43 research outputs found

    Adjustable viscoelasticity allows for efficient collective cell migration

    Get PDF
    Cell migration is essential for a wide range of biological processes such as embryo morphogenesis, wound healing, regeneration, and also in pathological conditions, such as cancer. In such contexts, cells are required to migrate as individual entities or as highly coordinated collectives, both of which requiring cells to respond to molecular and mechanical cues from their environment. However, whilst the function of chemical cues in cell migration is comparatively well understood, the role of tissue mechanics on cell migration is just starting to be studied. Recent studies suggest that the dynamic tuning of the viscoelasticity within a migratory cluster of cells, and the adequate elastic properties of its surrounding tissues, are essential to allow efficient collective cell migration in vivo. In this review we focus on the role of viscoelasticity in the control of collective cell migration in various cellular systems, mentioning briefly some aspects of single cell migration. We aim to provide details on how viscoelasticity of collectively migrating groups of cells and their surroundings is adjusted to ensure correct morphogenesis, wound healing, and metastasis. Finally, we attempt to show that environmental viscoelasticity triggers molecular changes within migrating clusters and that these new molecular setups modify clusters' viscoelasticity, ultimately allowing them to migrate across the challenging geometries of their microenvironment

    PDGF controls contact inhibition of locomotion by regulating N-cadherin during neural crest migration

    Get PDF
    A fundamental property of neural crest (NC) migration is contact inhibition of locomotion (CIL), a process by which cells change their direction of migration upon cell contact. CIL has been proven to be essential for NC migration in amphibians and zebrafish by controlling cell polarity in a cell contact-dependent manner. Cell contact during CIL requires the participation of the cell adhesion molecule N-cadherin, which starts to be expressed by NC cells as a consequence of the switch between E- and N-cadherins during epithelial-to-mesenchymal transition (EMT). However, the mechanism that controls the upregulation of N-cadherin remains unknown. Here, we show that platelet-derived growth factor receptor alpha (PDGFRα) and its ligand platelet-derived growth factor A (PDGF-A) are co-expressed in migrating cranial NC. Inhibition of PDGF-A/PDGFRα blocks NC migration by inhibiting N-cadherin and, consequently, impairing CIL. Moreover, we identify phosphatidylinositol-3-kinase (PI3K)/AKT as a downstream effector of the PDGFRα cellular response during CIL. Our results lead us to propose PDGF-A/ PDGFRα signalling as a tissue-autonomous regulator of CIL by controlling N-cadherin upregulation during EMT. Finally, we show that once NC cells have undergone EMT, the same PDGF-A/PDGFRα works as an NC chemoattractant, guiding their directional migration

    Delamination of neural crest cells requires transient and reversible Wnt inhibition mediated by DACT1/2

    Get PDF
    Delamination of neural crest (NC) cells is a bona fide physiological model of epithelial-to-mesenchymal transition (EMT), a process that is influenced by Wnt/β-catenin signalling. Using two in vivo models, we show that Wnt/β-catenin signalling is transiently inhibited at the time of NC delamination. In attempting to define the mechanism underlying this inhibition, we found that the scaffold proteins Dact1 and Dact2, which are expressed in pre-migratory NC cells, are required for NC delamination in Xenopus and chick embryos, whereas they do not affect the motile properties of migratory NC cells. Dact1/2 inhibit Wnt/β-catenin signalling upstream of the transcriptional activity of T cell factor (TCF), which is required for EMT to proceed. Dact1/2 regulate the subcellular distribution of β-catenin, preventing β-catenin from acting as a transcriptional co-activator to TCF, yet without affecting its stability. Together, these data identify a novel yet important regulatory element that inhibits β-catenin signalling, which then affects NC delamination

    Author Correction: Gap junction protein Connexin-43 is a direct transcriptional regulator of N-cadherin in vivo

    Get PDF
    Correction to: Nature Communications (2018); https://doi.org/10.1038/s41467-018-06368-x, published online 21 September 2018. The original version of this Article contained an error in the spelling of the author Alexandra Schambony, which was incorrectly given as Alexandra Schambon. This has now been corrected in both the PDF and HTML versions of the Article

    Gap junction protein Connexin-43 is a direct transcriptional regulator of N-cadherin in vivo

    Get PDF
    Connexins are the primary components of gap junctions, providing direct links between cells under many physiological processes. Here, we demonstrate that in addition to this canonical role, Connexins act as transcriptional regulators. We show that Connexin 43 (Cx43) controls neural crest cell migration in vivo by directly regulating N-cadherin transcription. This activity requires interaction between Cx43 carboxy tail and the basic transcription factor-3, which drives the translocation of Cx43 tail to the nucleus. Once in the nucleus they form a complex with PolII which directly binds to the N-cadherin promoter. We found that this mechanism is conserved between amphibian and mammalian cells. Given the strong evolutionary conservation of connexins across vertebrates, this may reflect a common mechanism of gene regulation by a protein whose function was previously ascribed only to gap junctional communication

    SPIN90 associates with mDia1 and the Arp2/3 complex to regulate cortical actin organization

    Get PDF
    Cell shape is controlled by the submembranous cortex, an actomyosin network mainly generated by two actin nucleators: the Arp2/3 complex and the formin mDia1. Changes in relative nucleator activity may alter cortical organization, mechanics and cell shape. Here we investigate how nucleation-promoting factors mediate interactions between nucleators. In vitro, the nucleation-promoting factor SPIN90 promotes formation of unbranched filaments by Arp2/3, a process thought to provide the initial filament for generation of dendritic networks. Paradoxically, in cells, SPIN90 appears to favour a formin-dominated cortex. Our in vitro experiments reveal that this feature stems mainly from two mechanisms: efficient recruitment of mDia1 to SPIN90-Arp2/3 nucleated filaments and formation of a ternary SPIN90-Arp2/3-mDia1 complex that greatly enhances filament nucleation. Both mechanisms yield rapidly elongating filaments with mDia1 at their barbed ends and SPIN90-Arp2/3 at their pointed ends. Thus, in networks, SPIN90 lowers branching densities and increases the proportion of long filaments elongated by mDia1

    Tissue stiffening coordinates morphogenesis by triggering collective cell migration in vivo.

    Get PDF
    Collective cell migration is essential for morphogenesis, tissue remodelling and cancer invasion. In vivo, groups of cells move in an orchestrated way through tissues. This movement involves mechanical as well as molecular interactions between cells and their environment. While the role of molecular signals in collective cell migration is comparatively well understood, how tissue mechanics influence collective cell migration in vivo remains unknown. Here we investigated the importance of mechanical cues in the collective migration of the Xenopus laevis neural crest cells, an embryonic cell population whose migratory behaviour has been likened to cancer invasion. We found that, during morphogenesis, the head mesoderm underlying the cephalic neural crest stiffens. This stiffening initiates an epithelial-to-mesenchymal transition in neural crest cells and triggers their collective migration. To detect changes in their mechanical environment, neural crest cells use mechanosensation mediated by the integrin-vinculin-talin complex. By performing mechanical and molecular manipulations, we show that mesoderm stiffening is necessary and sufficient to trigger neural crest migration. Finally, we demonstrate that convergent extension of the mesoderm, which starts during gastrulation, leads to increased mesoderm stiffness by increasing the cell density underneath the neural crest. These results show that convergent extension of the mesoderm has a role as a mechanical coordinator of morphogenesis, and reveal a link between two apparently unconnected processes-gastrulation and neural crest migration-via changes in tissue mechanics. Overall, we demonstrate that changes in substrate stiffness can trigger collective cell migration by promoting epithelial-to-mesenchymal transition in vivo. More broadly, our results raise the idea that tissue mechanics combines with molecular effectors to coordinate morphogenesis

    In Vivo and In Vitro Quantitative Analysis of Neural Crest Cell Migration

    No full text
    The neural crest is an embryonic cell population induced at the border of the neural plate from where it delaminates and migrates long distances across the embryo. Due to its extraordinary migratory capabilities, the neural crest has become a powerful system to study cellular and molecular aspects of collective and single cell migration both in vivo and in vitro. Here we provide detailed protocols used to perform quantitative analysis of molecular and cellular aspects of Xenopus laevis neural crest cell migration, both in vivo and in vitro
    corecore