34 research outputs found

    Establishment of cell-cell junctions depends on the oligomeric states of VE-cadherin.: Oligomerization of VE-cadherin at cell surface

    Get PDF
    International audienceSpecifically expressed at intercellular adherens junctions of endothelial cells, VE-cadherin is a receptor that exhibits particular self-association properties. Indeed, in vitro studies demonstrated that the extracellular part of VE-cadherin elaborates Ca(++)-dependent hexameric structures. We hypothesized that this assembly could be at the basis of a new cadherin-mediated cell-cell adhesion mechanism. To verify this assumption, we first demonstrated that VE-cadherin can elaborate hexamers at the cell surface of confluent endothelial cells. Second, mutations were introduced within the extracellular part of VE-cadherin to destabilize the hexamer. Following an in vitro screening, three mutants were selected, among which, one is able to elaborate only dimers. The selected mutations were expressed as C-terminal green fluorescent protein fusions in CHO cells. Despite their capacity to elaborate nascent cell-cell contacts, the mutants seem to be rapidly degraded and/or internalized. Altogether, our results suggest that the formation of VE-cadherin hexamers protects this receptor and might allow the elaboration of mature endothelial cell-cell junctions

    The Motor Protein Myosin-X Transports VE-Cadherin along Filopodia To Allow the Formation of Early Endothelial Cell-Cell Contacts: MYOSIN-X TRANSPORT OF VE-CADHERIN ALONG FILOPODIA

    Get PDF
    International audienceVascular endothelium (VE), the monolayer of endothelial cells that lines the vascular tree, undergoes damage at the basis of some vascular diseases. Its integrity is maintained by VE-cadherin, an adhesive receptor localized at cell-cell junctions. Here, we show that VE-cadherin is also located at the tip and along filopodia in sparse or subconfluent endothelial cells. We observed that VE-cadherin navigates along intrafilopodial actin filaments. We found that the actin motor protein myosin-X is colocalized and moves synchronously with filopodial VE-cadherin. Immunoprecipitation and pulldown assays confirmed that myosin-X is directly associated with the VE-cadherin complex. Furthermore, expression of a dominant-negative mutant of myosin-X revealed that myosin-X is required for VE-cadherin export to cell edges and filopodia. These features indicate that myosin-X establishes a link between the actin cytoskeleton and VE-cadherin, thereby allowing VEcadherin transportation along intrafilopodial actin cables. In conclusion, we propose that VE-cadherin trafficking along filopodia using myosin-X motor protein is a prerequisite for cell-cell junction formation. This mechanism may have functional consequences for endothelium repair in pathological settings

    Dynamique de la jonction adhérente (rôle d'EPLIN dans la stabilité des contacts intercellulaires de l'endothélium vasculaire)

    No full text
    L'endothelium vasculaire constitue la principale barrière entre le sang et les tissus régulant le passage de macromolécules et de cellules circulantes. Longtemps considéré comme une monocouche passive, l'endothélium joue d'importants rôles dans la régulation de la pression sanguine, de l'hémostase, des réponses immunitaires et inflammatoires. L'adhérence cellule/cellule est initiée dans l'endothélium vasculaire par des interactions homophiliques entre molécules de VE-cadhérine (= jonctions adhérentes). La dynamique de la jonction et du cytosquelette est importante pour le remodelage des jonctions intercellulaires qui a lieu au cours l'angiogenèse, de la vasculogenèse et lors de la réparation de l'endothélium. C'est pourquoi la détermination des mécanismes moléculaires sous-jacents est indispensable à la comprehension de phénomènes physiopathologiques (angiogenèse et progression tumorales, inflammation...). D'après la littérature, la protéine EPLIN intervient dans la formation du complexe E-cadhérine/alpha-caténine/EPLIN et stabilise l'actine corticale. Actuellement décrite comme spécifique des modèles épithéliaux, EPLIN peut-elle intervenir dans la liaison du complexe à base de VE-cadhérine au cytosquelette d'actine? De plus, il paraît essentiel de comprendre le rôle de cette protéine dans les cellules car son expression est fortement diminuée dans la plupart des cancers alors qu'à l'inverse sa surexpression bloque la prolifération cellulaire.The endothelium forms the main barrier regulating the passage of macromolecules and circulating cells between the blood and tissue. Historically viewed as a passive vascular lining, vascular endothelium plays important roles in the regulation of vascular pressure, hemostasis, immune and inflammatory responses. In vascular endothelium, cell/cell adhesion is mediated by homophilic interactions of VE-cadherin molecules (= adherens junctions). Cytoskeleton and junction dynamics are important for intercellular junctions remodelling that occurs during angiogenesis, vasculogenesis and endothelium repair. So, determining the underlying molecular mechanisms is essential for the comprehension of pathologic phenomena such as angiogenesis, tumor progression or inflammation. We learn from the literature that EPLIN is involved in E-cadherin/a-catenin/EPLIN complex formation and cortical actin stabilization. Usually described as a protein specific of epithelial models, we wondered if EPLIN is able to link VE-cadherin complex to actin cytoskeleton. Furthermore, it seems essential to understand its cellular role since it is downregulated in many cancers while in contrast its overexpression blocks cell proliferation.SAVOIE-SCD - Bib.électronique (730659901) / SudocGRENOBLE1/INP-Bib.électronique (384210012) / SudocGRENOBLE2/3-Bib.électronique (384219901) / SudocSudocFranceF

    Nouvelle architecture de la jonction adhérente endothéliale

    No full text
    Les cellules tumorales, comme les leucocytes, franchissent l'endothélium vasculaire au niveau des jonctions intercellulaires à l'endroit même où s'exprime la VE-cadhérine, la protéine majeure des jonctions adhérentes. Cette transmigration déstabilise transitoire ment les jonctions, ce qui affecte l'intégrité de ce tissu. Afin de comprendre les mécanismes sous-jacents à l'ouverture des jonctions, nous avons établi la composition protéique du complexe à base de VE-cadhérine des jonctions adhérentes matures de cellules endothéliales primaires confIuentes de type HUVEC. Pour cela, nous avons couplé immunoprécipitation et analyse protéomique par spectrométrie de masse (LC-nanoESI-MS/MS). Nous avons ainsi identifié de nouveaux partenaires du complexe à base de VE-cadhérine jamais identifiés auparavant au niveau de la jonction adhérente endothéliale. Parmi ceux-ci se trouvent des protéines liant l'actine telles l'annexine 2 et la moésine.Nos résultats indiquent que l'annexine 2, qui s'accumule à la membrane plasmique au niveau des radeaux de cholestérol, entre en interaction directe avec le complexe à base de VE-cadhérine. Ainsi, l'annexine 2 connecte le complexe jonctionnel à base de VE-cadhérine au cytosquelette d'actine dans les HUVEC confIuentes. L'utilisation de siRNA nous a permis d'établir que l'expression de l'annexine 2 est absolument nécessaire au maintien de la VEcadhérine à la membrane plasmique. Nos résultats suggèrent que l'annexine 2, connectée à l'actine, arrime le complexe à base de VE-cadhérine au niveau des radeaux de cholestérol. En limitant la diffusion membranaire du complexe jonctionnel, ces interactions aboutissent à une consolidation des jonctions adhérentes ce qui contribue à maintenir l'intégrité de l'endothélium vasculaire. Lorsque les HUVEC sont soumises à des molecules destabilisant les jonctions adhérentes, une délocalisation de l'annexine 2 de la membrane vers le cytosol et une perturbation de la localisation de la VE-cadhérine sont observés. Ceci suggère que le prérequis à l'ouverture des jonctions adhérentes nécessite une rupture de l'interaction existant entre le complexe à base de VE-cadhérine et l'annexine 2.La moésine, quant à elle, semble interagir avec le complexe à base de VE-cadhérine dans les jonctions immatures formées entre cellules sub-confIuentes. La moésine serait donc impliquée dans l'établissement des contacts intercellulaires précoces plutôt que dans la maturation des jonctions endothéliales.Tumoral cells as leucocytes get through the vascular endothelium across adherens junctions, mainly composed of VE-cadherin. This transmigration process disturbs transiently the junctions, impairing so the endothelium integrity.To understand the mechanisms leading to the opening of adherens junctions, we first determined the proteic composition of the VE-cadherin-based complex of primary endothelial cell (HUVEC) mature adherens junctions. T 0 do so, we developed a method coupling immunoprecipitation and mass spectrometry-based proteomic analysis (LCnanoESI -MS/MS). Partners of VE-cadherin, so far unknown such as annexin 2 and moesin, were so identified. Our results demonstrated that, in confIuent HUVECs, the VE-cadherin-based complex interacts with annexin 2 and that annexin 2 translocates from the cytoplasm to the plasma membrane as cell reach confIuence. Annexin 2, located in cholesterol rafts, binds both to the actin cytoskeleton and the VE-cadherin-based complex so the complex is docked to cholesterol rafts. These multiple connections prevent the lateral diffusion of the VE-cadherin-based complex thus strengthening adherens junctions in the ultimate steps of maturation. Moreover, we observed that the down-regulation of annexin 2 by siRNA induces a delocalization of VE-cadherin from adherens junctions and consequently a destabilization of the se junctions. Furthermore, our data suggest that the decoupling of the annexin 2/p IlGRENOBLE1-BU Sciences (384212103) / SudocSudocFranceF

    Architecture of the VE-cadherin hexamer.

    Get PDF
    International audienceVascular endothelial-cadherin (VE-cadherin) is the major constituent of the adherens junctions of endothelial cells and plays a key role in angiogenesis and vascular permeability. The ectodomains EC1-4 of VE-cadherin are known to form hexamers in solution. To examine the mechanism of homotypic association of VE-cadherin, we have made a 3D reconstruction of the EC1-4 hexamer using electron microscopy and produced a homology model based on the known structure of C-cadherin EC1-5. The hexamer consists of a trimer of dimers with each N-terminal EC1 module making an antiparallel dimeric contact, and the EC4 modules forming extensive trimeric interactions. Each EC1-4 molecule makes a helical curve allowing some torsional flexibility to the edifice. While there is no direct evidence for the existence of hexamers of cadherin at adherens junctions, the model that we have produced provides indirect evidence since it can be used to explain some of the disparate results for adherens junctions. It is in accord with the X-ray and electron microscopy results, which demonstrate that the EC1 dimer is central to homotypic cadherin interaction. It provides an explanation for the force measurements of the interaction between opposing cadherin layers, which have previously been interpreted as resulting from three different interdigitating interactions. It is in accord with observations of native junctions by cryo-electron microscopy. The fact that this hexameric model of VE-cadherin can be used to explain more of the existing data on adherens junctions than any other model alone argues in favour of the existence of the hexamer at the adherens junction. In the context of the cell-cell junction these cis-trimers close to the membrane, and trans-dimers from opposing membranes, would increase the avidity of the bond

    Vascular endothelial-cadherin tyrosine phosphorylation in angiogenic and quiescent adult tissues.: VE-cadherin tyrosine phosphorylation

    Get PDF
    International audienceVascular endothelial-cadherin (VE-cadherin) plays a key role in angiogenesis and in vascular permeability. The regulation of its biological activity may be a central mechanism in normal or pathological angiogenesis. VE-cadherin has been shown to be phosphorylated on tyrosine in vitro under various conditions, including stimulation by VEGF. In the present study, we addressed the question of the existence of a tyrosine phosphorylated form of VE-cadherin in vivo, in correlation with the quiescent versus angiogenic state of adult tissues. Phosphorylated VE-cadherin was detected in mouse lung, uterus, and ovary but not in other tissues unless mice were injected with peroxovanadate to block protein phosphatases. Remarkably, VE-cadherin tyrosine phosphorylation was dramatically increased in uterus and ovary, and not in other organs, during PMSG/hCG-induced angiogenesis. In parallel, we observed an increased association of VE-cadherin with Flk1 (VEGF receptor 2) during hormonal angiogenesis. Additionally, Src kinase was constitutively associated with VE-cadherin in both quiescent and angiogenic tissues and increased phosphorylation of VE-cadherin-associated Src was detected in uterus and ovary after hormonal treatment. Src-VE-cadherin association was detected in cultured endothelial cells, independent of VE-cadherin phosphorylation state and Src activation level. In this model, Src inhibition impaired VEGF-induced VE-cadherin phosphorylation, indicating that VE-cadherin phosphorylation was dependent on Src activation. We conclude that VE-cadherin is a substrate for tyrosine kinases in vivo and that its phosphorylation, together with that of associated Src, is increased by angiogenic stimulation. Physical association between Flk1, Src, and VE-cadherin may thus provide an efficient mechanism for amplification and perpetuation of VEGF-stimulated angiogenic processes
    corecore