colonies, higher levels of endogenously induced Nodal signaling combine with exogenous Wnt to drive endoderm differentiation. Inhibition of Nodal signaling restores dose-dependent posteriorization by Wnt. In the absence of Nodal inhibition, micropatterned colonies undergo spontaneous, elaborate morphogenesis concomitant with endoderm differentiation even in the absence of added extracellular matrix proteins like Matrigel. Our study shows how Wnt and Nodal combinatorially coordinate germ layer differentiation with AP patterning and establishes a system to study a natural self-organizing morphogenetic event in in vitro culture.

4.
Nat Commun ; 13(1): 6681, 2022 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-36335122

RESUMEN

Transitioning from pluripotency to differentiated cell fates is fundamental to both embryonic development and adult tissue homeostasis. Improving our understanding of this transition would facilitate our ability to manipulate pluripotent cells into tissues for therapeutic use. Here, we show that membrane voltage (Vm) regulates the exit from pluripotency and the onset of germ layer differentiation in the embryo, a process that affects both gastrulation and left-right patterning. By examining candidate genes of congenital heart disease and heterotaxy, we identify KCNH6, a member of the ether-a-go-go class of potassium channels that hyperpolarizes the Vm and thus limits the activation of voltage gated calcium channels, lowering intracellular calcium. In pluripotent embryonic cells, depletion of kcnh6 leads to membrane depolarization, elevation of intracellular calcium levels, and the maintenance of a pluripotent state at the expense of differentiation into ectodermal and myogenic lineages. Using high-resolution temporal transcriptome analysis, we identify the gene regulatory networks downstream of membrane depolarization and calcium signaling and discover that inhibition of the mTOR pathway transitions the pluripotent cell to a differentiated fate. By manipulating Vm using a suite of tools, we establish a bioelectric pathway that regulates pluripotency in vertebrates, including human embryonic stem cells.


Asunto(s)
Células Madre Pluripotentes , Animales , Humanos , Calcio/metabolismo , Potenciales de la Membrana , Diferenciación Celular/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo
5.
PLoS Comput Biol ; 18(6): e1009846, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35696439

RESUMEN

We introduce cytoNet, a cloud-based tool to characterize cell populations from microscopy images. cytoNet quantifies spatial topology and functional relationships in cell communities using principles of network science. Capturing multicellular dynamics through graph features, cytoNet also evaluates the effect of cell-cell interactions on individual cell phenotypes. We demonstrate cytoNet's capabilities in four case studies: 1) characterizing the temporal dynamics of neural progenitor cell communities during neural differentiation, 2) identifying communities of pain-sensing neurons in vivo, 3) capturing the effect of cell community on endothelial cell morphology, and 4) investigating the effect of laminin α4 on perivascular niches in adipose tissue. The analytical framework introduced here can be used to study the dynamics of complex cell communities in a quantitative manner, leading to a deeper understanding of environmental effects on cellular behavior. The versatile, cloud-based format of cytoNet makes the image analysis framework accessible to researchers across domains.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Células-Madre Neurales , Procesamiento de Imagen Asistido por Computador/métodos , Neuronas , Análisis Espacio-Temporal
6.
Nat Commun ; 13(1): 497, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35079017

RESUMEN

Morphogens are signaling molecules that convey positional information and dictate cell fates during development. Although ectopic expression in model organisms suggests that morphogen gradients form through diffusion, little is known about how morphogen gradients are created and interpreted during mammalian embryogenesis due to the combined difficulties of measuring endogenous morphogen levels and observing development in utero. Here we take advantage of a human gastruloid model to visualize endogenous Nodal protein in living cells, during specification of germ layers. We show that Nodal is extremely short range so that Nodal protein is limited to the immediate neighborhood of source cells. Nodal activity spreads through a relay mechanism in which Nodal production induces neighboring cells to transcribe Nodal. We further show that the Nodal inhibitor Lefty, while biochemically capable of long-range diffusion, also acts locally to control the timing of Nodal spread and therefore of mesoderm differentiation during patterning. Our study establishes a paradigm for tissue patterning by an activator-inhibitor pair.


Asunto(s)
Blastocisto/metabolismo , Gástrula/metabolismo , Gastrulación/genética , Células Madre Embrionarias Humanas/metabolismo , Proteína Nodal/genética , Blastocisto/citología , Línea Celular , Difusión , Técnica del Anticuerpo Fluorescente/métodos , Gástrula/citología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Células Madre Embrionarias Humanas/citología , Humanos , Hibridación Fluorescente in Situ/métodos , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Proteína Nodal/metabolismo
7.
J Math Biol ; 83(5): 55, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34727234

RESUMEN

In this paper we consider mathematical modeling of the dynamics of self-organized patterning of spatially confined human embryonic stem cells (hESCs) treated with BMP4 (gastruloids) described in recent experimental works (Warmflash in Nat Methods 11:847-854, 2014; Chhabra in PloS Biol 17: 3000498, 2019). In the first part of the paper we use the activator-inhibitor equations of Gierer and Meinhardt to identify 3 reaction-diffusion regimes for each of the three morphogenic proteins, BMP4, Wnt and Nodal, based on the characteristic features of the dynamic patterning. We identify appropriate boundary conditions which correspond to the experimental setup and perform numerical simulations of the reaction-diffusion (RD) systems, using the finite element approximation, to confirm that the RD systems in these regimes produce realistic dynamics of the protein concentrations. In the second part of the paper we use analytic tools to address the questions of the existence and stability of non-homogeneous steady states for the reaction-diffusion systems of the type considered in the first part of the paper.


Asunto(s)
Células Madre Embrionarias Humanas , Difusión , Humanos , Modelos Biológicos , Morfogénesis
8.
Nature ; 599(7884): 268-272, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34707290

RESUMEN

Understanding human organ formation is a scientific challenge with far-reaching medical implications1,2. Three-dimensional stem-cell cultures have provided insights into human cell differentiation3,4. However, current approaches use scaffold-free stem-cell aggregates, which develop non-reproducible tissue shapes and variable cell-fate patterns. This limits their capacity to recapitulate organ formation. Here we present a chip-based culture system that enables self-organization of micropatterned stem cells into precise three-dimensional cell-fate patterns and organ shapes. We use this system to recreate neural tube folding from human stem cells in a dish. Upon neural induction5,6, neural ectoderm folds into a millimetre-long neural tube covered with non-neural ectoderm. Folding occurs at 90% fidelity, and anatomically resembles the developing human neural tube. We find that neural and non-neural ectoderm are necessary and sufficient for folding morphogenesis. We identify two mechanisms drive folding: (1) apical contraction of neural ectoderm, and (2) basal adhesion mediated via extracellular matrix synthesis by non-neural ectoderm. Targeting these two mechanisms using drugs leads to morphological defects similar to neural tube defects. Finally, we show that neural tissue width determines neural tube shape, suggesting that morphology along the anterior-posterior axis depends on neural ectoderm geometry in addition to molecular gradients7. Our approach provides a new route to the study of human organ morphogenesis in health and disease.


Asunto(s)
Morfogénesis , Tubo Neural/anatomía & histología , Tubo Neural/embriología , Técnicas de Cultivo de Órganos/métodos , Ectodermo/citología , Ectodermo/embriología , Humanos , Modelos Biológicos , Placa Neural/citología , Placa Neural/embriología , Tubo Neural/citología , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología , Regeneración , Células Madre/citología
9.
Biol Open ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34435204

RESUMEN

Human embryonic stem cells (hESCs) possess an immense potential to generate clinically relevant cell types and unveil mechanisms underlying early human development. However, using hESCs for discovery or translation requires accurately identifying differentiated cell types through comparison with their in vivo counterparts. Here, we set out to determine the identity of much debated BMP-treated hESCs by comparing their transcriptome to recently published single cell transcriptomic data from early human embryos ( Xiang et al., 2020). Our analyses reveal several discrepancies in the published human embryo dataset, including misclassification of putative amnion, intermediate and inner cell mass cells. These misclassifications primarily resulted from similarities in pseudogene expression, highlighting the need to carefully consider gene lists when making comparisons between cell types. In the absence of a relevant human dataset, we utilized the recently published single cell transcriptome of the early post implantation monkey embryo to discern the identity of BMP-treated hESCs. Our results suggest that BMP-treated hESCs are transcriptionally more similar to amnion cells than trophectoderm cells in the monkey embryo. Together with prior studies, this result indicates that hESCs possess a unique ability to form mature trophectoderm subtypes via an amnion-like transcriptional state. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Amnios/citología , Embrión de Mamíferos/citología , Desarrollo Embrionario/fisiología , Células Madre Embrionarias Humanas/citología , Transcriptoma/fisiología , Animales , Proteína Morfogenética Ósea 4/administración & dosificación , Regulación del Desarrollo de la Expresión Génica , Haplorrinos , Humanos
10.
Cells Tissues Organs ; 210(3): 151-172, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34218225

RESUMEN

Epithelial to mesenchymal transition (EMT) is a highly conserved cellular process in several species, from worms to humans. EMT plays a fundamental role in early embryogenesis, wound healing, and cancer metastasis. For neural crest cell (NCC) development, EMT typically results in forming a migratory and potent cell population that generates a wide variety of cell and tissue, including cartilage, bone, connective tissue, endocrine cells, neurons, and glia amongst many others. The degree of conservation between the signaling pathways that regulate EMT during development and metastatic cancer (MC) has not been fully established, despite ample studies. This systematic review and meta-analysis dissects the major signaling pathways involved in EMT of NCC development and MC to unravel the similarities and differences. While the FGF, TGFß/BMP, SHH, and NOTCH pathways have been rigorously investigated in both systems, the EGF, IGF, HIPPO, Factor Receptor Superfamily, and their intracellular signaling cascades need to be the focus of future NCC studies. In general, meta-analyses of the associated signaling pathways show a significant number of overlapping genes (particularly ligands, transcription regulators, and targeted cadherins) involved in each signaling pathway of both systems without stratification by body segments and cancer type. Lack of stratification makes it difficult to meaningfully evaluate the intracellular downstream effectors of each signaling pathway. Finally, pediatric neuroblastoma and melanoma are NCC-derived malignancies, which emphasize the importance of uncovering the EMT events that convert NCC into treatment-resistant malignant cells.


Asunto(s)
Neoplasias , Cresta Neural , Movimiento Celular , Niño , Transición Epitelial-Mesenquimal , Humanos , Neoplasias/genética , Factor de Crecimiento Transformador beta
11.
PLoS Comput Biol ; 17(6): e1009034, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34061834

RESUMEN

Increasing interest has emerged in new mathematical approaches that simplify the study of complex differentiation processes by formalizing Waddington's landscape metaphor. However, a rational method to build these landscape models remains an open problem. Here we study vulval development in C. elegans by developing a framework based on Catastrophe Theory (CT) and approximate Bayesian computation (ABC) to build data-fitted landscape models. We first identify the candidate qualitative landscapes, and then use CT to build the simplest model consistent with the data, which we quantitatively fit using ABC. The resulting model suggests that the underlying mechanism is a quantifiable two-step decision controlled by EGF and Notch-Delta signals, where a non-vulval/vulval decision is followed by a bistable transition to the two vulval states. This new model fits a broad set of data and makes several novel predictions.


Asunto(s)
Caenorhabditis elegans/citología , Modelos Biológicos , Animales , Teorema de Bayes , Diferenciación Celular , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Proyectos de Investigación , Vulva/crecimiento & desarrollo
12.
PLoS One ; 16(6): e0245634, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34077425

RESUMEN

Self-organized patterning of mammalian embryonic stem cells on micropatterned surfaces has previously been established as an in vitro platform for early mammalian developmental studies, complimentary to in vivo studies. Traditional micropatterning methods, such as micro-contact printing (µCP), involve relatively complicated fabrication procedures, which restricts widespread adoption by biologists. Here, we demonstrate a rapid method of micropatterning by printing hydrogel micro-features onto a glass-bottomed culture vessel. The micro-features are printed using a projection stereolithography bioprinter yielding hydrogel structures that geometrically restrict the attachment of cells or proteins. Compared to traditional and physical photomasks, a digitally tunable virtual photomask is used in the projector to generate blue light patterns that enable rapid iteration with minimal cost and effort. We show that a protocol that makes use of this method together with LN521 coating, an extracellular matrix coating, creates a surface suitable for human embryonic stem cell (hESC) attachment and growth with minimal non-specific adhesion. We further demonstrate that self-patterning of hESCs following previously published gastrulation and ectodermal induction protocols achieves results comparable with those obtained with commercially available plates.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Hidrogeles/química , Microtecnología/métodos , Estereolitografía/instrumentación , Células Madre Embrionarias Humanas/fisiología , Humanos , Propiedades de Superficie
13.
Stem Cell Reports ; 16(5): 1065-1077, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979594

RESUMEN

Mammalian embryonic development is a complex process driven by self-organization. Understanding how a fertilized egg develops into an embryo composed of more than 200 cell types in precise spatial patterns remains one of the fundamental challenges in biology. Pluripotent stem cells have been used as in vitro models for investigating mammalian development, and represent promising building blocks for regenerative therapies. Recently, sophisticated stem cell-based models that recapitulate early embryonic fate patterning and morphogenesis have been developed. In this article, we review recent advances in stem cell models of embryos in particular focusing on signaling activities underpinning cell fate decisions in space and time.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Modelos Biológicos , Transducción de Señal , Animales , Cuerpos Embrioides/metabolismo , Desarrollo Embrionario , Humanos
14.
Stem Cell Reports ; 16(5): 1014-1020, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33770498

RESUMEN

Stem cell-based models of embryos are known by various names, with different naming conventions, leading to confusion regarding their composition and potential. We propose the need for a general term for the field to promote public engagement and the development of a systematic nomenclature system to differentiate between specific models.


Asunto(s)
Embrión de Mamíferos/citología , Embrión no Mamífero/citología , Modelos Biológicos , Células Madre/citología , Animales , Cuerpos Embrioides/citología , Humanos , Terminología como Asunto
15.
Methods Mol Biol ; 2258: 119-130, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33340358

RESUMEN

In the developing mammalian embryo, intercellular signaling allows cells to self-organize to create spatial patterns of different cell fates. This process is challenging to study because of the difficulty of observing or manipulating embryos on the spatial and temporal scales required. In vitro models can provide a complement to in vivo systems for addressing these issues. These models are also the only windows we have into early human development. Here we provide protocols for two systems based on differentiating human pluripotent stem cells in micropatterned colonies on defined size and shape. The first model replicates the patterning of the germ layers at gastrulation, while the second replicates the medial-lateral patterning of the ectoderm. These systems allow study of how signaling underlies self-organized patterning at stages of development which are otherwise inaccessible.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Ectodermo/fisiología , Gastrulación , Células Madre Embrionarias Humanas/fisiología , Comunicación Celular , Forma de la Célula , Tamaño de la Célula , Células Cultivadas , Ectodermo/citología , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Humanos , Microscopía Fluorescente , Transducción de Señal , Factores de Tiempo
16.
Nat Mater ; 20(2): 132-144, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33199861

RESUMEN

Despite its importance, understanding the early phases of human development has been limited by availability of human samples. The recent emergence of stem-cell-derived embryo models, a new field aiming to use stem cells to construct in vitro models to recapitulate snapshots of the development of the mammalian conceptus, opens up exciting opportunities to promote fundamental understanding of human development and advance reproductive and regenerative medicine. This Review provides a summary of the current knowledge of early mammalian development, using mouse and human conceptuses as models, and emphasizes their similarities and critical differences. We then highlight existing embryo models that mimic different aspects of mouse and human development. We further discuss bioengineering tools used for controlling multicellular interactions and self-organization critical for the development of these models. We conclude with a discussion of the important next steps and exciting future opportunities of stem-cell-derived embryo models for fundamental discovery and translation.


Asunto(s)
Embrión de Mamíferos/embriología , Células Madre Embrionarias Humanas/metabolismo , Modelos Biológicos , Células Madre Embrionarias de Ratones/metabolismo , Animales , Embrión de Mamíferos/citología , Células Madre Embrionarias Humanas/citología , Humanos , Ratones , Células Madre Embrionarias de Ratones/citología
17.
Phys Biol ; 18(4)2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33276350

RESUMEN

The way in which interactions between mechanics and biochemistry lead to the emergence of complex cell and tissue organization is an old question that has recently attracted renewed interest from biologists, physicists, mathematicians and computer scientists. Rapid advances in optical physics, microscopy and computational image analysis have greatly enhanced our ability to observe and quantify spatiotemporal patterns of signalling, force generation, deformation, and flow in living cells and tissues. Powerful new tools for genetic, biophysical and optogenetic manipulation are allowing us to perturb the underlying machinery that generates these patterns in increasingly sophisticated ways. Rapid advances in theory and computing have made it possible to construct predictive models that describe how cell and tissue organization and dynamics emerge from the local coupling of biochemistry and mechanics. Together, these advances have opened up a wealth of new opportunities to explore how mechanochemical patterning shapes organismal development. In this roadmap, we present a series of forward-looking case studies on mechanochemical patterning in development, written by scientists working at the interface between the physical and biological sciences, and covering a wide range of spatial and temporal scales, organisms, and modes of development. Together, these contributions highlight the many ways in which the dynamic coupling of mechanics and biochemistry shapes biological dynamics: from mechanoenzymes that sense force to tune their activity and motor output, to collectives of cells in tissues that flow and redistribute biochemical signals during development.


Asunto(s)
Fenómenos Biomecánicos , Morfogénesis , Transducción de Señal , Modelos Biológicos
18.
Curr Top Dev Biol ; 137: 279-305, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32143746

RESUMEN

Morphogens play an essential role in cell fate specification and patterning including in laying out the mammalian body plan during gastrulation. In vivo studies have shed light on the signaling pathways involved in this process and the phenotypes associated with their disruption, however, several important open questions remain regarding how morphogens function in space and time. Self-organized patterning systems based on embryonic stem cells have emerged as a powerful platform for beginning to address these questions that is complementary to in vivo approaches. Here we review recent progress in understanding morphogen signaling dynamics and patterning in early mammalian development by taking advantage of cutting-edge embryonic stem cell technology.


Asunto(s)
Tipificación del Cuerpo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Embrión no Mamífero/fisiología , Células Madre Embrionarias/fisiología , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Animales , Diferenciación Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Embrión no Mamífero/citología , Células Madre Embrionarias/citología
19.
Development ; 147(3)2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32014865

RESUMEN

The pluripotent epiblast gives rise to all tissues and organs in the adult body. Its differentiation starts at gastrulation, when the epiblast generates mesoderm and endoderm germ layers through epithelial-mesenchymal transition (EMT). Although gastrulation EMT coincides with loss of epiblast pluripotency, pluripotent cells in development and in vitro can adopt either mesenchymal or epithelial morphology. The relationship between epiblast cellular morphology and its pluripotency is not well understood. Here, using chicken epiblast and mammalian pluripotency stem cell (PSC) models, we show that PSCs undergo a mesenchymal-epithelial transition (MET) prior to EMT-associated pluripotency loss. Epiblast MET and its subsequent EMT are two distinct processes. The former, a partial MET, is associated with reversible initiation of pluripotency exit, whereas the latter, a full EMT, is associated with complete and irreversible pluripotency loss. We provide evidence that integrin-mediated cell-matrix interaction is a key player in pluripotency exit regulation. We propose that epiblast partial MET is an evolutionarily conserved process among all amniotic vertebrates and that epiblast pluripotency is restricted to an intermediate cellular state residing between the fully mesenchymal and fully epithelial states.


Asunto(s)
Endodermo/citología , Transición Epitelial-Mesenquimal/fisiología , Gastrulación/fisiología , Mesodermo/citología , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular , Línea Celular , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Humanos , Morfogénesis/genética
20.
PLoS Biol ; 17(10): e3000498, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31613879

RESUMEN

During gastrulation, the pluripotent epiblast self-organizes into the 3 germ layers-endoderm, mesoderm and ectoderm, which eventually form the entire embryo. Decades of research in the mouse embryo have revealed that a signaling cascade involving the Bone Morphogenic Protein (BMP), WNT, and NODAL pathways is necessary for gastrulation. In vivo, WNT and NODAL ligands are expressed near the site of gastrulation in the posterior of the embryo, and knockout of these ligands leads to a failure to gastrulate. These data have led to the prevailing view that a signaling gradient in WNT and NODAL underlies patterning during gastrulation; however, the activities of these pathways in space and time have never been directly observed. In this study, we quantify BMP, WNT, and NODAL signaling dynamics in an in vitro model of human gastrulation. Our data suggest that BMP signaling initiates waves of WNT and NODAL signaling activity that move toward the colony center at a constant rate. Using a simple mathematical model, we show that this wave-like behavior is inconsistent with a reaction-diffusion-based Turing system, indicating that there is no stable signaling gradient of WNT/NODAL. Instead, the final signaling state is homogeneous, and spatial differences arise only from boundary effects. We further show that the durations of WNT and NODAL signaling control mesoderm differentiation, while the duration of BMP signaling controls differentiation of CDX2-positive extra-embryonic cells. The identity of these extra-embryonic cells has been controversial, and we use RNA sequencing (RNA-seq) to obtain their transcriptomes and show that they closely resemble human trophoblast cells in vivo. The domain of BMP signaling is identical to the domain of differentiation of these trophoblast-like cells; however, neither WNT nor NODAL forms a spatial pattern that maps directly to the mesodermal region, suggesting that mesoderm differentiation is controlled dynamically by the combinatorial effect of multiple signals. We synthesize our data into a mathematical model that accurately recapitulates signaling dynamics and predicts cell fate patterning upon chemical and physical perturbations. Taken together, our study shows that the dynamics of signaling events in the BMP, WNT, and NODAL cascade in the absence of a stable signaling gradient control fate patterning of human gastruloids.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Gastrulación/genética , Mesodermo/metabolismo , Proteína Nodal/genética , Transducción de Señal , Proteínas Wnt/genética , Benzotiazoles/farmacología , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Gástrula/citología , Gástrula/efectos de los fármacos , Gástrula/metabolismo , Gastrulación/efectos de los fármacos , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Modelos Biológicos , Modelos Estadísticos , Proteína Nodal/deficiencia , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA