Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Cell Biol ; 26(4): 530-541, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499770

ABSTRACT

Embryonic induction is a key mechanism in development that corresponds to an interaction between a signalling and a responding tissue, causing a change in the direction of differentiation by the responding tissue. Considerable progress has been achieved in identifying inductive signals, yet how tissues control their responsiveness to these signals, known as competence, remains poorly understood. While the role of molecular signals in competence has been studied, how tissue mechanics influence competence remains unexplored. Here we investigate the role of hydrostatic pressure in controlling competence in neural crest cells, an embryonic cell population. We show that neural crest competence decreases concomitantly with an increase in the hydrostatic pressure of the blastocoel, an embryonic cavity in contact with the prospective neural crest. By manipulating hydrostatic pressure in vivo, we show that this increase leads to the inhibition of Yap signalling and impairs Wnt activation in the responding tissue, which would be required for neural crest induction. We further show that hydrostatic pressure controls neural crest induction in amphibian and mouse embryos and in human cells, suggesting a conserved mechanism across vertebrates. Our work sets out how tissue mechanics can interplay with signalling pathways to regulate embryonic competence.


Subject(s)
Embryonic Induction , Neural Crest , Animals , Humans , Mice , Hydrostatic Pressure , Neural Crest/metabolism , Prospective Studies , Wnt Proteins/metabolism
2.
Semin Cell Dev Biol ; 141: 63-73, 2023 05 30.
Article in English | MEDLINE | ID: mdl-35450765

ABSTRACT

Over the past two decades, molecular cell biology has graduated from a mostly analytic science to one with substantial synthetic capability. This success is built on a deep understanding of the structure and function of biomolecules and molecular mechanisms. For synthetic biology to achieve similar success at the scale of tissues and organs, an equally deep understanding of the principles of development is required. Here, we review some of the central concepts and recent progress in tissue patterning, morphogenesis and collective cell migration and discuss their value for synthetic developmental biology, emphasizing in particular the power of (guided) self-organization and the role of theoretical advances in making developmental insights applicable in synthesis.


Subject(s)
Developmental Biology , Synthetic Biology , Morphogenesis , Cell Movement
3.
Nat Commun ; 13(1): 4481, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35918320

ABSTRACT

Two-pore channels are endo-lysosomal cation channels with malleable selectivity filters that drive endocytic ion flux and membrane traffic. Here we show that TPC2 can differentially regulate its cation permeability when co-activated by its endogenous ligands, NAADP and PI(3,5)P2. Whereas NAADP rendered the channel Ca2+-permeable and PI(3,5)P2 rendered the channel Na+-selective, a combination of the two increased Ca2+ but not Na+ flux. Mechanistically, this was due to an increase in Ca2+ permeability independent of changes in ion selectivity. Functionally, we show that cell permeable NAADP and PI(3,5)P2 mimetics synergistically activate native TPC2 channels in live cells, globalizing cytosolic Ca2+ signals and regulating lysosomal pH and motility. Our data reveal that flux of different ions through the same pore can be independently controlled and identify TPC2 as a likely coincidence detector that optimizes lysosomal Ca2+ signaling.


Subject(s)
Calcium Channels , Calcium , Bias , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Cations/metabolism , Lysosomes/metabolism , NADP/metabolism
4.
EMBO J ; 40(18): e107245, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34396565

ABSTRACT

During embryonic development, signalling pathways orchestrate organogenesis by controlling tissue-specific gene expression programmes and differentiation. Although the molecular components of many common developmental signalling systems are known, our current understanding of how signalling inputs are translated into gene expression outputs in real-time is limited. Here we employ optogenetics to control the activation of Notch signalling during Drosophila embryogenesis with minute accuracy and follow target gene expression by quantitative live imaging. Light-induced nuclear translocation of the Notch Intracellular Domain (NICD) causes a rapid activation of target mRNA expression. However, target gene transcription gradually decays over time despite continuous photo-activation and nuclear NICD accumulation, indicating dynamic adaptation to the signalling input. Using mathematical modelling and molecular perturbations, we show that this adaptive transcriptional response fits to known motifs capable of generating near-perfect adaptation and can be best explained by state-dependent inactivation at the target cis-regulatory region. Taken together, our results reveal dynamic nuclear adaptation as a novel mechanism controlling Notch signalling output during tissue differentiation.


Subject(s)
Cell Nucleus/metabolism , Receptors, Notch/metabolism , Signal Transduction , Adaptation, Biological , Animals , Cell Nucleus/genetics , Drosophila/embryology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Organogenesis/genetics , Regulatory Sequences, Nucleic Acid
5.
Elife ; 92020 06 05.
Article in English | MEDLINE | ID: mdl-32501214

ABSTRACT

Quantitative microscopy is becoming increasingly crucial in efforts to disentangle the complexity of organogenesis, yet adoption of the potent new toolbox provided by modern data science has been slow, primarily because it is often not directly applicable to developmental imaging data. We tackle this issue with a newly developed algorithm that uses point cloud-based morphometry to unpack the rich information encoded in 3D image data into a straightforward numerical representation. This enabled us to employ data science tools, including machine learning, to analyze and integrate cell morphology, intracellular organization, gene expression and annotated contextual knowledge. We apply these techniques to construct and explore a quantitative atlas of cellular architecture for the zebrafish posterior lateral line primordium, an experimentally tractable model of complex self-organized organogenesis. In doing so, we are able to retrieve both previously established and novel biologically relevant patterns, demonstrating the potential of our data-driven approach.


Subject(s)
Cytological Techniques/methods , Imaging, Three-Dimensional/methods , Microscopy/methods , Organogenesis/physiology , Algorithms , Animals , Embryo, Nonmammalian/cytology , Zebrafish
6.
Curr Opin Cell Biol ; 66: 19-27, 2020 10.
Article in English | MEDLINE | ID: mdl-32408249

ABSTRACT

Morphogenesis of multicellular systems is governed by precise spatiotemporal regulation of biochemical reactions and mechanical forces which together with environmental conditions determine the development of complex organisms. Current efforts in the field aim at decoding the system-level principles underlying the regulation of developmental processes. Toward this goal, optogenetics, the science of regulation of protein function with light, is emerging as a powerful new tool to quantitatively perturb protein function in vivo with unprecedented precision in space and time. In this review, we provide an overview of how optogenetics is helping to address system-level questions of multicellular morphogenesis and discuss future directions.


Subject(s)
Morphogenesis , Optogenetics , Animals , Embryonic Development , Humans , Models, Biological
7.
Dev Cell ; 52(4): 492-508.e10, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32059773

ABSTRACT

How tissues migrate robustly through changing guidance landscapes is poorly understood. Here, quantitative imaging is combined with inducible perturbation experiments to investigate the mechanisms that ensure robust tissue migration in vivo. We show that tissues exposed to acute "chemokine floods" halt transiently before they perfectly adapt, i.e., return to the baseline migration behavior in the continued presence of elevated chemokine levels. A chemokine-triggered phosphorylation of the atypical chemokine receptor Cxcr7b reroutes it from constitutive ubiquitination-regulated degradation to plasma membrane recycling, thus coupling scavenging capacity to extracellular chemokine levels. Finally, tissues expressing phosphorylation-deficient Cxcr7b migrate normally in the presence of physiological chemokine levels but show delayed recovery when challenged with elevated chemokine concentrations. This work establishes that adaptation to chemokine fluctuations can be "outsourced" from canonical GPCR signaling to an autonomously acting scavenger receptor that both senses and dynamically buffers chemokine levels to increase the robustness of tissue migration.


Subject(s)
Cell Movement , Chemokines/metabolism , Embryo, Nonmammalian/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Communication , Chemokines/genetics , Embryo, Nonmammalian/cytology , Phosphorylation , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
8.
Development ; 146(20)2019 10 22.
Article in English | MEDLINE | ID: mdl-31641044

ABSTRACT

The development of multicellular organisms is controlled by highly dynamic molecular and cellular processes organized in spatially restricted patterns. Recent advances in optogenetics are allowing protein function to be controlled with the precision of a pulse of laser light in vivo, providing a powerful new tool to perturb developmental processes at a wide range of spatiotemporal scales. In this Primer, we describe the most commonly used optogenetic tools, their application in developmental biology and in the nascent field of synthetic morphogenesis.


Subject(s)
Developmental Biology/methods , Optogenetics/methods , Animals , Embryonic Development/physiology , Humans , Morphogenesis/physiology , Signal Transduction/physiology
9.
Dev Cell ; 49(1): 77-88.e7, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30880002

ABSTRACT

Phagocytic immune cells such as microglia can engulf and process pathogens and dying cells with high efficiency while still maintaining their dynamic behavior and morphology. Effective intracellular processing of ingested cells is likely to be crucial for microglial function, but the underlying cellular mechanisms are poorly understood. Using both living fish embryos and mammalian macrophages, we show that processing depends on the shrinkage and packaging of phagosomes into a unique cellular compartment, the gastrosome, with distinct molecular and ultra-structural characteristics. Loss of the transporter Slc37a2 blocks phagosomal shrinkage, resulting in the expansion of the gastrosome and the dramatic bloating of the cell. This, in turn, affects the ability of microglia to phagocytose and migrate toward brain injuries. Thus, this work identifies a conserved crucial step in the phagocytic pathway of immune cells and provides a potential entry point for manipulating their behavior in development and disease.


Subject(s)
Antiporters/genetics , Macrophages/metabolism , Membrane Transport Proteins/genetics , Microglia/metabolism , Phagosomes/ultrastructure , Animals , Apoptosis/genetics , Cell Compartmentation/genetics , HeLa Cells , Humans , Macrophages/ultrastructure , Mice , Microglia/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Phagocytes/ultrastructure , Phagocytosis/genetics , Phagosomes/genetics , RAW 264.7 Cells , Zebrafish/genetics , Zebrafish/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...