Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell Rep Methods ; 2(9): 100297, 2022 09 19.
Article in English | MEDLINE | ID: mdl-36160045

ABSTRACT

Organoids are carrying the promise of modeling complex disease phenotypes and serving as a powerful basis for unbiased drug screens, potentially offering a more efficient drug-discovery route. However, unsolved technical bottlenecks of reproducibility and scalability have prevented the use of current organoids for high-throughput screening. Here, we present a method that overcomes these limitations by using deep-learning-driven analysis for phenotypic drug screens based on highly standardized micropattern-based neural organoids. This allows us to distinguish between disease and wild-type phenotypes in complex tissues with extremely high accuracy as well as quantify two predictors of drug success: efficacy and adverse effects. We applied our approach to Huntington's disease (HD) and discovered that bromodomain inhibitors revert complex phenotypes induced by the HD mutation. This work demonstrates the power of combining machine learning with phenotypic drug screening and its successful application to reveal a potentially new druggable target for HD.


Subject(s)
Deep Learning , Huntington Disease , Humans , Huntington Disease/drug therapy , High-Throughput Screening Assays , Drug Evaluation, Preclinical , Reproducibility of Results , Organoids
2.
Elife ; 112022 04 22.
Article in English | MEDLINE | ID: mdl-35451959

ABSTRACT

The Hippo pathway, a highly conserved signaling cascade that functions as an integrator of molecular signals and biophysical states, ultimately impinges upon the transcription coactivator Yes-associated protein 1 (YAP). Hippo-YAP signaling has been shown to play key roles both at the early embryonic stages of implantation and gastrulation, and later during neurogenesis. To explore YAP's potential role in neurulation, we used self-organizing neuruloids grown from human embryonic stem cells on micropatterned substrates. We identified YAP activation as a key lineage determinant, first between neuronal ectoderm and nonneuronal ectoderm, and later between epidermis and neural crest, indicating that YAP activity can enhance the effect of BMP4 stimulation and therefore affect ectodermal specification at this developmental stage. Because aberrant Hippo-YAP signaling has been implicated in the pathology of Huntington's Disease (HD), we used isogenic mutant neuruloids to explore the relationship between signaling and the disease. We found that HD neuruloids demonstrate ectopic activation of gene targets of YAP and that pharmacological reduction of YAP's transcriptional activity can partially rescue the HD phenotype.


Subject(s)
Ectoderm , Huntington Disease , YAP-Signaling Proteins , Cell Cycle Proteins/metabolism , Ectoderm/metabolism , Humans , Neurogenesis , Neurulation , Signal Transduction/genetics , YAP-Signaling Proteins/genetics
3.
Nat Commun ; 12(1): 6768, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799555

ABSTRACT

Organizing centers secrete morphogens that specify the emergence of germ layers and the establishment of the body's axes during embryogenesis. While traditional experimental embryology tools have been instrumental in dissecting the molecular aspects of organizers in model systems, they are impractical in human in-vitro model systems to dissect the relationships between signaling and fate along embryonic coordinates. To systematically study human embryonic organizer centers, we devised a collection of optogenetic ePiggyBac vectors to express a photoactivatable Cre-loxP recombinase, that allows the systematic induction of organizer structures by shining blue-light on human embryonic stem cells (hESCs). We used a light stimulus to geometrically confine SHH expression in neuralizing hESCs. This led to the self-organization of mediolateral neural patterns. scRNA-seq analysis established that these structures represent the dorsal-ventral forebrain, at the end of the first month of development. Here, we show that morphogen light-stimulation is a scalable tool that induces self-organizing centers.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Human Embryonic Stem Cells/physiology , Prosencephalon/embryology , Cell Lineage/physiology , Embryology/methods , Gene Expression Regulation, Developmental/radiation effects , Genetic Vectors/genetics , Humans , Integrases/genetics , Light , Optogenetics/methods , Prosencephalon/metabolism , RNA-Seq , Signal Transduction/physiology , Signal Transduction/radiation effects , Single-Cell Analysis
4.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34608934

ABSTRACT

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of the CAG repeats in the huntingtin gene (HTT). Although HD has been shown to have a developmental component, how early during human embryogenesis the HTT-CAG expansion can cause embryonic defects remains unknown. Here, we demonstrate a specific and highly reproducible CAG length-dependent phenotypic signature in a synthetic model for human gastrulation derived from human embryonic stem cells (hESCs). Specifically, we observed a reduction in the extension of the ectodermal compartment that is associated with enhanced activin signaling. Surprisingly, rather than a cell-autonomous effect, tracking the dynamics of TGFß signaling demonstrated that HTT-CAG expansion perturbs the spatial restriction of activin response. This is due to defects in the apicobasal polarization in the context of the polarized epithelium of the 2D gastruloid, leading to ectopic subcellular localization of TGFß receptors. This work refines the earliest developmental window for the prodromal phase of HD to the first 2 weeks of human development, as modeled by our 2D gastruloids.


Subject(s)
Cell Lineage , Cell Polarity , Germ Layers/metabolism , Human Embryonic Stem Cells/metabolism , Huntingtin Protein/metabolism , Activins/metabolism , Animals , Cell Line , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Germ Layers/cytology , Germ Layers/embryology , Human Embryonic Stem Cells/cytology , Humans , Huntingtin Protein/genetics , Mice , Signal Transduction , Transforming Growth Factor beta/metabolism , Trinucleotide Repeat Expansion
5.
Dev Cell ; 56(13): 1930-1944.e5, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34051144

ABSTRACT

Using self-organizing human models of gastrulation, we previously showed that (1) BMP4 initiates the cascade of events leading to gastrulation, (2) BMP4 signal reception is restricted to the basolateral domain, and (3) in a human-specific manner, BMP4 directly induces the expression of NOGGIN. Here, we report the surprising discovery that in human epiblasts, NOGGIN and BMP4 were secreted into opposite extracellular spaces. Interestingly, apically presented NOGGIN could inhibit basally delivered BMP4. Apically imposed microfluidic flow demonstrated that NOGGIN traveled in the apical extracellular space. Our co-localization analysis detailed the endocytotic route that trafficked NOGGIN from the apical space to the basolateral intercellular space where BMP4 receptors were located. This apical-basal transcytosis was indispensable for NOGGIN inhibition. Taken together, the segregation of activator/inhibitor into distinct extracellular spaces challenges classical views of morphogen movement. We propose that the transport of morphogen inhibitors regulates the spatial availability of morphogens during embryogenesis.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Carrier Proteins/genetics , Cell Compartmentation/genetics , Extracellular Space/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Microfluidics , Morphogenesis/genetics , Signal Transduction/genetics , Transcytosis/genetics
6.
J Mol Med (Berl) ; 99(4): 569-579, 2021 04.
Article in English | MEDLINE | ID: mdl-33792755

ABSTRACT

In this article, we discuss the ethics of human embryoids, i.e., embryo-like structures made from pluripotent stem cells for modeling natural embryos. We argue that defining our social priorities is critical to design a consistent ethical guideline for research on those new entities. The absence of clear regulations on these emerging technologies stems from an unresolved debate surrounding natural human embryo research and one common opinion that one needs to solve the question of the moral status of the human embryo before regulating their surrogate. The recent NIH funding restrictions for research on human embryoids have made scientists even more unlikely to raise their voices. As a result, the scientific community has maintained a low profile while longing for a more favorable socio-political climate for their research. This article is a call for consistency among biomedical research on human materials, trying to position human embryoids within a spectrum of existing practice from stem cell research or IVF to research involving human subjects. We specifically note that the current practices in infertility clinics of freezing human embryos or disposing of them without any consideration for their potential benefits contradicts the assumption of special consideration for human material. Conversely, creating human embryoids for research purposes could ensure that no human material be used in vain, always serving humankind. We argue here that it is time to reconsider the full ban on embryo research (human embryos and embryoids) beyond the 14-day rule and that research on those entities should obey a sliding scale combining the completeness of the model (e.g., complete vs. partial) and the developmental stage: with more advanced completeness and developmental stage of the considered entity, being associated with more rigorous evaluation of societal benefits, statements of intention, and necessity of such research.


Subject(s)
Embryo Research/ethics , Embryoid Bodies , Stem Cell Research/ethics , Animals , Cryopreservation , Embryo Disposition/ethics , Embryo Disposition/legislation & jurisprudence , Embryo Research/legislation & jurisprudence , Embryo, Mammalian/cytology , Embryoid Bodies/cytology , Embryonic Development , Embryonic Stem Cells/cytology , Fertilization in Vitro/ethics , Fertilization in Vitro/legislation & jurisprudence , Gastrulation , Guidelines as Topic , Humans , Internationality , Mice , Morals , National Institutes of Health (U.S.) , Pluripotent Stem Cells/cytology , Stem Cell Research/legislation & jurisprudence , United States
7.
Nat Biotechnol ; 37(10): 1198-1208, 2019 10.
Article in English | MEDLINE | ID: mdl-31501559

ABSTRACT

Harnessing the potential of human embryonic stem cells to mimic normal and aberrant development with standardized models is a pressing challenge. Here we use micropattern technology to recapitulate early human neurulation in large numbers of nearly identical structures called neuruloids. Dual-SMAD inhibition followed by bone morphogenic protein 4 stimulation induced self-organization of neuruloids harboring neural progenitors, neural crest, sensory placode and epidermis. Single-cell transcriptomics unveiled the precise identities and timing of fate specification. Investigation of the molecular mechanism of neuruloid self-organization revealed a pulse of pSMAD1 at the edge that induced epidermis, whose juxtaposition to central neural fates specifies neural crest and placodes, modulated by fibroblast growth factor and Wnt. Neuruloids provide a unique opportunity to study the developmental aspects of human diseases. Using isogenic Huntington's disease human embryonic stem cells and deep neural network analysis, we show how specific phenotypic signatures arise in our model of early human development as a consequence of mutant huntingtin protein, outlining an approach for phenotypic drug screening.


Subject(s)
Ectoderm/physiology , Embryonic Stem Cells/physiology , Huntington Disease , Neurulation/physiology , Telencephalon/growth & development , Cell Culture Techniques , Cell Differentiation/physiology , Cell Line , Humans , Neurogenesis , Telencephalon/physiology
8.
Nat Cell Biol ; 21(7): 900-910, 2019 07.
Article in English | MEDLINE | ID: mdl-31263269

ABSTRACT

Breaking the anterior-posterior symmetry in mammals occurs at gastrulation. Much of the signalling network underlying this process has been elucidated in the mouse; however, there is no direct molecular evidence of events driving axis formation in humans. Here, we use human embryonic stem cells to generate an in vitro three-dimensional model of a human epiblast whose size, cell polarity and gene expression are similar to a day 10 human epiblast. A defined dose of BMP4 spontaneously breaks axial symmetry, and induces markers of the primitive streak and epithelial-to-mesenchymal transition. We show that WNT signalling and its inhibitor DKK1 play key roles in this process downstream of BMP4. Our work demonstrates that a model human epiblast can break axial symmetry despite the absence of asymmetry in the initial signal and of extra-embryonic tissues or maternal cues. Our three-dimensional model is an assay for the molecular events underlying human axial symmetry breaking.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Gene Expression Regulation, Developmental/physiology , Germ Layers/metabolism , Primitive Streak/metabolism , Tissue Culture Techniques , Cell Polarity/physiology , Epithelial-Mesenchymal Transition , Gastrulation/physiology , Humans , Primitive Streak/embryology , Signal Transduction/physiology
9.
Nat Cell Biol ; 21(6): 671-673, 2019 06.
Article in English | MEDLINE | ID: mdl-31160707
10.
Elife ; 72018 10 12.
Article in English | MEDLINE | ID: mdl-30311909

ABSTRACT

Self-organization of discrete fates in human gastruloids is mediated by a hierarchy of signaling pathways. How these pathways are integrated in time, and whether cells maintain a memory of their signaling history remains obscure. Here, we dissect the temporal integration of two key pathways, WNT and ACTIVIN, which along with BMP control gastrulation. CRISPR/Cas9-engineered live reporters of SMAD1, 2 and 4 demonstrate that in contrast to the stable signaling by SMAD1, signaling and transcriptional response by SMAD2 is transient, and while necessary for pluripotency, it is insufficient for differentiation. Pre-exposure to WNT, however, endows cells with the competence to respond to graded levels of ACTIVIN, which induces differentiation without changing SMAD2 dynamics. This cellular memory of WNT signaling is necessary for ACTIVIN morphogen activity. A re-evaluation of the evidence gathered over decades in model systems, re-enforces our conclusions and points to an evolutionarily conserved mechanism.


Subject(s)
Activins/metabolism , Gastrulation , Wnt Signaling Pathway , Animals , Base Sequence , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Endoderm/cytology , Genes, Reporter , Humans , Mesoderm/cytology , Mice , Nucleotide Motifs/genetics , Pluripotent Stem Cells/metabolism , Rats , Smad Proteins/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism
11.
Nat Mater ; 17(11): 1048, 2018 11.
Article in English | MEDLINE | ID: mdl-30232394

ABSTRACT

In the version of this Article originally published, Supplementary Videos 3-5 were incorrectly labelled; 3 should have been 5, 4 should have been 3 and 5 should have been 4. This has now been corrected.

12.
Nat Mater ; 17(8): 740-746, 2018 08.
Article in English | MEDLINE | ID: mdl-29967464

ABSTRACT

The diffusivity of macromolecules in the cytoplasm of eukaryotic cells varies over orders of magnitude and dictates the kinetics of cellular processes. However, a general description that associates the Brownian or anomalous nature of intracellular diffusion to the architectural and biochemical properties of the cytoplasm has not been achieved. Here we measure the mobility of individual fluorescent nanoparticles in living mammalian cells to obtain a comprehensive analysis of cytoplasmic diffusion. We identify a correlation between tracer size, its biochemical nature and its mobility. Inert particles with size equal or below 50 nm behave as Brownian particles diffusing in a medium of low viscosity with negligible effects of molecular crowding. Increasing the strength of non-specific interactions of the nanoparticles within the cytoplasm gradually reduces their mobility and leads to subdiffusive behaviour. These experimental observations and the transition from Brownian to subdiffusive motion can be captured in a minimal phenomenological model.


Subject(s)
Cytosol/metabolism , Nanoparticles/chemistry , Diffusion , HeLa Cells , Humans , Particle Size , Quantum Dots/chemistry , Quantum Dots/metabolism
13.
Nat Protoc ; 11(11): 2223-2232, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27735934

ABSTRACT

Fate allocation in the gastrulating embryo is spatially organized as cells differentiate into specialized cell types depending on their positions with respect to the body axes. There is a need for in vitro protocols that allow the study of spatial organization associated with this developmental transition. Although embryoid bodies and organoids can exhibit some spatial organization of differentiated cells, methods that generate embryoid bodies or organoids do not yield consistent and fully reproducible results. Here, we describe a micropatterning approach in which human embryonic stem cells are confined to disk-shaped, submillimeter colonies. After 42 h of BMP4 stimulation, cells form self-organized differentiation patterns in concentric radial domains, which express specific markers associated with the embryonic germ layers, reminiscent of gastrulating embryos. Our protocol takes 3 d; it uses commercial microfabricated slides (from CYTOO), human laminin-521 (LN-521) as extracellular matrix coating, and either conditioned or chemically defined medium (mTeSR). Differentiation patterns within individual colonies can be determined by immunofluorescence and analyzed with cellular resolution. Both the size of the micropattern and the type of medium affect the patterning outcome. The protocol is appropriate for personnel with basic stem cell culture training. This protocol describes a robust platform for quantitative analysis of the mechanisms associated with pattern formation at the onset of gastrulation.


Subject(s)
Human Embryonic Stem Cells/cytology , Microtechnology/methods , Cell Differentiation , Cell Line , Gastrulation , Humans
14.
Dev Cell ; 39(3): 302-315, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27746044

ABSTRACT

The earliest aspects of human embryogenesis remain mysterious. To model patterning events in the human embryo, we used colonies of human embryonic stem cells (hESCs) grown on micropatterned substrate and differentiated with BMP4. These gastruloids recapitulate the embryonic arrangement of the mammalian germ layers and provide an assay to assess the structural and signaling mechanisms patterning the human gastrula. Structurally, high-density hESCs localize their receptors to transforming growth factor ß at their lateral side in the center of the colony while maintaining apical localization of receptors at the edge. This relocalization insulates cells at the center from apically applied ligands while maintaining response to basally presented ones. In addition, BMP4 directly induces the expression of its own inhibitor, NOGGIN, generating a reaction-diffusion mechanism that underlies patterning. We develop a quantitative model that integrates edge sensing and inhibitors to predict human fate positioning in gastruloids and, potentially, the human embryo.


Subject(s)
Gastrula/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Animals , Body Patterning/drug effects , Bone Morphogenetic Protein 4/pharmacology , Carrier Proteins/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Polarity/drug effects , Cell Proliferation/drug effects , Colony-Forming Units Assay , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Feedback, Physiological/drug effects , Human Embryonic Stem Cells/drug effects , Humans , Ligands , Mice , Models, Biological , Phosphorylation/drug effects , Receptors, Cell Surface/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad1 Protein/metabolism , Time Factors , Transforming Growth Factor beta/metabolism
15.
Curr Top Dev Biol ; 116: 99-113, 2016.
Article in English | MEDLINE | ID: mdl-26970615

ABSTRACT

The developing embryo is a remarkable example of self-organization, where functional units are created in a complex spatiotemporal choreography. Recently, human embryonic stem cells (ESCs) have been used to recapitulate in vitro the self-organization programs that are executed in the embryo in vivo. This represents an unique opportunity to address self-organization in humans that is otherwise not addressable with current technologies. In this chapter, we review the recent literature on self-organization of human ESCs, with a particular focus on two examples: formation of embryonic germ layers and neural rosettes. Intriguingly, both activation and elimination of TGFß signaling can initiate self-organization, albeit with different molecular underpinnings. We discuss the mechanisms underlying the formation of these structures in vitro and explore future challenges in the field.


Subject(s)
Embryonic Stem Cells/cytology , Bone Morphogenetic Protein 4/metabolism , Embryonic Stem Cells/physiology , Humans , Transforming Growth Factor beta/metabolism
16.
Biophys J ; 109(9): 1785-97, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26536256

ABSTRACT

Recently developed optogenetic methods promise to revolutionize cell biology by allowing signaling perturbations to be controlled in space and time with light. However, a quantitative analysis of the relationship between a custom-defined illumination pattern and the resulting signaling perturbation is lacking. Here, we characterize the biophysical processes governing the localized recruitment of the Cryptochrome CRY2 to its membrane-anchored CIBN partner. We develop a quantitative framework and present simple procedures that enable predictive manipulation of protein distributions on the plasma membrane with a spatial resolution of 5 µm. We show that protein gradients of desired levels can be established in a few tens of seconds and then steadily maintained. These protein gradients can be entirely relocalized in a few minutes. We apply our approach to the control of the Cdc42 Rho GTPase activity. By inducing strong localized signaling perturbation, we are able to monitor the initiation of cell polarity and migration with a remarkable reproducibility despite cell-to-cell variability.


Subject(s)
Cell Membrane/metabolism , Cryptochromes/metabolism , Optogenetics/methods , cdc42 GTP-Binding Protein/metabolism , Animals , Diffusion , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Light , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , NIH 3T3 Cells , cdc42 GTP-Binding Protein/genetics , Red Fluorescent Protein
17.
Nano Lett ; 15(5): 3487-94, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25895433

ABSTRACT

Tools for controlling the spatial organization of proteins are a major prerequisite for deciphering mechanisms governing the dynamic architecture of living cells. Here, we have developed a generic approach for inducing and maintaining protein gradients inside living cells by means of biofunctionalized magnetic nanoparticles (MNPs). For this purpose, we tailored the size and surface properties of MNPs in order to ensure unhindered mobility in the cytosol. These MNPs with a core diameter below 50 nm could be rapidly relocalized in living cells by exploiting biased diffusion at weak magnetic forces in the femto-Newton range. In combination with MNP surface functionalization for specific in situ capturing of target proteins as well as efficient delivery into the cytosplasm, we here present a comprehensive technology for controlling intracellular protein gradients with a temporal resolution of a few tens of seconds.


Subject(s)
Cytosol/drug effects , Magnetite Nanoparticles/chemistry , Proteins/chemistry , Cell Line , Cytosol/chemistry , Humans , Magnetite Nanoparticles/administration & dosage , Particle Size , Proteins/administration & dosage , Surface Properties
18.
Nat Commun ; 5: 5093, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25298155

ABSTRACT

Magneto-fluorescent particles have been recognized as an emerging class of materials that exhibit great potential in advanced applications. However, synthesizing such magneto-fluorescent nanomaterials that simultaneously exhibit uniform and tunable sizes, high magnetic content loading, maximized fluorophore coverage at the surface and a versatile surface functionality has proven challenging. Here we report a simple approach for co-assembling magnetic nanoparticles with fluorescent quantum dots to form colloidal magneto-fluorescent supernanoparticles. Importantly, these supernanoparticles exhibit a superstructure consisting of a close-packed magnetic nanoparticle 'core', which is fully surrounded by a 'shell' of fluorescent quantum dots. A thin layer of silica coating provides high colloidal stability and biocompatibility, and a versatile surface functionality. We demonstrate that after surface pegylation, these silica-coated magneto-fluorescent supernanoparticles can be magnetically manipulated inside living cells while being optically tracked. Moreover, our silica-coated magneto-fluorescent supernanoparticles can also serve as an in vivo multi-photon and magnetic resonance dual-modal imaging probe.


Subject(s)
Fluorescent Dyes/chemistry , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Quantum Dots/chemistry , Fluorescence , Silicon Dioxide/chemistry
19.
J Cell Biol ; 206(6): 707-17, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25202028

ABSTRACT

Oriented cell divisions are necessary for the development of epithelial structures. Mitotic spindle orientation requires the precise localization of force generators at the cell cortex via the evolutionarily conserved LGN complex. However, polarity cues acting upstream of this complex in vivo in the vertebrate epithelia remain unknown. In this paper, we show that Dlg1 is localized at the basolateral cell cortex during mitosis and is necessary for planar spindle orientation in the chick neuroepithelium. Live imaging revealed that Dlg1 is required for directed spindle movements during metaphase. Mechanistically, we show that direct interaction between Dlg1 and LGN promotes cortical localization of the LGN complex. Furthermore, in human cells dividing on adhesive micropatterns, homogenously localized Dlg1 recruited LGN to the mitotic cortex and was also necessary for proper spindle orientation. We propose that Dlg1 acts primarily to recruit LGN to the cortex and that Dlg1 localization may additionally provide instructive cues for spindle orientation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Polarity/genetics , Epithelium/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Spindle Apparatus/genetics , Animals , Cell Line, Tumor , Chick Embryo , Discs Large Homolog 1 Protein , HeLa Cells , Humans , Mitosis , RNA Interference , RNA, Small Interfering
20.
Nat Methods ; 11(8): 847-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973948

ABSTRACT

Embryos allocate cells to the three germ layers in a spatially ordered sequence. Human embryonic stem cells (hESCs) can generate the three germ layers in culture; however, differentiation is typically heterogeneous and spatially disordered. We show that geometric confinement is sufficient to trigger self-organized patterning in hESCs. In response to BMP4, colonies reproducibly differentiated to an outer trophectoderm-like ring, an inner ectodermal circle and a ring of mesendoderm expressing primitive-streak markers in between. Fates were defined relative to the boundary with a fixed length scale: small colonies corresponded to the outer layers of larger ones. Inhibitory signals limited the range of BMP4 signaling to the colony edge and induced a gradient of Activin-Nodal signaling that patterned mesendodermal fates. These results demonstrate that the intrinsic tendency of stem cells to make patterns can be harnessed by controlling colony geometries and provide a quantitative assay for studying paracrine signaling in early development.


Subject(s)
Body Patterning , Embryo, Mammalian , Embryonic Stem Cells/cytology , Bone Morphogenetic Proteins/antagonists & inhibitors , Cell Differentiation , Gastrulation , Humans , Transforming Growth Factor beta/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...