Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Cell ; 187(11): 2838-2854.e17, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38744282

ABSTRACT

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.


Subject(s)
Blastomeres , Cell Lineage , Embryo, Mammalian , Female , Humans , Blastomeres/cytology , Blastomeres/metabolism , Cell Division , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development , Germ Layers/cytology , Germ Layers/metabolism , Male , Animals , Mice
2.
EMBO J ; 41(24): e111132, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36345783

ABSTRACT

The cerebral cortex contains billions of neurons, and their disorganization or misspecification leads to neurodevelopmental disorders. Understanding how the plethora of projection neuron subtypes are generated by cortical neural stem cells (NSCs) is a major challenge. Here, we focused on elucidating the transcriptional landscape of murine embryonic NSCs, basal progenitors (BPs), and newborn neurons (NBNs) throughout cortical development. We uncover dynamic shifts in transcriptional space over time and heterogeneity within each progenitor population. We identified signature hallmarks of NSC, BP, and NBN clusters and predict active transcriptional nodes and networks that contribute to neural fate specification. We find that the expression of receptors, ligands, and downstream pathway components is highly dynamic over time and throughout the lineage implying differential responsiveness to signals. Thus, we provide an expansive compendium of gene expression during cortical development that will be an invaluable resource for studying neural developmental processes and neurodevelopmental disorders.


Subject(s)
Neural Stem Cells , Neurons , Animals , Mice , Cell Differentiation , Cell Lineage/genetics , Cerebral Cortex , Embryonic Stem Cells , Neurogenesis/genetics , Neurons/metabolism
3.
Development ; 150(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-37249125

ABSTRACT

Tissue patterning during embryonic development is remarkably precise. Here, we numerically determine the impact of the cell diameter, gradient length and the morphogen source on the variability of morphogen gradients. We show that the positional error increases with the gradient length relative to the size of the morphogen source, and with the square root of the cell diameter and the readout position. We provide theoretical explanations for these relationships, and show that they enable high patterning precision over developmental time for readouts that scale with expanding tissue domains, as observed in the Drosophila wing disc. Our analysis suggests that epithelial tissues generally achieve higher patterning precision with small cross-sectional cell areas. An extensive survey of measured apical cell areas shows that they are indeed small in developing tissues that are patterned by morphogen gradients. Enhanced precision may thus have led to the emergence of pseudostratification in epithelia, a phenomenon for which the evolutionary benefit had so far remained elusive.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Cross-Sectional Studies , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryonic Development , Cell Size , Body Patterning/genetics , Gene Expression Regulation, Developmental , Models, Biological , Drosophila melanogaster/genetics
4.
Development ; 149(21)2022 11 01.
Article in English | MEDLINE | ID: mdl-36205075

ABSTRACT

Kidneys develop via iterative branching of the ureteric epithelial tree and subsequent nephrogenesis at the branch points. Nephrons form in the cap mesenchyme as the metanephric mesenchyme (MM) condenses around the epithelial ureteric buds (UBs). Previous work has demonstrated that FGF8 is important for the survival of nephron progenitor cells (NPCs), and early deletion of Fgf8 leads to the cessation of nephron formation, which results in post-natal lethality. We now reveal a previously unreported function of FGF8. By combining transgenic mouse models, quantitative imaging assays and data-driven computational modelling, we show that FGF8 has a strong chemokinetic effect and that this chemokinetic effect is important for the condensation of NPCs to the UB. The computational model shows that the motility must be lower close to the UB to achieve NPC attachment. We conclude that the FGF8 signalling pathway is crucial for the coordination of NPC condensation at the UB. Chemokinetic effects have also been described for other FGFs and may be generally important for the formation of mesenchymal condensates.


Subject(s)
Kidney , Nephrons , Mice , Animals , Nephrons/metabolism , Kidney/metabolism , Organogenesis , Fibroblast Growth Factors/metabolism , Stem Cells/metabolism , Mice, Transgenic , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism
5.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35037942

ABSTRACT

Generating comprehensive image maps, while preserving spatial three-dimensional (3D) context, is essential in order to locate and assess quantitatively specific cellular features and cell-cell interactions during organ development. Despite recent advances in 3D imaging approaches, our current knowledge of the spatial organization of distinct cell types in the embryonic pancreatic tissue is still largely based on two-dimensional histological sections. Here, we present a light-sheet fluorescence microscopy approach to image the pancreas in three dimensions and map tissue interactions at key time points in the mouse embryo. We demonstrate the utility of the approach by providing volumetric data, 3D distribution of three main cellular components (epithelial, mesenchymal and endothelial cells) within the developing pancreas, and quantification of their relative cellular abundance within the tissue. Interestingly, our 3D images show that endocrine cells are constantly and increasingly in contact with endothelial cells forming small vessels, whereas the interactions with mesenchymal cells decrease over time. These findings suggest distinct cell-cell interaction requirements for early endocrine cell specification and late differentiation. Lastly, we combine our image data in an open-source online repository (referred to as the Pancreas Embryonic Cell Atlas).


Subject(s)
Imaging, Three-Dimensional/methods , Pancreas/anatomy & histology , Animals , Embryo, Mammalian/anatomy & histology , Embryonic Development , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelium/anatomy & histology , Homeobox Protein Nkx-2.5/deficiency , Homeobox Protein Nkx-2.5/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence
6.
Proc Natl Acad Sci U S A ; 119(20): e2117075119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35561223

ABSTRACT

Neurulation is the process in early vertebrate embryonic development during which the neural plate folds to form the neural tube. Spinal neural tube folding in the posterior neuropore changes over time, first showing a median hinge point, then both the median hinge point and dorsolateral hinge points, followed by dorsolateral hinge points only. The biomechanical mechanism of hinge point formation in the mammalian neural tube is poorly understood. Here we employ a mechanical finite element model to study neural tube formation. The computational model mimics the mammalian neural tube using microscopy data from mouse and human embryos. While intrinsic curvature at the neural plate midline has been hypothesized to drive neural tube folding, intrinsic curvature was not sufficient for tube closure in our simulations. We achieved neural tube closure with an alternative model combining mesoderm expansion, nonneural ectoderm expansion, and neural plate adhesion to the notochord. Dorsolateral hinge points emerged in simulations with low mesoderm expansion and zippering. We propose that zippering provides the biomechanical force for dorsolateral hinge point formation in settings where the neural plate lateral sides extend above the mesoderm. Together, these results provide a perspective on the biomechanical and molecular mechanism of mammalian spinal neurulation.


Subject(s)
Neural Tube , Neurulation , Animals , Ectoderm/embryology , Humans , Mice , Neural Plate/embryology , Neural Tube/embryology , Neurulation/physiology , Notochord/embryology
7.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33946098

ABSTRACT

During lung development, epithelial branches expand preferentially in a longitudinal direction. This bias in outgrowth has been linked to a bias in cell shape and in the cell division plane. How this bias arises is unknown. Here, we show that biased epithelial outgrowth occurs independent of the surrounding mesenchyme, of preferential turnover of the extracellular matrix at the bud tips and of FGF signalling. There is also no evidence for actin-rich filopodia at the bud tips. Rather, we find epithelial tubes to be collapsed during early lung and kidney development, and we observe fluid flow in the narrow tubes. By simulating the measured fluid flow inside segmented narrow epithelial tubes, we show that the shear stress levels on the apical surface are sufficient to explain the reported bias in cell shape and outgrowth. We use a cell-based vertex model to confirm that apical shear forces, unlike constricting forces, can give rise to both the observed bias in cell shapes and tube elongation. We conclude that shear stress may be a more general driver of biased tube elongation beyond its established role in angiogenesis. This article has an associated 'The people behind the papers' interview.


Subject(s)
Biomechanical Phenomena , Kidney/growth & development , Lung/growth & development , Organogenesis , Animals , Biophysics , Cell Shape , Epithelial Cells/cytology , Extracellular Matrix , Female , Male , Mesoderm/metabolism , Mice , Models, Biological , Morphogenesis , Pseudopodia
8.
Nat Methods ; 16(9): 932, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31406386

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Nat Methods ; 16(8): 743-749, 2019 08.
Article in English | MEDLINE | ID: mdl-31363225

ABSTRACT

Cellular behavior is controlled by the interplay of diverse biomolecules. Most experimental methods, however, can only monitor a single molecule class or reaction type at a time. We developed an in vitro nuclear magnetic resonance spectroscopy (NMR) approach, which permitted dynamic quantification of an entire 'heterotypic' network-simultaneously monitoring three distinct molecule classes (metabolites, proteins and RNA) and all elementary reaction types (bimolecular interactions, catalysis, unimolecular changes). Focusing on an eight-reaction co-transcriptional RNA folding network, in a single sample we recorded over 35 time points with over 170 observables each, and accurately determined five core reaction constants in multiplex. This reconstruction revealed unexpected cross-talk between the different reactions. We further observed dynamic phase-separation in a system of five distinct RNA-binding domains in the course of the RNA transcription reaction. Our Systems NMR approach provides a deeper understanding of biological network dynamics by combining the dynamic resolution of biochemical assays and the multiplexing ability of 'omics'.


Subject(s)
Gene Regulatory Networks , Metabolome , Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/analysis , RNA/analysis , HEK293 Cells , Humans , Nucleic Acid Conformation , Protein Conformation , Proteins/chemistry , RNA/chemistry , RNA Folding
10.
Development ; 144(19): 3465-3474, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28974640

ABSTRACT

During embryonic and adult neurogenesis, neural stem cells (NSCs) generate the correct number and types of neurons in a temporospatial fashion. Control of NSC activity and fate is crucial for brain formation and homeostasis. Neurogenesis in the embryonic and adult brain differ considerably, but Notch signaling and inhibitor of DNA-binding (ID) factors are pivotal in both. Notch and ID factors regulate NSC maintenance; however, it has been difficult to evaluate how these pathways potentially interact. Here, we combined mathematical modeling with analysis of single-cell transcriptomic data to elucidate unforeseen interactions between the Notch and ID factor pathways. During brain development, Notch signaling dominates and directly regulates Id4 expression, preventing other ID factors from inducing NSC quiescence. Conversely, during adult neurogenesis, Notch signaling and Id2/3 regulate neurogenesis in a complementary manner and ID factors can induce NSC maintenance and quiescence in the absence of Notch. Our analyses unveil key molecular interactions underlying NSC maintenance and mechanistic differences between embryonic and adult neurogenesis. Similar Notch and ID factor interactions may be crucial in other stem cell systems.


Subject(s)
Homeostasis , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Receptors, Notch/metabolism , Transcription Factor HES-1/metabolism , Animals , Cell Differentiation , Cell Proliferation , Computer Simulation , Embryo, Mammalian/metabolism , Feedback, Physiological , Gene Expression Regulation, Developmental , Humans , Mice , Models, Biological , Protein Binding , Signal Transduction
11.
Development ; 144(5): 837-843, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28246213

ABSTRACT

A fundamental question in developmental biology is how organ size is controlled. We have previously shown that the area growth rate in the Drosophila eye primordium declines inversely proportionally to the increase in its area. How the observed reduction in the growth rate is achieved is unknown. Here, we explore the dilution of the cytokine Unpaired (Upd) as a possible candidate mechanism. In the developing eye, upd expression is transient, ceasing at the time when the morphogenetic furrow first emerges. We confirm experimentally that the diffusion and stability of the JAK/STAT ligand Upd are sufficient to control eye disc growth via a dilution mechanism. We further show that sequestration of Upd by ectopic expression of an inactive form of the receptor Domeless (Dome) results in a substantially lower growth rate, but the area growth rate still declines inversely proportionally to the area increase. This growth rate-to-area relationship is no longer observed when Upd dilution is prevented by the continuous, ectopic expression of Upd. We conclude that a mechanism based on the dilution of the growth modulator Upd can explain how growth termination is controlled in the eye disc.


Subject(s)
Cytokines/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Eye/growth & development , Photoreceptor Cells, Invertebrate/physiology , Transcription Factors/metabolism , Animals , Computer Simulation , DNA-Binding Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Kinetics , Mutation , STAT Transcription Factors/metabolism , Signal Transduction
12.
Nature ; 511(7507): 46-51, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24990743

ABSTRACT

The large spectrum of limb morphologies reflects the wide evolutionary diversification of the basic pentadactyl pattern in tetrapods. In even-toed ungulates (artiodactyls, including cattle), limbs are adapted for running as a consequence of progressive reduction of their distal skeleton to symmetrical and elongated middle digits with hoofed phalanges. Here we analyse bovine embryos to establish that polarized gene expression is progressively lost during limb development in comparison to the mouse. Notably, the transcriptional upregulation of the Ptch1 gene, which encodes a Sonic hedgehog (SHH) receptor, is disrupted specifically in the bovine limb bud mesenchyme. This is due to evolutionary alteration of a Ptch1 cis-regulatory module, which no longer responds to graded SHH signalling during bovine handplate development. Our study provides a molecular explanation for the loss of digit asymmetry in bovine limb buds and suggests that modifications affecting the Ptch1 cis-regulatory landscape have contributed to evolutionary diversification of artiodactyl limbs.


Subject(s)
Biological Evolution , Extremities/anatomy & histology , Extremities/embryology , Hedgehog Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Body Patterning , Cattle , Female , Gene Expression Regulation, Developmental/genetics , Limb Buds/anatomy & histology , Limb Buds/embryology , Male , Mesoderm/metabolism , Mice , Mice, Transgenic , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Regulatory Sequences, Nucleic Acid/genetics
13.
Development ; 143(9): 1482-90, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26965369

ABSTRACT

The size and shape of organs is species specific, and even in species in which organ size is strongly influenced by environmental cues, such as nutrition or temperature, it follows defined rules. Therefore, mechanisms must exist to ensure a tight control of organ size within a given species, while being flexible enough to allow for the evolution of different organ sizes in different species. We combined computational modeling and quantitative measurements to analyze growth control in the Drosophila eye disc. We find that the area growth rate declines inversely proportional to the increasing total eye disc area. We identify two growth laws that are consistent with the growth data and that would explain the extraordinary robustness and evolutionary plasticity of the growth process and thus of the final adult eye size. These two growth laws correspond to very different control mechanisms and we discuss how each of these laws constrains the set of candidate biological mechanisms for growth control in the Drosophila eye disc.


Subject(s)
Drosophila melanogaster/embryology , Imaginal Discs/growth & development , Optic Disk/growth & development , Algorithms , Animals , Computer Simulation , Models, Biological , Organ Size/physiology
14.
Phys Biol ; 17(1): 016006, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31711053

ABSTRACT

During morphogenesis, epithelial tubes elongate. In the case of the mammalian lung, biased elongation has been linked to a bias in cell shape and cell division, but it has remained unclear whether a bias in cell shape along the axis of outgrowth is sufficient for biased outgrowth and how it arises. Here, we use our 2D cell-based tissue simulation software [Formula: see text] to investigate the conditions for biased epithelial outgrowth. We show that the observed bias in cell shape and cell division can result in the observed bias in outgrowth only in the case of strong cortical tension, and comparison to biological data suggests that the cortical tension in epithelia is likely sufficient. We explore mechanisms that may result in the observed bias in cell division and cell shapes. To this end, we test the possibility that the surrounding tissue or extracellular matrix acts as a mechanical constraint that biases growth in the longitudinal direction. While external compressive forces can result in the observed bias in outgrowth, we find that they do not result in the observed bias in cell shapes. We conclude that other mechanisms must exist that generate the bias in lung epithelial outgrowth.


Subject(s)
Epithelial Cells/cytology , Lung/physiology , Cell Division , Computer Simulation , Epithelium/growth & development , Software
15.
J Theor Biol ; 481: 233-248, 2019 11 21.
Article in English | MEDLINE | ID: mdl-30529487

ABSTRACT

Parameter estimation is a major challenge in computational modeling of biological processes. This is especially the case in image-based modeling where the inherently quantitative output of the model is measured against image data, which is typically noisy and non-quantitative. In addition, these models can have a high computational cost, limiting the number of feasible simulations, and therefore rendering most traditional parameter estimation methods unsuitable. In this paper, we present a pipeline that uses Gaussian process learning to estimate biological parameters from noisy, non-quantitative image data when the model has a high computational cost. This approach is first successfully tested on a parametric function with the goal of retrieving the original parameters. We then apply it to estimating parameters in a biological setting by fitting artificial in-situ hybridization (ISH) data of the developing murine limb bud. We expect that this method will be of use in a variety of modeling scenarios where quantitative data is missing and the use of standard parameter estimation approaches in biological modeling is prohibited by the computational cost of the model.


Subject(s)
Algorithms , Computer Simulation/economics , Embryo, Mammalian/embryology , Hindlimb/embryology , Image Processing, Computer-Assisted/economics , Models, Biological , Animals , In Situ Hybridization , Mice
16.
Brief Bioinform ; 17(4): 616-27, 2016 07.
Article in English | MEDLINE | ID: mdl-26510443

ABSTRACT

One of the major challenges in biology concerns the integration of data across length and time scales into a consistent framework: how do macroscopic properties and functionalities arise from the molecular regulatory networks-and how can they change as a result of mutations? Morphogenesis provides an excellent model system to study how simple molecular networks robustly control complex processes on the macroscopic scale despite molecular noise, and how important functional variants can emerge from small genetic changes. Recent advancements in three-dimensional imaging technologies, computer algorithms and computer power now allow us to develop and analyse increasingly realistic models of biological control. Here, we present our pipeline for image-based modelling that includes the segmentation of images, the determination of displacement fields and the solution of systems of partial differential equations on the growing, embryonic domains. The development of suitable mathematical models, the data-based inference of parameter sets and the evaluation of competing models are still challenging, and current approaches are discussed.


Subject(s)
Organogenesis , Computer Simulation , Models, Biological , Systems Biology
17.
Development ; 141(23): 4526-36, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25359721

ABSTRACT

Early branching events during lung development are stereotyped. Although key regulatory components have been defined, the branching mechanism remains elusive. We have now used a developmental series of 3D geometric datasets of mouse embryonic lungs as well as time-lapse movies of cultured lungs to obtain physiological geometries and displacement fields. We find that only a ligand-receptor-based Turing model in combination with a particular geometry effect that arises from the distinct expression domains of ligands and receptors successfully predicts the embryonic areas of outgrowth and supports robust branch outgrowth. The geometry effect alone does not support bifurcating outgrowth, while the Turing mechanism alone is not robust to noisy initial conditions. The negative feedback between the individual Turing modules formed by fibroblast growth factor 10 (FGF10) and sonic hedgehog (SHH) enlarges the parameter space for which the embryonic growth field is reproduced. We therefore propose that a signaling mechanism based on FGF10 and SHH directs outgrowth of the lung bud via a ligand-receptor-based Turing mechanism and a geometry effect.


Subject(s)
Fibroblast Growth Factor 10/metabolism , Hedgehog Proteins/metabolism , Lung/embryology , Models, Biological , Morphogenesis/physiology , Signal Transduction/physiology , Animals , Mice , Time-Lapse Imaging
18.
Bioinformatics ; 32(12): 1905-6, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26861819

ABSTRACT

UNLABELLED: MATLAB is popular in biological research for creating and simulating models that use ordinary differential equations (ODEs). However, sharing or using these models outside of MATLAB is often problematic. A community standard such as Systems Biology Markup Language (SBML) can serve as a neutral exchange format, but translating models from MATLAB to SBML can be challenging-especially for legacy models not written with translation in mind. We developed MOCCASIN (Model ODE Converter for Creating Automated SBML INteroperability) to help. MOCCASIN can convert ODE-based MATLAB models of biochemical reaction networks into the SBML format. AVAILABILITY AND IMPLEMENTATION: MOCCASIN is available under the terms of the LGPL 2.1 license (http://www.gnu.org/licenses/lgpl-2.1.html). Source code, binaries and test cases can be freely obtained from https://github.com/sbmlteam/moccasin CONTACT: : mhucka@caltech.edu SUPPLEMENTARY INFORMATION: More information is available at https://github.com/sbmlteam/moccasin.


Subject(s)
Computational Biology/methods , Computer Simulation , Software , Systems Biology , Models, Biological , Programming Languages
19.
PLoS Comput Biol ; 12(9): e1005052, 2016 09.
Article in English | MEDLINE | ID: mdl-27626238

ABSTRACT

Patterning and growth are linked during early development and have to be tightly controlled to result in a functional tissue or organ. During the development of the Drosophila eye, this linkage is particularly clear: the growth of the eye primordium mainly results from proliferating cells ahead of the morphogenetic furrow (MF), a moving signaling wave that sweeps across the tissue from the posterior to the anterior side, that induces proliferating cells anterior to it to differentiate and become cell cycle quiescent in its wake. Therefore, final eye disc size depends on the proliferation rate of undifferentiated cells and on the speed with which the MF sweeps across the eye disc. We developed a spatio-temporal model of the growing eye disc based on the regulatory interactions controlled by the signals Decapentaplegic (Dpp), Hedgehog (Hh) and the transcription factor Homothorax (Hth) and explored how the signaling patterns affect the movement of the MF and impact on eye disc growth. We used published and new quantitative data to parameterize the model. In particular, two crucial parameter values, the degradation rate of Hth and the diffusion coefficient of Hh, were measured. The model is able to reproduce the linear movement of the MF and the termination of growth of the primordium. We further show that the model can explain several mutant phenotypes, but fails to reproduce the previously observed scaling of the Dpp gradient in the anterior compartment.


Subject(s)
Drosophila/growth & development , Eye/growth & development , Morphogenesis/physiology , Signal Transduction/physiology , Animals , Cell Proliferation , Computational Biology , Computer Simulation , Drosophila Proteins/metabolism , Spatio-Temporal Analysis
20.
Development ; 140(20): 4119-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24086076

ABSTRACT

Morphogens are signaling factors that direct cell fate and tissue development at a distance from their source, and various modes of transport and interpretation have been suggested for morphogens. The recent EMBO Workshop on 'Morphogen gradients', which took place in Oxford, UK in June 2013, centered on the formation and interpretation of such morphogen gradients during development. This meeting allowed an exchange of views in light of recent results. Here, we provide a brief overview of the talks, organized in relation to several major themes of discussion at the meeting: (1) morphogen gradient formation; (2) morphogen gradient interpretation; (3) signaling networks and feedback in morphogenesis; (4) emergence of patterns; (5) scaling of patterns; (6) the control of growth; and (7) new techniques in the field.


Subject(s)
Morphogenesis , Animals , Body Patterning , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Drosophila Proteins/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Homeodomain Proteins/metabolism , Nodal Protein/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Transport , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL